1
|
Verlinden TJM, Lamers WH, Herrler A, Köhler SE. The differences in the anatomy of the thoracolumbar and sacral autonomic outflow are quantitative. Clin Auton Res 2024; 34:79-97. [PMID: 38403748 PMCID: PMC10944453 DOI: 10.1007/s10286-024-01023-6] [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: 07/17/2023] [Accepted: 10/12/2023] [Indexed: 02/27/2024]
Abstract
PURPOSE We have re-evaluated the anatomical arguments that underlie the division of the spinal visceral outflow into sympathetic and parasympathetic divisions. METHODOLOGY Using a systematic literature search, we mapped the location of catecholaminergic neurons throughout the mammalian peripheral nervous system. Subsequently, a narrative method was employed to characterize segment-dependent differences in the location of preganglionic cell bodies and the composition of white and gray rami communicantes. RESULTS AND CONCLUSION One hundred seventy studies were included in the systematic review, providing information on 389 anatomical structures. Catecholaminergic nerve fibers are present in most spinal and all cranial nerves and ganglia, including those that are known for their parasympathetic function. Along the entire spinal autonomic outflow pathways, proximal and distal catecholaminergic cell bodies are common in the head, thoracic, and abdominal and pelvic region, which invalidates the "short-versus-long preganglionic neuron" argument. Contrary to the classically confined outflow levels T1-L2 and S2-S4, preganglionic neurons have been found in the resulting lumbar gap. Preganglionic cell bodies that are located in the intermediolateral zone of the thoracolumbar spinal cord gradually nest more ventrally within the ventral motor nuclei at the lumbar and sacral levels, and their fibers bypass the white ramus communicans and sympathetic trunk to emerge directly from the spinal roots. Bypassing the sympathetic trunk, therefore, is not exclusive for the sacral outflow. We conclude that the autonomic outflow displays a conserved architecture along the entire spinal axis, and that the perceived differences in the anatomy of the autonomic thoracolumbar and sacral outflow are quantitative.
Collapse
Affiliation(s)
- Thomas J M Verlinden
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands.
| | - Wouter H Lamers
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andreas Herrler
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - S Eleonore Köhler
- Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
2
|
Zhao Q, Jiang C, Zhao L, Dai X, Yi S. Unleashing Axonal Regeneration Capacities: Neuronal and Non-neuronal Changes After Injuries to Dorsal Root Ganglion Neuron Central and Peripheral Axonal Branches. Mol Neurobiol 2024; 61:423-433. [PMID: 37620687 DOI: 10.1007/s12035-023-03590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Peripheral nerves obtain remarkable regenerative capacity while central nerves can hardly regenerate following nerve injury. Sensory neurons in the dorsal root ganglion (DRG) are widely used to decipher the dissimilarity between central and peripheral axonal regeneration as axons of DRG neurons bifurcate into the regeneration-incompetent central projections and the regeneration-competent peripheral projections. A conditioning peripheral branch injury facilitates central axonal regeneration and enables the growth and elongation of central axons. Peripheral axonal injury stimulates neuronal calcium influx, alters the start-point chromatin states, increases chromatin accessibility, upregulates the expressions of regeneration-promoting genes and the synthesis of proteins, and supports axonal regeneration. Following central axonal injury, the responses of DRG neurons are modest, resulting in poor intrinsic growth ability. Some non-neuronal cells in DRGs, for instance satellite glial cells, also exhibit diminished injury responses to central axon injury as compared with peripheral axon injury. Moreover, DRG central and peripheral axonal branches are respectively surrounded by inhibitory glial scars generated by central glial cells and a permissive microenvironment generated by Schwann cells and macrophages. The aim of this review is to look at changes of DRG neurons and non-neuronal cells after peripheral and central axon injuries and summarize the contributing roles of both neuronal intrinsic regenerative capacities and surrounding microenvironments in axonal regeneration.
Collapse
Affiliation(s)
- Qian Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Chunyi Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Li Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Xiu Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
3
|
Zhao S, Tang C, Weinberger J, Gao D, Hou S. Sprouting of afferent and efferent inputs to pelvic organs after spinal cord injury. J Neuropathol Exp Neurol 2023; 83:20-29. [PMID: 38102789 PMCID: PMC10746698 DOI: 10.1093/jnen/nlad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Neural plasticity occurs within the central and peripheral nervous systems after spinal cord injury (SCI). Although central alterations have extensively been studied, it is largely unknown whether afferent and efferent fibers in pelvic viscera undergo similar morphological changes. Using a rat spinal cord transection model, we conducted immunohistochemistry to investigate afferent and efferent innervations to the kidney, colon, and bladder. Approximately 3-4 weeks after injury, immunostaining demonstrated that tyrosine hydroxylase (TH)-labeled postganglionic sympathetic fibers and calcitonin gene-related peptide (CGRP)-immunoreactive sensory terminals sprout in the renal pelvis and colon. Morphologically, sprouted afferent or efferent projections showed a disorganized structure. In the bladder, however, denser CGRP-positive primary sensory fibers emerged in rats with SCI, whereas TH-positive sympathetic efferent fibers did not change. Numerous CGRP-positive afferents were observed in the muscle layer and the lamina propria of the bladder following SCI. TH-positive efferent inputs displayed hypertrophy with large diameters, but their innervation patterns were sustained. Collectively, afferent or efferent inputs sprout widely in the pelvic organs after SCI, which may be one of the morphological bases underlying functional adaptation or maladaptation.
Collapse
Affiliation(s)
- Shunyi Zhao
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Chuanxi Tang
- Department of Neurobiology and Cell Biology, Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jeremy Weinberger
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
| | - Dianshuai Gao
- Department of Neurobiology and Cell Biology, Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Shaoping Hou
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, USA
| |
Collapse
|
4
|
Pandey M, Zhang JH, Adikaram PR, Kittock C, Lue N, Awe A, Degner K, Jacob N, Staples J, Thomas R, Kohnen AB, Ganesan S, Kabat J, Chen CK, Simonds WF. Specific regulation of mechanical nociception by Gβ5 involves GABA-B receptors. JCI Insight 2023; 8:e134685. [PMID: 37219953 PMCID: PMC10371342 DOI: 10.1172/jci.insight.134685] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
Mechanical, thermal, and chemical pain sensation is conveyed by primary nociceptors, a subset of sensory afferent neurons. The intracellular regulation of the primary nociceptive signal is an area of active study. We report here the discovery of a Gβ5-dependent regulatory pathway within mechanical nociceptors that restrains antinociceptive input from metabotropic GABA-B receptors. In mice with conditional knockout (cKO) of the gene that encodes Gβ5 (Gnb5) targeted to peripheral sensory neurons, we demonstrate the impairment of mechanical, thermal, and chemical nociception. We further report the specific loss of mechanical nociception in Rgs7-Cre+/- Gnb5fl/fl mice but not in Rgs9-Cre+/- Gnb5fl/fl mice, suggesting that Gβ5 might specifically regulate mechanical pain in regulator of G protein signaling 7-positive (Rgs7+) cells. Additionally, Gβ5-dependent and Rgs7-associated mechanical nociception is dependent upon GABA-B receptor signaling since both were abolished by treatment with a GABA-B receptor antagonist and since cKO of Gβ5 from sensory cells or from Rgs7+ cells potentiated the analgesic effects of GABA-B agonists. Following activation by the G protein-coupled receptor Mrgprd agonist β-alanine, enhanced sensitivity to inhibition by baclofen was observed in primary cultures of Rgs7+ sensory neurons harvested from Rgs7-Cre+/- Gnb5fl/fl mice. Taken together, these results suggest that the targeted inhibition of Gβ5 function in Rgs7+ sensory neurons might provide specific relief for mechanical allodynia, including that contributing to chronic neuropathic pain, without reliance on exogenous opioids.
Collapse
Affiliation(s)
- Mritunjay Pandey
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jian-Hua Zhang
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Poorni R. Adikaram
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Claire Kittock
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Nicole Lue
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Adam Awe
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Katherine Degner
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Nirmal Jacob
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Jenna Staples
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Rachel Thomas
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Allison B. Kohnen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Sundar Ganesan
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Juraj Kabat
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Ching-Kang Chen
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - William F. Simonds
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
5
|
Rockel AF, Wagner N, Spenger P, Ergün S, Wörsdörfer P. Neuro-mesodermal assembloids recapitulate aspects of peripheral nervous system development in vitro. Stem Cell Reports 2023; 18:1155-1165. [PMID: 37084722 DOI: 10.1016/j.stemcr.2023.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/23/2023] Open
Abstract
Here we describe a novel neuro-mesodermal assembloid model that recapitulates aspects of peripheral nervous system (PNS) development such as neural crest cell (NCC) induction, migration, and sensory as well as sympathetic ganglion formation. The ganglia send projections to the mesodermal as well as neural compartment. Axons in the mesodermal part are associated with Schwann cells. In addition, peripheral ganglia and nerve fibers interact with a co-developing vascular plexus, forming a neurovascular niche. Finally, developing sensory ganglia show response to capsaicin indicating their functionality. The presented assembloid model could help to uncover mechanisms of human NCC induction, delamination, migration, and PNS development. Moreover, the model could be used for toxicity screenings or drug testing. The co-development of mesodermal and neuroectodermal tissues and a vascular plexus along with a PNS allows us to investigate the crosstalk between neuroectoderm and mesoderm and between peripheral neurons/neuroblasts and endothelial cells.
Collapse
Affiliation(s)
- Anna F Rockel
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Nicole Wagner
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Peter Spenger
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany
| | - Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070 Würzburg, Germany.
| |
Collapse
|
6
|
Atherton MA, Park S, Horan NL, Nicholson S, Dolan JC, Schmidt BL, Scheff NN. Sympathetic modulation of tumor necrosis factor alpha-induced nociception in the presence of oral squamous cell carcinoma. Pain 2023; 164:27-42. [PMID: 35714327 PMCID: PMC9582047 DOI: 10.1097/j.pain.0000000000002655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/08/2022] [Indexed: 01/09/2023]
Abstract
ABSTRACT Head and neck squamous cell carcinoma (HNSCC) causes more severe pain and psychological stress than other types of cancer. Despite clinical evidence linking pain, stress, and cancer progression, the underlying relationship between pain and sympathetic neurotransmission in oral cancer is unknown. We found that human HNSCC tumors and mouse tumor tissue are innervated by peripheral sympathetic and sensory nerves. Moreover, β-adrenergic 1 and 2 receptors (β-ARs) are overexpressed in human oral cancer cell lines, and norepinephrine treatment increased β-AR2 protein expression as well as cancer cell proliferation in vitro. We have recently demonstrated that inhibition of tumor necrosis factor alpha (TNFα) signaling reduces oral cancer-induced nociceptive behavior. Norepinephrine-treated cancer cell lines secrete more TNFα which, when applied to tongue-innervating trigeminal neurons, evoked a larger Ca 2+ transient; TNF-TNFR inhibitor blocked the increase in the evoked Ca 2+ transient. Using an orthotopic xenograft oral cancer model, we found that mice demonstrated significantly less orofacial cancer-induced nociceptive behavior during systemic β-adrenergic inhibitory treatment with propranolol. Furthermore, chemical sympathectomy using guanethidine led to a significant reduction in tumor size and nociceptive behavior. We infer from these results that sympathetic signaling modulates oral cancer pain through TNFα secretion and tumorigenesis. Further investigation of the role of neurocancer communication in cancer progression and pain is warranted.
