1
|
Razee A, Banerjee S, Hong J, Magaki S, Fishbein G, Ajijola OA, Umar S. Thoracic Spinal Cord Neuroinflammation as a Novel Therapeutic Target in Pulmonary Hypertension. Hypertension 2023; 80:1297-1310. [PMID: 37092338 PMCID: PMC10192067 DOI: 10.1161/hypertensionaha.122.20782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Pulmonary hypertension (PH) is associated with aberrant sympathoexcitation leading to right ventricular failure (RVF), arrhythmias, and death. Microglial activation and neuroinflammation have been implicated in sympathoexcitation in experimental PH. We recently reported the first evidence of thoracic spinal cord (TSC) neuroinflammation in PH rats. Here, we hypothesize that PH is associated with increased cardiopulmonary afferent signaling leading to TSC-specific neuroinflammation and sympathoexcitation. Furthermore, inhibition of TSC neuroinflammation rescues experimental PH and RVF. METHODS We performed transcriptomic analysis and its validation on the TSC of monocrotaline (n=8) and Sugen hypoxia (n=8) rat models of severe PH-RVF. A group of monocrotaline rats received either daily intrathecal microglial activation inhibitor minocycline (200 μg/kg per day, n=5) or PBS (n=5) from day 14 through 28. Echocardiography and right ventricle-catheterization were performed terminally. Real-time quantitative reverse transcription PCR, immunolocalization, microglia+astrocyte quantification, and terminal deoxynucleotidyl transferase dUTP nick end labeling were assessed. Plasma catecholamines were measured by ELISA. Human spinal cord autopsy samples (Control n=3; pulmonary arterial hypertension n=3) were assessed to validate preclinical findings. RESULTS Increased cardiopulmonary afferent signaling was demonstrated in preclinical and clinical PH. Our findings delineated common dysregulated genes and pathways highlighting neuroinflammation and apoptosis in the remodeled TSC and highlighted increased sympathoexcitation in both rat models. Moreover, we validated significantly increased microglial and astrocytic activation and CX3CL1 expression in TSC of human pulmonary arterial hypertension. Finally, amelioration of TSC neuroinflammation by minocycline in monocrotaline rats inhibited microglial activation, decreased proinflammatory cytokines, sympathetic nervous system activation and significantly attenuated PH and RVF. CONCLUSIONS Targeting neuroinflammation and associated molecular pathways and genes in the TSC may yield novel therapeutic strategies for PH and RVF.
Collapse
Affiliation(s)
- Asif Razee
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Somanshu Banerjee
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jason Hong
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Los Angeles, CA, USA
| | - Shino Magaki
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Greg Fishbein
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Olujimi A. Ajijola
- UCLA Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, Los Angeles, CA, USA
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine Division of Molecular Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
2
|
Development of Neurogenic Detrusor Overactivity after Thoracic Spinal Cord Injury Is Accompanied by Time-Dependent Changes in Lumbosacral Expression of Axonal Growth Regulators. Int J Mol Sci 2022; 23:ijms23158667. [PMID: 35955811 PMCID: PMC9368817 DOI: 10.3390/ijms23158667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Thoracic spinal cord injury (SCI) results in urinary dysfunction, which majorly affects the quality of life of SCI patients. Abnormal sprouting of lumbosacral bladder afferents plays a crucial role in this condition. Underlying mechanisms may include changes in expression of regulators of axonal growth, including chondroitin sulphate proteoglycans (CSPGs), myelin-associated inhibitors (MAIs) and repulsive guidance molecules, known to be upregulated at the injury site post SCI. Here, we confirmed lumbosacral upregulation of the growth-associated protein GAP43 in SCI animals with bladder dysfunction, indicating the occurrence of axonal sprouting. Neurocan and Phosphacan (CSPGs), as well as Nogo-A (MAI), at the same spinal segments were upregulated 7 days post injury (dpi) but returned to baseline values 28 dpi. In turn, qPCR analysis of the mRNA levels for receptors of those repulsive molecules in dorsal root ganglia (DRG) neurons showed a time-dependent decrease in receptor expression. In vitro assays with DRG neurons from SCI rats demonstrated that exposure to high levels of NGF downregulated the expression of some, but not all, receptors for those regulators of axonal growth. The present results, therefore, show significant molecular changes at the lumbosacral cord and DRGs after thoracic lesion, likely critically involved in neuroplastic events leading to urinary impairment.
Collapse
|
3
|
Abstract
N-methyl-d-aspartate receptors (NMDARs) and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are excitatory neurotransmission receptors of the central nervous system and play vital roles in synaptic plasticity. Although not fully elucidated, visceral hypersensitivity is one of the most well-characterized pathophysiologic abnormalities of functional gastrointestinal diseases and appears to be associated with increased synaptic plasticity. In this study, we review the updated findings on the physiology of NMDARs and AMPARs and their relation to visceral hypersensitivity, which propose directions for future research in this field with evolving importance.
Collapse
|
4
|
Natoli S. The multiple faces of ketamine in anaesthesia and analgesia. Drugs Context 2021; 10:dic-2020-12-8. [PMID: 33995542 PMCID: PMC8074779 DOI: 10.7573/dic.2020-12-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Objective Ketamine is an anaesthetic agent with a unique dissociative profile and pharmacological effects ranging from the induction and maintenance of anaesthesia to analgesia and sedation, depending on the dose. This article provides information for the clinical use of ketamine in anaesthesia, in both conventional and special circumstances. Methods This is a non-systematic review of the literature, through a PubMed search up to February 2021. Results With a favourable pharmacokinetic profile, ketamine is used in hospital and prehospital settings for emergency situations. It is suitable for patients with many heart conditions and, unlike other anaesthetics, its potential for cardiorespiratory depression is low. Furthermore, it may be used when venous access is difficult as it may be administered through various routes. Ketamine is the anaesthetic of choice for patients with bronchospasm thanks to its bronchodilatory and anti-inflammatory properties. Conclusion With a favourable pharmacokinetic profile, ketamine is used in hospital and prehospital settings for emergency situations and is suitable for patients with many cardiac and respiratory conditions.
Collapse
Affiliation(s)
- Silvia Natoli
- Department of Clinical Science and Translational Medicine and Unit of Pain Therapy, Polyclinic of Tor Vergata, University of Rome, Tor Vergata, Rome, Italy
| |
Collapse
|
5
|
Nicotinamide adenine dinucleotide phosphate oxidase 2-derived reactive oxygen species contribute to long-term potentiation of C-fiber-evoked field potentials in spinal dorsal horn and persistent mirror-image pain following high-frequency stimulus of the sciatic nerve. Pain 2021; 161:758-772. [PMID: 32195784 DOI: 10.1097/j.pain.0000000000001761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
High-frequency stimulation (HFS) of the sciatic nerve has been reported to produce long-term potentiation (LTP) and long-lasting pain hypersensitivity in rats. However, the central underlying mechanism remains unclear. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) belongs to a group of electron-transporting transmembrane enzymes that produce reactive oxygen species (ROS). Here, we found that NOX2 was upregulated in the lumbar spinal dorsal horn after HFS of the left sciatic nerve, which induced bilateral pain and spinal LTP in both male and female rats. Blocking NOX2 with blocking peptide or shRNA prevented the development of bilateral mechanical allodynia, the induction of spinal LTP, and the phosphorylation of N-methyl-d-aspartate (NMDA) receptor 2B (GluN2B) and nuclear factor kappa-B (NF-κB) p65 after HFS. Moreover, NOX2 shRNA reduced the frequency and