Collapse
Affiliation(s)
- Megan A Atherton
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Stella Park
- Bluestone Center for Clinical Research, DDS Program, College of Dentistry, New York University, New York, NY, United States
| | - Nicole L Horan
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Samuel Nicholson
- Bluestone Center for Clinical Research, DDS Program, College of Dentistry, New York University, New York, NY, United States
| | - John C Dolan
- Bluestone Center for Clinical Research, DDS Program, College of Dentistry, New York University, New York, NY, United States
| | - Brian L Schmidt
- Bluestone Center for Clinical Research, DDS Program, College of Dentistry, New York University, New York, NY, United States
| | - Nicole N Scheff
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Huzard D, Martin M, Maingret F, Chemin J, Jeanneteau F, Mery PF, Fossat P, Bourinet E, François A. The impact of C-tactile low-threshold mechanoreceptors on affective touch and social interactions in mice. SCIENCE ADVANCES 2022; 8:eabo7566. [PMID: 35767616 PMCID: PMC9242590 DOI: 10.1126/sciadv.abo7566] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Affective touch is necessary for proper neurodevelopment and sociability. However, it remains unclear how the neurons innervating the skin detect affective and social behaviors. The C low-threshold mechanoreceptors (C-LTMRs), a specific population of somatosensory neurons in mice, appear particularly well suited, physiologically and anatomically, to perceive affective and social touch. However, their contribution to sociability has not been resolved yet. Our observations revealed that C-LTMR functional deficiency induced social isolation and reduced tactile interactions in adulthood. Conversely, transient increase in C-LTMR excitability in adults, using chemogenetics, was rewarding, promoted touch-seeking behaviors, and had prosocial influences on group dynamics. This work provides the first empirical evidence that specific peripheral inputs alone can drive complex social behaviors. It demonstrates the existence of a specialized neuronal circuit, originating in the skin, wired to promote interactions with other individuals.
Collapse
Affiliation(s)
- Damien Huzard
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Miquel Martin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - François Maingret
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Jean Chemin
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pierre-François Mery
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Pascal Fossat
- Institut des Maladies Neurodégénératives, Université de Bordeaux, CNRS, Bordeaux, France
| | - Emmanuel Bourinet
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Amaury François
- Institut de Génomique Fonctionnelle (IGF), Université de Montpellier, CNRS, INSERM, Montpellier, France
- Corresponding author.
| |
Collapse
|
8
|
Finno CJ, Chen Y, Park S, Lee JH, Perez-Flores MC, Choi J, Yamoah EN. Cisplatin Neurotoxicity Targets Specific Subpopulations and K + Channels in Tyrosine-Hydroxylase Positive Dorsal Root Ganglia Neurons. Front Cell Neurosci 2022; 16:853035. [PMID: 35586548 PMCID: PMC9108181 DOI: 10.3389/fncel.2022.853035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Among the features of cisplatin chemotherapy-induced peripheral neuropathy are chronic pain and innocuous mechanical hypersensitivity. The complete etiology of the latter remains unknown. Here, we show that cisplatin targets a heterogeneous population of tyrosine hydroxylase-positive (TH+) primary afferent dorsal root ganglion neurons (DRGNs) in mice, determined using single-cell transcriptome and electrophysiological analyses. TH+ DRGNs regulate innocuous mechanical sensation through C-low threshold mechanoreceptors. A differential assessment of wild-type and vitamin E deficient TH+ DRGNs revealed heterogeneity and specific functional phenotypes. The TH+ DRGNs comprise; fast-adapting eliciting one action potential (AP; 1-AP), moderately-adapting (≥2-APs), in responses to square-pulse current injection, and spontaneously active (SA). Cisplatin increased the input resistance and AP frequency but reduced the temporal coding feature of 1-AP and ≥2-APs neurons. By contrast, cisplatin has no measurable effect on the SA neurons. Vitamin E reduced the cisplatin-mediated increased excitability but did not improve the TH+ neuron temporal coding properties. Cisplatin mediates its effect by targeting outward K+ current, likely carried through K2P18.1 (Kcnk18), discovered through the differential transcriptome studies and heterologous expression. Studies show a potential new cellular target for chemotherapy-induced peripheral neuropathy and implicate the possible neuroprotective effects of vitamin E in cisplatin chemotherapy.
Collapse
Affiliation(s)
- Carrie J. Finno
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Yingying Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Seojin Park
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Jeong Han Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | | | - Jinsil Choi
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Reno, Reno, NV, United States
| |
Collapse
|
9
|
Zheng Q, Xie W, Lückemeyer DD, Lay M, Wang XW, Dong X, Limjunyawong N, Ye Y, Zhou FQ, Strong JA, Zhang JM, Dong X. Synchronized cluster firing, a distinct form of sensory neuron activation, drives spontaneous pain. Neuron 2022; 110:209-220.e6. [PMID: 34752775 PMCID: PMC8776619 DOI: 10.1016/j.neuron.2021.10.019] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/01/2021] [Accepted: 10/13/2021] [Indexed: 01/21/2023]
Abstract
Spontaneous pain refers to pain occurring without external stimuli. It is a primary complaint in chronic pain conditions and remains difficult to treat. Moreover, the mechanisms underlying spontaneous pain remain poorly understood. Here we employed in vivo imaging of dorsal root ganglion (DRG) neurons and discovered a distinct form of abnormal spontaneous activity following peripheral nerve injury: clusters of adjacent DRG neurons firing synchronously and sporadically. The level of cluster firing correlated directly with nerve injury-induced spontaneous pain behaviors. Furthermore, we demonstrated that cluster firing is triggered by activity of sympathetic nerves, which sprout into DRGs after injury, and identified norepinephrine as a key neurotransmitter mediating this unique firing. Chemogenetic and pharmacological manipulations of sympathetic activity and norepinephrine receptors suggest that they are necessary and sufficient for DRG cluster firing and spontaneous pain behavior. Therefore, blocking sympathetically mediated cluster firing may be a new paradigm for treating spontaneous pain.
Collapse
Affiliation(s)
- Qin Zheng
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Debora D Lückemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mark Lay
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Yaqing Ye
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA.
| |
Collapse
|
10
|
Morelli C, Castaldi L, Brown SJ, Streich LL, Websdale A, Taberner FJ, Cerreti B, Barenghi A, Blum KM, Sawitzke J, Frank T, Steffens LK, Doleschall B, Serrao J, Ferrarini D, Lechner SG, Prevedel R, Heppenstall PA. Identification of a population of peripheral sensory neurons that regulates blood pressure. Cell Rep 2021; 35:109191. [PMID: 34077727 PMCID: PMC8187988 DOI: 10.1016/j.celrep.2021.109191] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/14/2020] [Accepted: 05/09/2021] [Indexed: 02/08/2023] Open
Abstract
The vasculature is innervated by a network of peripheral afferents that sense and regulate blood flow. Here, we describe a system of non-peptidergic sensory neurons with cell bodies in the spinal ganglia that regulate vascular tone in the distal arteries. We identify a population of mechanosensitive neurons, marked by tropomyosin receptor kinase C (TrkC) and tyrosine hydroxylase in the dorsal root ganglia, which projects to blood vessels. Local stimulation of TrkC neurons decreases vessel diameter and blood flow, whereas systemic activation increases systolic blood pressure and heart rate variability via the sympathetic nervous system. Ablation of the neurons provokes variability in local blood flow, leading to a reduction in systolic blood pressure, increased heart rate variability, and ultimately lethality within 48 h. Thus, a population of TrkC+ sensory neurons forms part of a sensory-feedback mechanism that maintains cardiovascular homeostasis through the autonomic nervous system. TrkC+/Th+ DRG neurons project to blood vessels Local stimulation of TrkC+ DRG neurons decreases vessel diameter and blood flow Systemic activation of TrkC+ DRG neurons increases blood pressure and heart rate Ablation of TrkC+ neurons dysregulates cardiovascular homeostasis and is lethal
Collapse
Affiliation(s)
- Chiara Morelli
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany; Collaboration for joint PhD degree between EMBL Heidelberg, Heidelberg, Germany, and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Laura Castaldi
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Sam J Brown
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | - Lina L Streich
- Collaboration for joint PhD degree between EMBL Heidelberg, Heidelberg, Germany, and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Francisco J Taberner
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | | | - Kevin M Blum
- Center for Regenerative Medicine, the Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Biomedical Engineering, the Ohio State University, Columbus, OH, USA
| | | | - Tessa Frank
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | - Laura K Steffens
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Joana Serrao
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy
| | | | - Stefan G Lechner
- Institute of Pharmacology, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Robert Prevedel
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany; Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Paul A Heppenstall
- EMBL Rome, Via Ramarini 32, Monterotondo 00015, Italy; Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany.
| |
Collapse
|
11
|
Schiller M, Azulay-Debby H, Boshnak N, Elyahu Y, Korin B, Ben-Shaanan TL, Koren T, Krot M, Hakim F, Rolls A. Optogenetic activation of local colonic sympathetic innervations attenuates colitis by limiting immune cell extravasation. Immunity 2021; 54:1022-1036.e8. [PMID: 33932356 PMCID: PMC8116309 DOI: 10.1016/j.immuni.2021.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 01/16/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
The sympathetic nervous system is composed of an endocrine arm, regulating blood adrenaline and noradrenaline, and a local arm, a network of fibers innervating immune organs. Here, we investigated the impact of the local arm of the SNS in an inflammatory response in the colon. Intra-rectal insertion of an optogenetic probe in mice engineered to express channelrhodopsin-2 in tyrosine hydroxylase cells activated colonic sympathetic fibers. In contrast to systemic application of noradrenaline, local activation of sympathetic fibers attenuated experimental colitis and reduced immune cell abundance. Gene expression profiling showed decreased endothelial expression of the adhesion molecule MAdCAM-1 upon optogenetic stimulation; this decrease was sensitive to adrenergic blockers and 6-hydroxydopamine. Antibody blockade of MAdCAM-1 abrogated the optogenetic effect on immune cell extravasation into the colon and the pathology. Thus, sympathetic fibers control colonic inflammation by regulating immune cell extravasation from circulation, a mechanism likely relevant in multiple organs.
Collapse
Affiliation(s)
- Maya Schiller
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Hilla Azulay-Debby
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Nadia Boshnak
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Yehezqel Elyahu
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 8410501, Beer-Sheva, Israel
| | - Ben Korin
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Tamar L Ben-Shaanan
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Tamar Koren
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Maria Krot
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel
| | - Fahed Hakim
- Cancer Research Center, EMMS Nazareth, 16100, Nazareth, Israel; Azrieli faculty of medicine, Bar-Ilan university, 1311502, Safad, Israel
| | - Asya Rolls
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; Department of Neuroscience, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 3525422, Haifa, Israel; The Technion Integrated Cancer Center, Technion-Israel Institute of Technology, 3525422, Haifa, Israel.
| |
Collapse
|
12
|
Valek L, Tran B, Wilken-Schmitz A, Trautmann S, Heidler J, Schmid T, Brüne B, Thomas D, Deller T, Geisslinger G, Auburger G, Tegeder I. Prodromal sensory neuropathy in Pink1 -/- SNCA A53T double mutant Parkinson mice. Neuropathol Appl Neurobiol 2021; 47:1060-1079. [PMID: 33974284 DOI: 10.1111/nan.12734] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/28/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022]
Abstract
AIMS Parkinson's disease (PD) is frequently associated with a prodromal sensory neuropathy manifesting with sensory loss and chronic pain. We have recently shown that PD-associated sensory neuropathy in patients is associated with high levels of glucosylceramides. Here, we assessed the underlying pathology and mechanisms in Pink1-/- SNCAA53T double mutant mice. METHODS We studied nociceptive and olfactory behaviour and the neuropathology of dorsal root ganglia (DRGs), including ultrastructure, mitochondrial respiration, transcriptomes, outgrowth and calcium currents of primary neurons, and tissue ceramides and sphingolipids before the onset of a PD-like disease that spontaneously develops in Pink1-/- SNCAA53T double mutant mice beyond 15 months of age. RESULTS Similar to PD patients, Pink1-/- SNCAA53T mice developed a progressive prodromal sensory neuropathy with a loss of thermal sensitivity starting as early as 4 months of age. In analogy to human plasma, lipid analyses revealed an accumulation of glucosylceramides (GlcCer) in the DRGs and sciatic nerves, which was associated with pathological mitochondria, impairment of mitochondrial respiration, and deregulation of transient receptor potential channels (TRPV and TRPA) at mRNA, protein and functional levels in DRGs. Direct exposure of DRG neurons to GlcCer caused transient hyperexcitability, followed by a premature decline of the viability of sensory neurons cultures upon repeated GlcCer application. CONCLUSIONS The results suggest that pathological GlcCer contribute to prodromal sensory disease in PD mice via mitochondrial damage and calcium channel hyperexcitability. GlcCer-associated sensory neuron pathology might be amenable to GlcCer lowering therapeutic strategies.
Collapse
Affiliation(s)
- Lucie Valek
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Bao Tran
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Annett Wilken-Schmitz
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Sandra Trautmann
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Juliana Heidler
- Functional Proteomics Group, Faculty of Medicine, Goethe-University, Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| | - Dominique Thomas
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany.,Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), Frankfurt, Germany
| | - Georg Auburger
- Experimental Neurology, Faculty of Medicine, Goethe-University, Frankfurt, Germany
| | - Irmgard Tegeder
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University of Frankfurt, Frankfurt, Germany
| |
Collapse
|
13
|
Brumberg J, Kuzkina A, Lapa C, Mammadova S, Buck A, Volkmann J, Sommer C, Isaias IU, Doppler K. Dermal and cardiac autonomic fiber involvement in Parkinson's disease and multiple system atrophy. Neurobiol Dis 2021; 153:105332. [PMID: 33722614 DOI: 10.1016/j.nbd.2021.105332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 12/01/2022] Open
Abstract
Pathological aggregates of alpha-synuclein in peripheral dermal nerve fibers can be detected in patients with idiopathic Parkinson's disease and multiple system atrophy. This study combines skin biopsy staining for p-alpha-synuclein depositions and radionuclide imaging of the heart with [123I]-metaiodobenzylguanidine to explore peripheral denervation in both diseases. To this purpose, 42 patients with a clinical diagnosis of Parkinson's disease or multiple system atrophy were enrolled. All patients underwent a standardized clinical work-up including neurological evaluation, neurography, and blood samples. Skin biopsies were obtained from the distal and proximal leg, back, and neck for immunofluorescence double labeling with anti-p-alpha-synuclein and anti-PGP9.5. All patients underwent myocardial [123I]-metaiodobenzylguanidine scintigraphy. Dermal p-alpha-synuclein was observed in 47.6% of Parkinson's disease patients and was mainly found in autonomic structures. 81.0% of multiple system atrophy patients had deposits with most of cases in somatosensory fibers. The [123I]-metaiodobenzylguanidine heart-to-mediastinum ratio was lower in Parkinson's disease than in multiple system atrophy patients (1.94 ± 0.63 vs. 2.91 ± 0.96; p < 0.0001). Irrespective of the diagnosis, uptake was lower in patients with than without p-alpha-synuclein in autonomic structures (1.42 ± 0.51 vs. 2.74 ± 0.83; p < 0.0001). Rare cases of Parkinson's disease with p-alpha-synuclein in somatosensory fibers and multiple system atrophy patients with deposits in autonomic structures or both fiber types presented with clinically overlapping features. In conclusion, this study suggests that alpha-synuclein contributes to peripheral neurodegeneration and mediates the impairment of cardiac sympathetic neurons in patients with synucleinopathies. Furthermore, it indicates that Parkinson's disease and multiple system atrophy share pathophysiologic mechanisms of peripheral nervous system dysfunction with a clinical overlap.
Collapse
Affiliation(s)
- Joachim Brumberg
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Oberdϋrrbacher Straβe 6, 97080 Würzburg, Germany.
| | - Anastasia Kuzkina
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Oberdϋrrbacher Straβe 6, 97080 Würzburg, Germany; Nuclear Medicine, Medical Faculty, University of Augsburg, Stenglinstraβe 2, 86156 Augsburg, Germany
| | - Sona Mammadova
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Oberdϋrrbacher Straβe 6, 97080 Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| | - Ioannis U Isaias
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg and Julius-Maximilian-University Würzburg, Josef-Schneider-Straβe 11, 97080 Würzburg, Germany
| |
Collapse
|
14
|
Sapio MR, Vazquez FA, Loydpierson AJ, Maric D, Kim JJ, LaPaglia DM, Puhl HL, Lu VB, Ikeda SR, Mannes AJ, Iadarola MJ. Comparative Analysis of Dorsal Root, Nodose and Sympathetic Ganglia for the Development of New Analgesics. Front Neurosci 2021; 14:615362. [PMID: 33424545 PMCID: PMC7793666 DOI: 10.3389/fnins.2020.615362] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.
Collapse
Affiliation(s)
- Matthew R Sapio
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Fernando A Vazquez
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Amelia J Loydpierson
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jenny J Kim
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Danielle M LaPaglia
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Henry L Puhl
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Van B Lu
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Stephen R Ikeda
- Section on Neurotransmitter Signaling, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Andrew J Mannes
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| | - Michael J Iadarola
- Anesthesia Section, Department of Perioperative Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States
| |
Collapse
|
15
|
Adelman PC, Baumbauer KM, Friedman R, Shah M, Wright M, Young E, Jankowski MP, Albers KM, Koerber HR. Single-cell q-PCR derived expression profiles of identified sensory neurons. Mol Pain 2020; 15:1744806919884496. [PMID: 31588843 PMCID: PMC6820183 DOI: 10.1177/1744806919884496] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sensory neurons are chemically and functionally heterogeneous, and this heterogeneity has been examined extensively over the last several decades. These studies have employed a variety of different methodologies, including anatomical, electrophysiological, and molecular approaches. Recent studies using next-generation sequencing techniques have examined the transcriptome of single sensory neurons. Although these reports have provided a wealth of exciting new information on the heterogeneity of sensory neurons, correlation with functional types is lacking. Here, we employed retrograde tracing of cutaneous and muscle afferents to examine the variety of mRNA expression profiles of individual, target-specific sensory neurons. In addition, we used an ex vivo skin/nerve/dorsal root ganglion/spinal cord preparation to record and characterize the functional response properties of individual cutaneous sensory neurons that were then intracellularly labeled with fluorescent dyes, recovered from dissociated cultures, and analyzed for gene expression. We found that by using single-cell quantitative polymerase chain reaction techniques and a set of 28 genes, we can identify transcriptionally distinct groups. We have also used calcium imaging and single-cell quantitative polymerase chain reaction to determine the correlation between levels of mRNA expression and functional protein expression and how functional properties correlated with the different transcriptional groups. These studies show that although transcriptomics does map to functional types, within any one functional subgroup, there are highly variable patterns of gene expression. Thus, studies that rely on the expression pattern of one or a few genes as a stand in for physiological experiments, runs a high risk of data misinterpretation with respect to function.
Collapse
Affiliation(s)
- Peter C Adelman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Kyle M Baumbauer
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Robert Friedman
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Mansi Shah
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Margaret Wright
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Erin Young
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Michael P Jankowski
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - H Richard Koerber
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| |
Collapse
|
16
|
Habib AM, Nagi K, Thillaiappan NB, Sukumaran V, Akhtar S. Vitamin D and Its Potential Interplay With Pain Signaling Pathways. Front Immunol 2020; 11:820. [PMID: 32547536 PMCID: PMC7270292 DOI: 10.3389/fimmu.2020.00820] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
About 50 million of the U.S. adult population suffer from chronic pain. It is a complex disease in its own right for which currently available analgesics have been deemed woefully inadequate since ~20% of the sufferers derive no benefit. Vitamin D, known for its role in calcium homeostasis and bone metabolism, is thought to be of clinical benefit in treating chronic pain without the side-effects of currently available analgesics. A strong correlation between hypovitaminosis D and incidence of bone pain is known. However, the potential underlying mechanisms by which vitamin D might exert its analgesic effects are poorly understood. In this review, we discuss pathways involved in pain sensing and processing primarily at the level of dorsal root ganglion (DRG) neurons and the potential interplay between vitamin D, its receptor (VDR) and known specific pain signaling pathways including nerve growth factor (NGF), glial-derived neurotrophic factor (GDNF), epidermal growth factor receptor (EGFR), and opioid receptors. We also discuss how vitamin D/VDR might influence immune cells and pain sensitization as well as review the increasingly important topic of vitamin D toxicity. Further in vitro and in vivo experimental studies will be required to study these potential interactions specifically in pain models. Such studies could highlight the potential usefulness of vitamin D either alone or in combination with existing analgesics to better treat chronic pain.
Collapse
Affiliation(s)
| | | | | | | | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
17
|
Finno CJ, Peterson J, Kang M, Park S, Bordbari MH, Durbin-Johnson B, Settles M, Perez-Flores MC, Lee JH, Yamoah EN. Single-Cell RNA-seq Reveals Profound Alterations in Mechanosensitive Dorsal Root Ganglion Neurons with Vitamin E Deficiency. iScience 2019; 21:720-735. [PMID: 31733517 PMCID: PMC6864320 DOI: 10.1016/j.isci.2019.10.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/16/2019] [Accepted: 10/28/2019] [Indexed: 12/26/2022] Open
Abstract
Ninety percent of Americans consume less than the estimated average requirements of dietary vitamin E (vitE). Severe vitE deficiency due to genetic mutations in the tocopherol transfer protein (TTPA) in humans results in ataxia with vitE deficiency (AVED), with proprioceptive deficits and somatosensory degeneration arising from dorsal root ganglia neurons (DRGNs). Single-cell RNA-sequencing of DRGNs was performed in Ttpa-/- mice, an established model of AVED. In stark contrast to expected changes in proprioceptive neurons, Ttpa-/- DRGNs showed marked upregulation of voltage-gated Ca2+ and K+ channels in mechanosensitive, tyrosine-hydroxylase positive (TH+) DRGNs. The ensuing significant conductance changes resulted in reduced excitability in mechanosensitive Ttpa-/- DRGNs. A highly supplemented vitE diet (600 mg dl-α-tocopheryl acetate/kg diet) prevented the cellular and molecular alterations and improved mechanosensation. VitE deficiency profoundly alters the molecular signature and functional properties of mechanosensitive TH+ DRGN, representing an intriguing shift of the prevailing paradigm from proprioception to mechanical sensation.
Collapse
Affiliation(s)
- Carrie J Finno
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA.
| | - Janel Peterson
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Mincheol Kang
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV 89557, USA
| | - Seojin Park
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV 89557, USA
| | - Matthew H Bordbari
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Blythe Durbin-Johnson
- Bioinformatics Core Facility, Genome Center, University of California, Davis, CA 95616, USA
| | - Matthew Settles
- Bioinformatics Core Facility, Genome Center, University of California, Davis, CA 95616, USA
| | - Maria C Perez-Flores
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV 89557, USA
| | - Jeong H Lee
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV 89557, USA
| | - Ebenezer N Yamoah
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV 89557, USA
| |
Collapse
|
18
|
Synaptic Organization of VGLUT3 Expressing Low-Threshold Mechanosensitive C Fiber Terminals in the Rodent Spinal Cord. eNeuro 2019; 6:eN-NWR-0007-19. [PMID: 30783617 PMCID: PMC6378328 DOI: 10.1523/eneuro.0007-19.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/11/2019] [Accepted: 01/16/2019] [Indexed: 11/21/2022] Open
Abstract
Low-threshold mechanosensitive C fibers (C-LTMRs) that express the vesicular glutamate transporter VGLUT3 are thought to signal affective touch, and may also play a role in mechanical allodynia. However, the nature of the central termination of C-LTMRs in the dorsal horn remains largely unexplored. Here, we used light and electron microscopy in combination with VGLUT3 immunolabeling as a marker of C-LTMR terminations to investigate this issue. VGLUT3+ C-LTMRs formed central terminals of Type II glomeruli in the inner part of lamina II of the dorsal horn, often establishing multiple asymmetric synapses with postsynaptic dendrites but also participating in synaptic configurations with presynaptic axons and dendrites. Unexpectedly, essentially all VGLUT3+ C-LTMR terminals showed substantial VGLUT1 expression in the rat, whereas such terminals in mice lacked VGLUT1. Most VGLUT3+ C-LTMR terminals exhibited weak-to-moderate VGLUT2 expression. Further, C-LTMR terminals formed numerous synapses with excitatory protein kinase Cγ (PKCγ) interneurons and inhibitory parvalbumin neurons, whereas synapses with calretinin neurons were scarce. C-LTMR terminals rarely if ever established synapses with neurokinin 1 receptor (NK1R)-possessing dendrites traversing lamina II. Thus, VGLUT3+ C-LTMR terminals appear to largely correspond to neurofilament-lacking central terminals of Type II glomeruli in inner lamina II and can thus be identified at the ultrastructural level by morphological criteria. The participation of C-LTMR terminals in Type II glomeruli involving diverse populations of interneuron indicates highly complex modes of integration of C-LTMR mediated signaling in the dorsal horn. Furthermore, differences in VGLUT1 expression indicate distinct species differences in synaptic physiology of C-LTMR terminals.
Collapse
|
19
|
Abstract
Nociceptive signals conveyed to the dorsal horn of the spinal cord by primary nociceptors are subject to extensive modulation by local neurons and by supraspinal descending pathways to the spinal cord before being relayed to higher brain centers. Descending modulatory pathways to the spinal cord comprise, among others, noradrenergic, serotonergic, γ-aminobutyric acid (GABA)ergic, and dopaminergic fibers. The contributions of noradrenaline, serotonin, and GABA to pain modulation have been extensively investigated. In contrast, the contributions of dopamine to pain modulation remain poorly understood. The focus of this review is to summarize the current knowledge of the contributions of dopamine to pain modulation. Hypothalamic A11 dopaminergic neurons project to all levels of the spinal cord and provide the main source of spinal dopamine. Dopamine receptors are expressed in primary nociceptors as well as in spinal neurons located in different laminae in the dorsal horn of the spinal cord, suggesting that dopamine can modulate pain signals by acting at both presynaptic and postsynaptic targets. Here, I will review the literature on the effects of dopamine and dopamine receptor agonists/antagonists on the excitability of primary nociceptors, the effects of dopamine on the synaptic transmission between primary nociceptors and dorsal horn neurons, and the effects of dopamine on pain in rodents. Published data support both anti-nociceptive effects of dopamine mediated by D2-like receptors and pro-nociceptive effects mediated by D1-like receptors.
Collapse
Affiliation(s)
- Michelino Puopolo
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| |
Collapse
|
20
|
Guimarães MZP, De Vecchi R, Vitória G, Sochacki JK, Paulsen BS, Lima I, Rodrigues da Silva F, da Costa RFM, Castro NG, Breton L, Rehen SK. Generation of iPSC-Derived Human Peripheral Sensory Neurons Releasing Substance P Elicited by TRPV1 Agonists. Front Mol Neurosci 2018; 11:277. [PMID: 30186108 PMCID: PMC6113370 DOI: 10.3389/fnmol.2018.00277] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/23/2018] [Indexed: 01/01/2023] Open
Abstract
Neural crest stem cells (NCPCs) have been shown to differentiate into various cell types and tissues during embryonic development, including sensory neurons. The few studies addressing the generation of NCPCs and peripheral sensory neurons (PSNs) from human induced pluripotent stem cells (hiPSCs), generated sensory cells without displaying robust activity. Here, we describe an efficient strategy for hiPSCs differentiation into NCPCs and functional PSNs using chemically defined media and factors to achieve efficient differentiation, confirmed by the expression of specific markers. After 10 days hiPSCs differentiated into NCPCs, cells were then maintained in neural induction medium containing defined growth factors for PSNs differentiation, followed by 10 days in neonatal human epidermal keratinocytes- (HEKn-) conditioned medium (CM). We observed a further increase in PSN markers expression and neurites length after CM treatment. The resulting neurons elicited action potentials after current injection and released substance P (SP) in response to nociceptive agents such as anandamide and resiniferatoxin. Anandamide induced substance P release via activation of TRPV1 and not CB1. Transcriptomic analysis of the PSNs revealed the main dorsal root ganglia neuronal markers and a transcriptional profile compatible with C fiber-low threshold mechanoreceptors. TRPV1 was detected by immunofluorescence and RNA-Seq in multiple experiments. In conclusion, the developed strategy generated PSNs useful for drug screening that could be applied to patient-derived hiPSCs, consisting in a powerful tool to model human diseases in vitro.
Collapse
Affiliation(s)
- Marília Z P Guimarães
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo De Vecchi
- L'Oréal Research & Innovation, Rio de Janeiro, Brazil.,Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Gabriela Vitória
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | | | - Bruna S Paulsen
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Igor Lima
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Felipe Rodrigues da Silva
- Department of Genetics and Evolution, Institute of Biology, University of Campinas, Campinas, Brazil.,Embrapa Informática Agropecuária, Campinas, Brazil
| | | | - Newton G Castro
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lionel Breton
- L'Oréal Research & Innovation, Aulnay-sous-Bois, France
| | - Stevens K Rehen
- D'Or Institute for Research and Education, Rio de Janeiro, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Nascimento AI, Mar FM, Sousa MM. The intriguing nature of dorsal root ganglion neurons: Linking structure with polarity and function. Prog Neurobiol 2018; 168:86-103. [PMID: 29729299 DOI: 10.1016/j.pneurobio.2018.05.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 11/26/2022]
Abstract
Dorsal root ganglion (DRG) neurons are the first neurons of the sensory pathway. They are activated by a variety of sensory stimuli that are then transmitted to the central nervous system. An important feature of DRG neurons is their unique morphology where a single process -the stem axon- bifurcates into a peripheral and a central axonal branch, with different functions and cellular properties. Distinctive structural aspects of the two DRG neuron branches may have important implications for their function in health and disease. However, the link between DRG axonal branch structure, polarity and function has been largely neglected in the field, and relevant information is rather scattered across the literature. In particular, ultrastructural differences between the two axonal branches are likely to account for the higher transport and regenerative ability of the peripheral DRG neuron axon when compared to the central one. Nevertheless, the cell intrinsic factors contributing to this central-peripheral asymmetry are still unknown. Here we critically review the factors that may underlie the functional asymmetry between the peripheral and central DRG axonal branches. Also, we discuss the hypothesis that DRG neurons may assemble a structure resembling the axon initial segment that may be responsible, at least in part, for their polarity and electrophysiological features. Ultimately, we suggest that the clarification of the axonal ultrastructure of DRG neurons using state-of-the-art techniques will be crucial to understand the physiology of this peculiar cell type.
Collapse
Affiliation(s)
- Ana Isabel Nascimento
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar-ICBAS, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Fernando Milhazes Mar
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular-IBMC and Instituto de Inovação e Investigação em Saúde, University of Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.
| |
Collapse
|
22
|
Nociceptive DRG neurons express muscle lim protein upon axonal injury. Sci Rep 2017; 7:643. [PMID: 28377582 PMCID: PMC5428558 DOI: 10.1038/s41598-017-00590-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/03/2017] [Indexed: 01/21/2023] Open
Abstract
Muscle lim protein (MLP) has long been regarded as a cytosolic and nuclear muscular protein. Here, we show that MLP is also expressed in a subpopulation of adult rat dorsal root ganglia (DRG) neurons in response to axonal injury, while the protein was not detectable in naïve cells. Detailed immunohistochemical analysis of L4/L5 DRG revealed ~3% of MLP-positive neurons 2 days after complete sciatic nerve crush and maximum ~10% after 4–14 days. Similarly, in mixed cultures from cervical, thoracic, lumbar and sacral DRG ~6% of neurons were MLP-positive after 2 days and maximal 17% after 3 days. In both, histological sections and cell cultures, the protein was detected in the cytosol and axons of small diameter cells, while the nucleus remained devoid. Moreover, the vast majority could not be assigned to any of the well characterized canonical DRG subpopulations at 7 days after nerve injury. However, further analysis in cell culture revealed that the largest population of MLP expressing cells originated from non-peptidergic IB4-positive nociceptive neurons, which lose their ability to bind the lectin upon axotomy. Thus, MLP is mostly expressed in a subset of axotomized nociceptive neurons and can be used as a novel marker for this population of cells.
Collapse
|
23
|
Oroszova Z, Hricova L, Stropkovska A, Lukacova N, Pavel J. The Characterization of AT 1 Expression in the Dorsal Root Ganglia After Chronic Constriction Injury. Cell Mol Neurobiol 2017; 37:545-554. [PMID: 27319015 DOI: 10.1007/s10571-016-0396-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 06/13/2016] [Indexed: 10/21/2022]
Abstract
To clarify the role of Angiotensin II in the regulation of sensory signaling, we characterized the AT1 expression in neuronal subpopulation of lower lumbar dorsal root ganglia under normal conditions and its alteration in neuropathic pain model. The characterization of AT1 expression was done under control and after the chronic constriction injury induced by four loose ligatures of the sciatic nerve representing the model of posttraumatic painful peripheral neuropathy. Major Angiotensin II receptor type was expressed in approximately 43 % of small-sized and 62 % of large-sized neurons in control. The AT1 overexpression after sciatic nerve ligation lasting 7 days was detected predominantly in small-sized AT1 immunoreactive neurons (about 38 % increase). Chronic constriction injury caused a statistically marked increase in number of the small-sized peptidergic (CGRP immunoreactive) neuronal subpopulation expressing AT1 (about 64 %). The subpopulations of AT1-immunoreactive and nonpeptide-containing primary sensory neurons revealed by IB4 binding, tyrosine hydroxylase- and parvalbumin-immunoreactive neurons were not markedly changed. Our results indicate that: (1) the AT1 overexpression after the chronic constriction injury is an important factor in Angiotensin II-potentiated pain perception; (2) Angiotensin II is involved in pathological mechanisms of neuropathic pain and this effect can be mediated perhaps in combination with other neuropeptides synthesized in the primary sensory neurons.
Collapse
Affiliation(s)
- Zuzana Oroszova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Slovak Academy of Sciences, Soltesovej 4-6, 040 01, Kosice, Slovak Republic
| | - Ludmila Hricova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Slovak Academy of Sciences, Soltesovej 4-6, 040 01, Kosice, Slovak Republic
| | - Andrea Stropkovska
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Slovak Academy of Sciences, Soltesovej 4-6, 040 01, Kosice, Slovak Republic
| | - Nadezda Lukacova
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Slovak Academy of Sciences, Soltesovej 4-6, 040 01, Kosice, Slovak Republic
| | - Jaroslav Pavel
- Department of Neurodegeneration, Plasticity and Repair, Institute of Neurobiology, Slovak Academy of Sciences, Soltesovej 4-6, 040 01, Kosice, Slovak Republic.
| |
Collapse
|
24
|
Ekhteiari Salmas R, Durdagi S, Gulhan MF, Duruyurek M, Abdullah HI, Selamoglu Z. The effects of pollen, propolis, and caffeic acid phenethyl ester on tyrosine hydroxylase activity and total RNA levels in hypertensive rats caused by nitric oxide synthase inhibition: experimental, docking and molecular dynamic studies. J Biomol Struct Dyn 2017; 36:609-620. [PMID: 28132600 DOI: 10.1080/07391102.2017.1288660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ramin Ekhteiari Salmas
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Mehmet Fuat Gulhan
- Department of Medicinal and Aromatic Plants, Vocational School of Technical Sciences, Aksaray, Turkey
| | - Merve Duruyurek
- Faculty of Arts and Science, Department of Biotechnology, Omer Halisdemir University, Nigde 51240, Turkey
| | - Huda I. Abdullah
- Department of Pharmacology, New York Medical College, Valhalla 10595, NY, USA
| | - Zeliha Selamoglu
- Faculty of Arts and Science, Department of Biotechnology, Omer Halisdemir University, Nigde 51240, Turkey
| |
Collapse
|
25
|
Dorsal root ganglion neurons and tyrosine hydroxylase--an intriguing association with implications for sensation and pain. Pain 2016; 157:314-320. [PMID: 26447702 DOI: 10.1097/j.pain.0000000000000381] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
26
|
Seal RP. Do the distinct synaptic properties of VGLUTs shape pain? Neurochem Int 2016; 98:82-8. [PMID: 27180049 DOI: 10.1016/j.neuint.2016.04.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 04/26/2016] [Accepted: 04/28/2016] [Indexed: 11/25/2022]
Abstract
The somatosensory system transmits touch, temperature, itch and pain. Three vesicular glutamate transporter isoforms mediate the release of glutamate throughout the mammalian nervous system with largely non-overlapping distributions and unique roles at the synapse. This review discusses the contribution of each of these essential transporters to circuits underlying pain and other somatosensory behaviors throughout postnatal development and in the adult. A better understanding of the individual contributions of the VGLUT isoforms could provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
27
|
Huang Y, Zhang Q, Song NN, Zhang L, Sun YL, Hu L, Chen JY, Zhu W, Li J, Ding YQ. Lrp5/6 are required for cerebellar development and for suppressing TH expression in Purkinje cells via β-catenin. Mol Brain 2016; 9:7. [PMID: 26772978 PMCID: PMC4714458 DOI: 10.1186/s13041-015-0183-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/25/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The cerebellum is responsible for coordinating motor functions and has a unique laminated architecture. Purkinje cells are inhibitory neurons and represent the only output from the cerebellar cortex. Tyrosine hydroxylase (TH) is the key enzyme for the synthesis of catecholamines, including dopamine and noradrenaline, and it is normally not expressed in cerebellar neurons. RESULTS We report here that the low-density lipoprotein receptors (Lrp) 5 and 6, Wnt co-receptors, are required for the development of the cerebellum and for suppressing ectopic TH expression in Purkinje cells. Simultaneous inactivation of Lrp 5 and 6 by Nestin-Cre results in defective lamination and foliation of the cerebellum during postnatal development. Surprisingly, TH is ectopically expressed by Purkinje cells, although they still keep its other neurochemical characteristics. These phenotypes are also observed in the cerebellum of GFAP-Cre;β-catenin(flox/flox) mice, and AAV2-Cre-mediated gene deletion leads to ectopic TH expression in Purkinje cells of β-catenin(flox/flox) mice as well. CONCLUSIONS Our results revealed a new role of the canonical Lrp5/6-β-catenin pathway in regulating the morphogenesis of the cerebellum during postnatal development.
Collapse
Affiliation(s)
- Ying Huang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qiong Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ning-Ning Song
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yu-Ling Sun
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Ling Hu
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jia-Ying Chen
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China
| | - Weidong Zhu
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.,Clinical and Translational Research Center, East Hospital, Shanghai, 200120, China
| | - Jue Li
- Department of Prevention, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Department of Anatomy and Neurobiology, Collaborative Innovation Centerfor Brain Science, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
28
|
Chakraborty S, Rebecchi M, Kaczocha M, Puopolo M. Dopamine modulation of transient receptor potential vanilloid type 1 (TRPV1) receptor in dorsal root ganglia neurons. J Physiol 2016; 594:1627-42. [PMID: 26563747 DOI: 10.1113/jp271198] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/04/2015] [Indexed: 01/11/2023] Open
Abstract
The transient receptor potential vanilloid type 1 (TRPV1) receptor plays a key role in the modulation of nociceptor excitability. To address whether dopamine can modulate the activity of TRPV1 channels in nociceptive neurons, the effects of dopamine and dopamine receptor agonists were tested on the capsaicin-activated current recorded from acutely dissociated small diameter (<27 μm) dorsal root ganglia (DRG) neurons. Dopamine or SKF 81297 (an agonist at D1/D5 receptors), caused inhibition of both inward and outward currents by ∼60% and ∼48%, respectively. The effect of SKF 81297 was reversed by SCH 23390 (an antagonist at D1/D5 receptors), confirming that it was mediated by activation of D1/D5 dopamine receptors. In contrast, quinpirole (an agonist at D2 receptors) had no significant effect on the capsaicin-activated current. Inhibition of the capsaicin-activated current by SKF 81297 was mediated by G protein coupled receptors (GPCRs), and highly dependent on external calcium. The inhibitory effect of SKF 81297 on the capsaicin-activated current was not affected when the protein kinase A (PKA) activity was blocked with H89, or when the protein kinase C (PKC) activity was blocked with bisindolylmaleimide II (BIM). In contrast, when the calcium-calmodulin-dependent protein kinase II (CaMKII) was blocked with KN-93, the inhibitory effect of SKF 81297 on the capsaicin-activated current was greatly reduced, suggesting that activation of D1/D5 dopamine receptors may be preferentially linked to CaMKII activity. We suggest that modulation of TRPV1 channels by dopamine in nociceptive neurons may represent a way for dopamine to modulate incoming noxious stimuli.
Collapse
Affiliation(s)
- Saikat Chakraborty
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Mario Rebecchi
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| |
Collapse
|
29
|
Malet M, Brumovsky PR. VGLUTs and Glutamate Synthesis-Focus on DRG Neurons and Pain. Biomolecules 2015; 5:3416-37. [PMID: 26633536 PMCID: PMC4693284 DOI: 10.3390/biom5043416] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 12/14/2022] Open
Abstract
The amino acid glutamate is the principal excitatory transmitter in the nervous system, including in sensory neurons that convey pain sensation from the periphery to the brain. It is now well established that a family of membrane proteins, termed vesicular glutamate transporters (VGLUTs), serve a critical function in these neurons: they incorporate glutamate into synaptic vesicles. VGLUTs have a central role both under normal neurotransmission and pathological conditions, such as neuropathic or inflammatory pain. In the present short review, we will address VGLUTs in the context of primary afferent neurons. We will focus on the role of VGLUTs in pain triggered by noxious stimuli, peripheral nerve injury, and tissue inflammation, as mostly explored in transgenic mice. The possible interplay between glutamate biosynthesis and VGLUT-dependent packaging in synaptic vesicles, and its potential impact in various pain states will be presented.
Collapse
Affiliation(s)
- Mariana Malet
- Institute of Research on Translational Medicine, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Austral University, Avenida Juan D. Perón 1500, Pilar, Buenos Aires 1629AHJ, Argentina.
| | - Pablo R Brumovsky
- Institute of Research on Translational Medicine, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Austral University, Avenida Juan D. Perón 1500, Pilar, Buenos Aires 1629AHJ, Argentina.
| |
Collapse
|
30
|
Nerve growth factor–induced synapse-like structures in contralateral sensory ganglia contribute to chronic mirror-image pain. Pain 2015; 156:2295-2309. [DOI: 10.1097/j.pain.0000000000000280] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Lee DW, Cho PS, Lee HK, Lee SH, Jung SJ, Oh SB. Trans-activation of TRPV1 by D1R in mouse dorsal root ganglion neurons. Biochem Biophys Res Commun 2015; 465:832-7. [DOI: 10.1016/j.bbrc.2015.08.096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/21/2015] [Indexed: 10/23/2022]
|
32
|
Nascimento FP, Magnussen C, Yousefpour N, Ribeiro-da-Silva A. Sympathetic fibre sprouting in the skin contributes to pain-related behaviour in spared nerve injury and cuff models of neuropathic pain. Mol Pain 2015; 11:59. [PMID: 26376854 PMCID: PMC4574171 DOI: 10.1186/s12990-015-0062-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/08/2015] [Indexed: 02/06/2023] Open
Abstract
Background Cuff and spared nerve injury (SNI) in the sciatic territory are widely used to model neuropathic pain. Because nociceptive information is first detected in skin, it is important to understand how alterations in peripheral innervation contribute to pain in each model. Over 16 weeks in male rats, changes in sensory and autonomic innervation of the skin were described after cuff and SNI using immunohistochemistry to label myelinated (neurofilament 200 positive—NF200+) and peptidergic (calcitonin gene-related peptide positive—CGRP+) primary afferents and sympathetic fibres (dopamine β-hydroxylase positive—DBH+) Results Cuff and SNI caused an early loss and later reinnervation of NF200 and CGRP fibres in the plantar hind paw skin. In both models, DBH+ fibres sprouted into the upper dermis of the plantar skin 4 and 6 weeks after injury. Despite these similarities, behavioural pain measures were significantly different in each model. Sympathectomy using guanethidine significantly alleviated mechanical allodynia 6 weeks after cuff, when peak sympathetic sprouting was observed, having no effect at 2 weeks, when fibres were absent. In SNI animals, mechanical allodynia in the lateral paw was significantly improved by guanethidine at 2 and 6 weeks, and the development of cold hyperalgesia, which roughly paralleled the appearance of ectopic sympathetic fibres, was alleviated by guanethidine at 6 weeks. Sympathetic fibres did not sprout into the dorsal root ganglia at 2 or 6 weeks, indicating their unimportance to pain behaviour in these two models. Conclusions Alterations in sympathetic innervation in the skin represents an important mechanism that contributes to pain in cuff and SNI models of neuropathic pain.
Collapse
Affiliation(s)
- Francisney P Nascimento
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, QC, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, H3A 0G1, Canada.
| | - Claire Magnussen
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, QC, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, H3A 0G1, Canada.
| | - Noosha Yousefpour
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, QC, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, H3A 0G1, Canada.
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, McGill University, McIntyre Medical Building, 3655 Promenade Sir William Osler, Room 1215, Montreal, QC, H3G 1Y6, Canada. .,Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, H3A 0G1, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 0C7, Canada.
| |
Collapse
|
33
|
Spinal dopaminergic projections control the transition to pathological pain plasticity via a D1/D5-mediated mechanism. J Neurosci 2015; 35:6307-17. [PMID: 25904784 DOI: 10.1523/jneurosci.3481-14.2015] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The mechanisms that lead to the maintenance of chronic pain states are poorly understood, but their elucidation could lead to new insights into how pain becomes chronic and how it can potentially be reversed. We investigated the role of spinal dorsal horn neurons and descending circuitry in plasticity mediating a transition to pathological pain plasticity suggesting the presence of a chronic pain state using hyperalgesic priming. We found that when dorsal horn neurokinin 1 receptor-positive neurons or descending serotonergic neurons were ablated before hyperalgesic priming, IL-6- and carrageenan-induced mechanical hypersensitivity was impaired, and subsequent prostaglandin E2 (PGE2) response was blunted. However, when these neurons were lesioned after the induction of priming, they had no effect on the PGE2 response, reflecting differential mechanisms driving plasticity in a primed state. In stark contrast, animals with a spinally applied dopaminergic lesion showed intact IL-6- and carrageenan-induced mechanical hypersensitivity, but the subsequent PGE2 injection failed to cause mechanical hypersensitivity. Moreover, ablating spinally projecting dopaminergic neurons after the resolution of the IL-6- or carrageenan-induced response also reversed the maintenance of priming as assessed through mechanical hypersensitivity and the mouse grimace scale. Pharmacological antagonism of spinal dopamine D1/D5 receptors reversed priming, whereas D1/D5 agonists induced mechanical hypersensitivity exclusively in primed mice. Strikingly, engagement of D1/D5 coupled with anisomycin in primed animals reversed a chronic pain state, consistent with reconsolidation-like effects in the spinal dorsal horn. These findings demonstrate a novel role for descending dopaminergic neurons in the maintenance of pathological pain plasticity.
Collapse
|
34
|
McCarthy CJ, Tomasella E, Malet M, Seroogy KB, Hökfelt T, Villar MJ, Gebhart GF, Brumovsky PR. Axotomy of tributaries of the pelvic and pudendal nerves induces changes in the neurochemistry of mouse dorsal root ganglion neurons and the spinal cord. Brain Struct Funct 2015; 221:1985-2004. [PMID: 25749859 DOI: 10.1007/s00429-015-1019-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/24/2015] [Indexed: 12/31/2022]
Abstract
Using immunohistochemical techniques, we characterized changes in the expression of several neurochemical markers in lumbar 4-sacral 2 (L4-S2) dorsal root ganglion (DRG) neuron profiles (NPs) and the spinal cord of BALB/c mice after axotomy of the L6 and S1 spinal nerves, major tributaries of the pelvic (targeting pelvic visceral organs) and pudendal (targeting perineum and genitalia) nerves. Sham animals were included. Expression of cyclic AMP-dependent transcription factor 3 (ATF3), calcitonin gene-related peptide (CGRP), transient receptor potential cation channel subfamily V, member 1 (TRPV1), tyrosine hydroxylase (TH) and vesicular glutamate transporters (VGLUT) types 1 and -2 was analysed seven days after injury. L6-S1 axotomy induced dramatic de novo expression of ATF3 in many L6-S1 DRG NPs, and parallel significant downregulations in the percentage of CGRP-, TRPV1-, TH- and VGLUT2-immunoreactive (IR) DRG NPs, as compared to their expression in uninjured DRGs (contralateral L6-S1-AXO; sham mice); VGLUT1 expression remained unaltered. Sham L6-S1 DRGs only showed a small ipsilateral increase in ATF3-IR NPs (other markers were unchanged). L6-S1-AXO induced de novo expression of ATF3 in several lumbosacral spinal cord motoneurons and parasympathetic preganglionic neurons; in sham mice the effect was limited to a few motoneurons. Finally, a moderate decrease in CGRP- and TRPV1-like-immunoreactivities was observed in the ipsilateral superficial dorsal horn neuropil. In conclusion, injury of a mixed visceral/non-visceral nerve leads to considerable neurochemical alterations in DRGs matched, to some extent, in the spinal cord. Changes in these and potentially other nociception-related molecules could contribute to pain due to injury of nerves in the abdominopelvic cavity.
Collapse
Affiliation(s)
- Carly J McCarthy
- Faculty of Biomedical Sciences, School of Biomedical Sciences, Austral University, Av. Juan D. Perón 1500, Pilar, B1629AHJ, Buenos Aires, Argentina
| | - Eugenia Tomasella
- Faculty of Biomedical Sciences, School of Biomedical Sciences, Austral University, Av. Juan D. Perón 1500, Pilar, B1629AHJ, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariana Malet
- Faculty of Biomedical Sciences, School of Biomedical Sciences, Austral University, Av. Juan D. Perón 1500, Pilar, B1629AHJ, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Kim B Seroogy
- Department of Neurology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Marcelo J Villar
- Faculty of Biomedical Sciences, School of Biomedical Sciences, Austral University, Av. Juan D. Perón 1500, Pilar, B1629AHJ, Buenos Aires, Argentina
| | - G F Gebhart
- Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Pablo R Brumovsky
- Faculty of Biomedical Sciences, School of Biomedical Sciences, Austral University, Av. Juan D. Perón 1500, Pilar, B1629AHJ, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina. .,Department of Anesthesiology, Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
35
|
Yu L, Ding Y, Spencer A, Ma J, Lu R, Rudkin BB, Yuan C. Dorsal root ganglion progenitors differentiate to gamma-aminobutyric acid- and choline acetyltransferase-positive neurons. Neural Regen Res 2015; 7:485-91. [PMID: 25745432 PMCID: PMC4348992 DOI: 10.3969/j.issn.1673-5374.2012.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 02/11/2012] [Indexed: 11/18/2022] Open
Abstract
This study examined the isolation and differentiation of dorsal root ganglion progenitor cells for therapeutic use in neurodegenerative diseases. Rat embryonic dorsal root ganglia progenitors were isolated and purified using the differential adhesion method combined with cytosine arabinoside treatment. After culture in serum-free medium supplemented with B27, basic fibroblast growth factor and epidermal growth factor, these cells remained viable and survived for more than 18 months in vitro. Most cells differentiated to neurons that were immunoreactive for gamma-aminobutyric acid and choline acetyltransferase as detected by immunohistochemical staining. In addition, nerve growth factor and neurotrophic tyrosine kinase receptor expression were also observed in dorsal root ganglion progenitors and differentiated cells. K252a, an inhibitor that blocks nerve growth factor-induced signaling, inhibited cell survival, suggesting the possible existence of a nerve growth factor autocrine loop in these proliferating cells.
Collapse
Affiliation(s)
- Lingli Yu
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China ; Differentiation & Cell Cycle Group, Laboratory of Molecular and Cellular Science, UMR 5239 CNRS/ENS Lyon/University of Lyon 1, Ecole Normale Supérieure de Lyon, IFR128 "Biosciences Lyon-Gerland" Lyon, 69007, France
| | - Yindi Ding
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China ; Differentiation & Cell Cycle Group, Laboratory of Molecular and Cellular Science, UMR 5239 CNRS/ENS Lyon/University of Lyon 1, Ecole Normale Supérieure de Lyon, IFR128 "Biosciences Lyon-Gerland" Lyon, 69007, France
| | - Ambre Spencer
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China ; Differentiation & Cell Cycle Group, Laboratory of Molecular and Cellular Science, UMR 5239 CNRS/ENS Lyon/University of Lyon 1, Ecole Normale Supérieure de Lyon, IFR128 "Biosciences Lyon-Gerland" Lyon, 69007, France
| | - Ji Ma
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China
| | - Ruisheng Lu
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China
| | - Brian B Rudkin
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China ; Differentiation & Cell Cycle Group, Laboratory of Molecular and Cellular Science, UMR 5239 CNRS/ENS Lyon/University of Lyon 1, Ecole Normale Supérieure de Lyon, IFR128 "Biosciences Lyon-Gerland" Lyon, 69007, France
| | - Chonggang Yuan
- Laboratory of Molecular and Cellular Neurophysiology, School of Life Science, East China Normal University, Shanghai 200062, China
| |
Collapse
|
36
|
François A, Schüetter N, Laffray S, Sanguesa J, Pizzoccaro A, Dubel S, Mantilleri A, Nargeot J, Noël J, Wood JN, Moqrich A, Pongs O, Bourinet E. The Low-Threshold Calcium Channel Cav3.2 Determines Low-Threshold Mechanoreceptor Function. Cell Rep 2015; 10:370-382. [PMID: 25600872 DOI: 10.1016/j.celrep.2014.12.042] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 11/14/2014] [Accepted: 12/18/2014] [Indexed: 12/12/2022] Open
Abstract
The T-type calcium channel Cav3.2 emerges as a key regulator of sensory functions, but its expression pattern within primary afferent neurons and its contribution to modality-specific signaling remain obscure. Here, we elucidate this issue using a unique knockin/flox mouse strain wherein Cav3.2 is replaced by a functional Cav3.2-surface-ecliptic GFP fusion. We demonstrate that Cav3.2 is a selective marker of two major low-threshold mechanoreceptors (LTMRs), Aδ- and C-LTMRs, innervating the most abundant skin hair follicles. The presence of Cav3.2 along LTMR-fiber trajectories is consistent with critical roles at multiple sites, setting their strong excitability. Strikingly, the C-LTMR-specific knockout uncovers that Cav3.2 regulates light-touch perception and noxious mechanical cold and chemical sensations and is essential to build up that debilitates allodynic symptoms of neuropathic pain, a mechanism thought to be entirely A-LTMR specific. Collectively, our findings support a fundamental role for Cav3.2 in touch/pain pathophysiology, validating their critic pharmacological relevance to relieve mechanical and cold allodynia.
Collapse
Affiliation(s)
- Amaury François
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Niklas Schüetter
- Department of Physiology, University of Saarland, School of Medicine, Kirrberger Straße 1, 66424 Homburg, Germany
| | - Sophie Laffray
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Juan Sanguesa
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Anne Pizzoccaro
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Stefan Dubel
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Annabelle Mantilleri
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, 13288 Marseille, France
| | - Joel Nargeot
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France
| | - Jacques Noël
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; Université de Nice Sophia Antipolis, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS, 660 route des lucioles, 06560 Valbonne, France
| | - John N Wood
- Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, UMR 7288, 13288 Marseille, France
| | - Olaf Pongs
- Department of Physiology, University of Saarland, School of Medicine, Kirrberger Straße 1, 66424 Homburg, Germany
| | - Emmanuel Bourinet
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, 141 rue de la Cardonille, 34094 Montpellier, France; CNRS UMR5203, 34095 Montpellier, France; INSERM, U661, 34095 Montpellier, France; Université de Montpellier, 34095 Montpellier, France.
| |
Collapse
|
37
|
Goswami SC, Mishra SK, Maric D, Kaszas K, Gonnella GL, Clokie SJ, Kominsky HD, Gross JR, Keller JM, Mannes AJ, Hoon MA, Iadarola MJ. Molecular signatures of mouse TRPV1-lineage neurons revealed by RNA-Seq transcriptome analysis. THE JOURNAL OF PAIN 2014; 15:1338-1359. [PMID: 25281809 DOI: 10.1016/j.jpain.2014.09.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/10/2014] [Accepted: 09/19/2014] [Indexed: 12/22/2022]
Abstract
UNLABELLED Disorders of pain neural systems are frequently chronic and, when recalcitrant to treatment, can severely degrade the quality of life. The pain pathway begins with sensory neurons in dorsal root or trigeminal ganglia, and the neuronal subpopulations that express the transient receptor potential cation channel, subfamily V, member 1 (TRPV1) ion channel transduce sensations of painful heat and inflammation and play a fundamental role in clinical pain arising from cancer and arthritis. In the present study, we elucidate the complete transcriptomes of neurons from the TRPV1 lineage and a non-TRPV1 neuroglial population in sensory ganglia through the combined application of next-gen deep RNA-Seq, genetic neuronal labeling with fluorescence-activated cell sorting, or neuron-selective chemoablation. RNA-Seq accurately quantitates gene expression, a difficult parameter to determine with most other methods, especially for very low and very high expressed genes. Differentially expressed genes are present at every level of cellular function from the nucleus to the plasma membrane. We identified many ligand receptor pairs in the TRPV1 population, suggesting that autonomous presynaptic regulation may be a major regulatory mechanism in nociceptive neurons. The data define, in a quantitative, cell population-specific fashion, the molecular signature of a distinct and clinically important group of pain-sensing neurons and provide an overall framework for understanding the transcriptome of TRPV1 nociceptive neurons. PERSPECTIVE Next-gen RNA-Seq, combined with molecular genetics, provides a comprehensive and quantitative measurement of transcripts in TRPV1 lineage neurons and a contrasting transcriptome from non-TRPV1 neurons and cells. The transcriptome highlights previously unrecognized protein families, identifies multiple molecular circuits for excitatory or inhibitory autocrine and paracrine signaling, and suggests new combinatorial approaches to pain control.
Collapse
Affiliation(s)
- Samridhi C Goswami
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Santosh K Mishra
- Molecular Genetics Unit, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, Bethesda, Maryland
| | - Dragan Maric
- Laboratory of Neurophysiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Krisztian Kaszas
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Gian Luigi Gonnella
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Samuel J Clokie
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Hal D Kominsky
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jacklyn R Gross
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jason M Keller
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Andrew J Mannes
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Mark A Hoon
- Molecular Genetics Unit, Laboratory of Sensory Biology, National Institute of Dental and Craniofacial Research, Bethesda, Maryland
| | - Michael J Iadarola
- Anesthesia Section, Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
38
|
Minett MS, Falk S, Santana-Varela S, Bogdanov YD, Nassar MA, Heegaard AM, Wood JN. Pain without nociceptors? Nav1.7-independent pain mechanisms. Cell Rep 2014; 6:301-12. [PMID: 24440715 PMCID: PMC3969273 DOI: 10.1016/j.celrep.2013.12.033] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/22/2013] [Accepted: 12/20/2013] [Indexed: 11/20/2022] Open
Abstract
Nav1.7, a peripheral neuron voltage-gated sodium channel, is essential for pain and olfaction in mice and humans. We examined the role of Nav1.7 as well as Nav1.3, Nav1.8, and Nav1.9 in different mouse models of chronic pain. Constriction-injury-dependent neuropathic pain is abolished when Nav1.7 is deleted in sensory neurons, unlike nerve-transection-related pain, which requires the deletion of Nav1.7 in sensory and sympathetic neurons for pain relief. Sympathetic sprouting that develops in parallel with nerve-transection pain depends on the presence of Nav1.7 in sympathetic neurons. Mechanical and cold allodynia required distinct sets of neurons and different repertoires of sodium channels depending on the nerve injury model. Surprisingly, pain induced by the chemotherapeutic agent oxaliplatin and cancer-induced bone pain do not require the presence of Nav1.7 sodium channels or Nav1.8-positive nociceptors. Thus, similar pain phenotypes arise through distinct cellular and molecular mechanisms. Therefore, rational analgesic drug therapy requires patient stratification in terms of mechanisms and not just phenotype. Phenotypically identical pain models have different underlying molecular mechanisms Nav1.7 expression is required for sympathetic sprouting after neuronal damage Oxaliplatin and cancer-induced bone pain are both Nav1.7-independent Deleting Nav1.7 in adult mice reverses nerve damage-induced neuropathic pain
Collapse
Affiliation(s)
- Michael S Minett
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Sarah Falk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Yury D Bogdanov
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Mohammed A Nassar
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
39
|
Galbavy W, Safaie E, Rebecchi MJ, Puopolo M. Inhibition of tetrodotoxin-resistant sodium current in dorsal root ganglia neurons mediated by D1/D5 dopamine receptors. Mol Pain 2013; 9:60. [PMID: 24283218 PMCID: PMC4220807 DOI: 10.1186/1744-8069-9-60] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/22/2013] [Indexed: 12/25/2022] Open
Abstract
Background Dopaminergic fibers originating from area A11 of the hypothalamus project to different levels of the spinal cord and represent the major source of dopamine. In addition, tyrosine hydroxylase, the rate-limiting enzyme for the synthesis of catecholamines, is expressed in 8-10% of dorsal root ganglia (DRG) neurons, suggesting that dopamine may be released in the dorsal root ganglia. Dopamine has been shown to modulate calcium current in DRG neurons, but the effects of dopamine on sodium current and on the firing properties of small DRG neurons are poorly understood. Results The effects of dopamine and dopamine receptor agonists were tested on the tetrodotoxin-resistant (TTX-R) sodium current recorded from acutely dissociated small (diameter ≤ 25 μm) DRG neurons. Dopamine (20 μM) and SKF 81297 (10 μM) caused inhibition of TTX-R sodium current in small DRG neurons by 23% and 37%, respectively. In contrast, quinpirole (20 μM) had no effects on the TTX-R sodium current. Inhibition by SKF 81297 of the TTX-R sodium current was not affected when the protein kinase A (PKA) activity was blocked with the PKA inhibitory peptide (6–22), but was greatly reduced when the protein kinase C (PKC) activity was blocked with the PKC inhibitory peptide (19–36), suggesting that activation of D1/D5 dopamine receptors is linked to PKC activity. Expression of D1and D5 dopamine receptors in small DRG neurons, but not D2 dopamine receptors, was confirmed by Western blotting and immunofluorescence analysis. In current clamp experiments, the number of action potentials elicited in small DRG neurons by current injection was reduced by ~ 30% by SKF 81297. Conclusions We conclude that activation of D1/D5 dopamine receptors inhibits TTX-R sodium current in unmyelinated nociceptive neurons and dampens their intrinsic excitability by reducing the number of action potentials in response to stimulus. Increasing or decreasing levels of dopamine in the dorsal root ganglia may serve to adjust the sensitivity of nociceptors to noxious stimuli.
Collapse
Affiliation(s)
| | | | | | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY 11794, USA.
| |
Collapse
|
40
|
VGLUTs in Peripheral Neurons and the Spinal Cord: Time for a Review. ISRN NEUROLOGY 2013; 2013:829753. [PMID: 24349795 PMCID: PMC3856137 DOI: 10.1155/2013/829753] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/25/2013] [Indexed: 02/07/2023]
Abstract
Vesicular glutamate transporters (VGLUTs) are key molecules for the incorporation of glutamate in synaptic vesicles across the nervous system, and since their discovery in the early 1990s, research on these transporters has been intense and productive. This review will focus on several aspects of VGLUTs research on neurons in the periphery and the spinal cord. Firstly, it will begin with a historical account on the evolution of the morphological analysis of glutamatergic systems and the pivotal role played by the discovery of VGLUTs. Secondly, and in order to provide an appropriate framework, there will be a synthetic description of the neuroanatomy and neurochemistry of peripheral neurons and the spinal cord. This will be followed by a succinct description of the current knowledge on the expression of VGLUTs in peripheral sensory and autonomic neurons and neurons in the spinal cord. Finally, this review will address the modulation of VGLUTs expression after nerve and tissue insult, their physiological relevance in relation to sensation, pain, and neuroprotection, and their potential pharmacological usefulness.
Collapse
|
41
|
George L, Chaverra M, Wolfe L, Thorne J, Close-Davis M, Eibs A, Riojas V, Grindeland A, Orr M, Carlson GA, Lefcort F. Familial dysautonomia model reveals Ikbkap deletion causes apoptosis of Pax3+ progenitors and peripheral neurons. Proc Natl Acad Sci U S A 2013; 110:18698-703. [PMID: 24173031 PMCID: PMC3831979 DOI: 10.1073/pnas.1308596110] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Familial dysautonomia (FD) is a devastating developmental and progressive peripheral neuropathy caused by a mutation in the gene inhibitor of kappa B kinase complex-associated protein (IKBKAP). To identify the cellular and molecular mechanisms that cause FD, we generated mice in which Ikbkap expression is ablated in the peripheral nervous system and identify the steps in peripheral nervous system development that are Ikbkap-dependent. We show that Ikbkap is not required for trunk neural crest migration or pathfinding, nor for the formation of dorsal root or sympathetic ganglia, or the adrenal medulla. Instead, Ikbkap is essential for the second wave of neurogenesis during which the majority of tropomyosin-related kinase A (TrkA(+)) nociceptors and thermoreceptors arise. In its absence, approximately half the normal complement of TrkA(+) neurons are lost, which we show is partly due to p53-mediated premature differentiation and death of mitotically-active progenitors that express the paired-box gene Pax3 and give rise to the majority of TrkA(+) neurons. By the end of sensory development, the number of TrkC neurons is significantly increased, which may result from an increase in Runx3(+) cells. Furthermore, our data demonstrate that TrkA(+) (but not TrkC(+)) sensory and sympathetic neurons undergo exacerbated Caspase 3-mediated programmed cell death in the absence of Ikbkap and that this death is not due to a reduction in nerve growth factor synthesis. In summary, these data suggest that FD does not result from a failure in trunk neural crest migration, but rather from a critical function for Ikbkap in TrkA progenitors and TrkA(+) neurons.
Collapse
Affiliation(s)
- Lynn George
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
- Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT 59101; and
| | - Marta Chaverra
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Lindsey Wolfe
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Julian Thorne
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Mattheson Close-Davis
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Amy Eibs
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | - Vickie Riojas
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| | | | - Miranda Orr
- McLaughlin Research Institute, Great Falls, MT 59405
| | | | - Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717
| |
Collapse
|
42
|
Petrie CN, Smithson LJ, Crotty AM, Michalski B, Fahnestock M, Kawaja MD. Overexpression of nerve growth factor by murine smooth muscle cells: Role of the p75 neurotrophin receptor on sympathetic and sensory sprouting. J Comp Neurol 2013; 521:2621-43. [DOI: 10.1002/cne.23302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 11/06/2012] [Accepted: 01/03/2013] [Indexed: 01/06/2023]
|
43
|
Target-derived neurotrophins coordinate transcription and transport of bclw to prevent axonal degeneration. J Neurosci 2013; 33:5195-207. [PMID: 23516285 DOI: 10.1523/jneurosci.3862-12.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Establishment of neuronal circuitry depends on both formation and refinement of neural connections. During this process, target-derived neurotrophins regulate both transcription and translation to enable selective axon survival or elimination. However, it is not known whether retrograde signaling pathways that control transcription are coordinated with neurotrophin-regulated actions that transpire in the axon. Here we report that target-derived neurotrophins coordinate transcription of the antiapoptotic gene bclw with transport of bclw mRNA to the axon, and thereby prevent axonal degeneration in rat and mouse sensory neurons. We show that neurotrophin stimulation of nerve terminals elicits new bclw transcripts that are immediately transported to the axons and translated into protein. Bclw interacts with Bax and suppresses the caspase6 apoptotic cascade that fosters axonal degeneration. The scope of bclw regulation at the levels of transcription, transport, and translation provides a mechanism whereby sustained neurotrophin stimulation can be integrated over time, so that axonal survival is restricted to neurons connected within a stable circuit.
Collapse
|
44
|
Abstract
The hairs of the skin not only function to prevent heat loss but also have important sensory functions. Recent work has now established that each hair of the skin is innervated by one or more of three types of mechanoreceptor ending. Each of these three mechanoreceptor types possesses distinct molecular features and detects distinctive information about skin touch, which is relayed to specific brain locations in a somatotopic fashion.
Collapse
Affiliation(s)
- Stefan G. Lechner
- Department of Neuroscience, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gary R. Lewin
- Department of Neuroscience, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
45
|
Brumovsky PR, La JH, McCarthy CJ, Hökfelt T, Gebhart GF. Dorsal root ganglion neurons innervating pelvic organs in the mouse express tyrosine hydroxylase. Neuroscience 2012; 223:77-91. [PMID: 22858598 DOI: 10.1016/j.neuroscience.2012.07.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/02/2012] [Accepted: 07/18/2012] [Indexed: 12/30/2022]
Abstract
Previous studies in rat and mouse documented that a subpopulation of dorsal root ganglion (DRG) neurons innervating non-visceral tissues express tyrosine hydroxylase (TH). Here we studied whether or not mouse DRG neurons retrogradely traced with Fast Blue (FB) from colorectum or urinary bladder also express immunohistochemically detectable TH. The lumbar sympathetic chain (LSC) and major pelvic ganglion (MPG) were included in the analysis. Previously characterized antibodies against TH, norepinephrine transporter type 1 (NET-1) and calcitonin gene-related peptide (CGRP) were used. On average, ∼14% of colorectal and ∼17% of urinary bladder DRG neurons expressed TH and spanned virtually all neuronal sizes, although more often in the medium-sized to small ranges. Also, they were more abundant in lumbosacral than thoracolumbar DRGs, and often coexpressed CGRP. We also detected several TH-immunoreactive (IR) colorectal and urinary bladder neurons in the LSC and the MPG, more frequently in the former. No NET-1-IR neurons were detected in DRGs, whereas the majority of FB-labeled, TH-IR neurons in the LSC and MPG coexpressed this marker (as did most other TH-IR neurons not labeled from the target organs). TH-IR nerve fibers were detected in all layers of the colorectum and the urinary bladder, with some also reaching the basal mucosal cells. Most TH-IR fibers in these organs lacked CGRP. Taken together, we show: (1) that a previously undescribed population of colorectal and urinary bladder DRG neurons expresses TH, often CGRP but not NET-1, suggesting the absence of a noradrenergic phenotype; and (2) that TH-IR axons/terminals in the colon or urinary bladder, naturally expected to derive from autonomic sources, could also originate from sensory neurons.
Collapse
Affiliation(s)
- P R Brumovsky
- Center for Pain Research, Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | | | | | |
Collapse
|
46
|
Chapman RJ, Lall VK, Maxeiner S, Willecke K, Deuchars J, King AE. Localization of neurones expressing the gap junction protein Connexin45 within the adult spinal dorsal horn: a study using Cx45-eGFP reporter mice. Brain Struct Funct 2012; 218:751-65. [PMID: 22638825 PMCID: PMC3637643 DOI: 10.1007/s00429-012-0426-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 04/30/2012] [Indexed: 12/21/2022]
Abstract
Connexin (Cx) proteins localized to neuronal and glial syncytia provide the ultrastructural components for intercellular communication via gap junctions. In this study, a Cx45 reporter mouse model in which the Cx45 coding sequence is substituted for enhanced green fluorescent protein (eGFP) was used to characterize Cx45 expressing neurones within adult mouse spinal cord. eGFP-immunoreactive (eGFP-IR) cells were localized at all rostro-caudal levels to laminae I-III of the dorsal horn (DH), areas associated with nociception. The neuronal rather than glial phenotype of these cells in DH was confirmed by co-localisation of eGFP-IR with the neuronal marker NeuN. Further immunohistochemical studies revealed that eGFP-IR interneurones co-express the calcium-binding protein calbindin, and to a lesser extent calretinin. In contrast, eGFP-IR profiles did not co-localize with either parvalbumin or GAD-67, both of which are linked to inhibitory interneurones. Staining with the primary afferent markers isolectin-B4 (IB4) and calcitonin gene-related peptide revealed that eGFP-IR somata within laminae I-III receive close appositions from the former, presumed non-peptidergic nociceptive afferents of peripheral origin. The presence of 5-HT terminals in close apposition to eGFP-IR interneuronal somata suggests modulation via descending pathways. These data demonstrate a highly localized expression of Cx45 in a population of interneurones within the mouse superficial dorsal horn. The implications of these data in the context of the putative role of Cx45 and gap junctions in spinal somatosensory processing and pain are discussed.
Collapse
Affiliation(s)
- R J Chapman
- Institute for Membrane and Systems Biology, University of Leeds, Leeds LS2 9JT, UK
| | | | | | | | | | | |
Collapse
|
47
|
Wojtalla A, Herweck F, Granzow M, Klein S, Trebicka J, Huss S, Lerner R, Lutz B, Schildberg FA, Knolle PA, Sauerbruch T, Singer MV, Zimmer A, Siegmund SV. The endocannabinoid N-arachidonoyl dopamine (NADA) selectively induces oxidative stress-mediated cell death in hepatic stellate cells but not in hepatocytes. Am J Physiol Gastrointest Liver Physiol 2012; 302:G873-87. [PMID: 22301114 DOI: 10.1152/ajpgi.00241.2011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The endocannabinoid system is a crucial regulator of hepatic fibrogenesis. We have previously shown that the endocannabinoid anandamide (AEA) is a lipid mediator that blocks proliferation and induces death in hepatic stellate cells (HSCs), the main fibrogenic cell type in the liver, but not in hepatocytes. However, the effects of other endocannabinoids such as N-arachidonoyl dopamine (NADA) have not yet been investigated. The NADA-synthesizing enzyme tyrosine hydroxylase was mainly expressed in sympathetic neurons in portal tracts. Its expression pattern stayed unchanged in normal or fibrotic liver. NADA dose dependently induced cell death in culture-activated primary murine or human HSCs after 2-4 h, starting from 5 μM. Despite caspase 3 cleavage, NADA-mediated cell death showed typical features of necrosis, including ATP depletion. Although the cannabinoid receptors CB1, CB2, or transient receptor potential cation channel subfamily V, member 1 were expressed in HSCs, their pharmacological or genetic blockade failed to inhibit NADA-mediated death, indicating a cannabinoid-receptor-independent mechanism. Interestingly, membrane cholesterol depletion with methyl-β-cyclodextrin inhibited AEA- but not NADA-induced death. NADA significantly induced reactive oxygen species formation in HSCs. The antioxidant glutathione (GSH) significantly decreased NADA-induced cell death. Similar to AEA, primary hepatocytes were highly resistant against NADA-induced death. Resistance to NADA in hepatocytes was due to high levels of GSH, since GSH depletion significantly increased NADA-induced death. Moreover, high expression of the AEA-degrading enzyme fatty acid amide hydrolase (FAAH) in hepatocytes also conferred resistance towards NADA-induced death, since pharmacological or genetic FAAH inhibition significantly augmented hepatocyte death. Thus the selective induction of cell death in HSCs proposes NADA as a novel antifibrogenic mediator.
Collapse
|
48
|
Combinatorial expression of Brn3 transcription factors in somatosensory neurons: genetic and morphologic analysis. J Neurosci 2012; 32:995-1007. [PMID: 22262898 DOI: 10.1523/jneurosci.4755-11.2012] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The three members of the Brn3 family of POU-domain transcription factors (Brn3a/Pou4f1, Brn3b/Pou4f2, and Brn3c/Pou4f3) are expressed in overlapping subsets of visual, auditory/vestibular, and somatosensory neurons. Using unmarked Brn3-null alleles and Brn3 conditional alleles in which gene loss is coupled to expression of an alkaline phosphatase reporter, together with sparse Cre-mediated recombination, we describe the following: (1) the overlapping patterns of Brn3 gene expression in somatosensory neurons; (2) the manner in which these patterns correlate with molecular markers, peripheral afferent arbor morphologies, and dorsal horn projections; and (3) the consequences for these neurons of deleting individual Brn3 genes in the mouse. We observe broad expression of Brn3a among DRG neurons, but subtype-restricted expression of Brn3b and Brn3c. We also observe a nearly complete loss of hair follicle-associated sensory endings among Brn3a(-/-) neurons. Together with earlier analyses of Brn3 gene expression patterns in the retina and inner ear, these experiments suggest a deep functional similarity among primary somatosensory neurons, spiral and vestibular ganglion neurons, and retinal ganglion cells. This work also demonstrates the utility of sparse genetically directed labeling for visualizing individual somatosensory afferent arbors and for defining cell-autonomous mutant phenotypes.
Collapse
|
49
|
Laux A, Delalande F, Mouheiche J, Stuber D, Van Dorsselaer A, Bianchi E, Bezard E, Poisbeau P, Goumon Y. Localization of endogenous morphine-like compounds in the mouse spinal cord. J Comp Neurol 2012; 520:1547-61. [DOI: 10.1002/cne.22811] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Sex differences in the expression of serotonin-synthesizing enzymes in mouse trigeminal ganglia. Neuroscience 2011; 199:429-37. [PMID: 22056601 DOI: 10.1016/j.neuroscience.2011.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 10/18/2011] [Accepted: 10/20/2011] [Indexed: 01/29/2023]
Abstract
Migraine headaches are more prevalent in women and often occur during the early phases of the menstrual cycle, implying a link between migraine and ovarian steroids. Serotonin (5-HT) and its receptors have been proposed to play a key role in the pathophysiology of migraine. The trigeminal ganglion (TG) has been proposed as a site for 5-HT synthesis based on the expression of the rate limiting enzyme in peripheral 5-HT synthesis, tryptophan hydroxylase 1 (TPH1), in female rodent trigeminal ganglia. Tryptophan hydroxylase levels vary over the estrus cycle, however, the expression and potential regulation of other enzymes involved in 5-HT synthesis has not been reported in this tissue. C57/BL6 mice of both sexes expressed TPH1 and aromatic amino acid decarboxylase (AADC), the key enzymes involved in 5-HT synthesis. Levels of both enzymes were significantly higher in juvenile males compared with females. In naturally cycling females TPH1 and AADC expression was highest during proestrus when compared with the other phases of the cycle, and this regulation was mirrored at the mRNA level. In situ hybridization experiments detected TPH1 and AADC mRNA in presumptive neurons in the trigeminal ganglion. Both key enzymes involved in the synthesis of 5-HT are expressed in mouse trigeminal ganglion and are localized to neurons. The levels of these enzymes are dependent on gender and estrus cycle stage, suggesting that ovarian steroids might play a role in the regulation of sensory neuron 5-HT synthesis.
Collapse
|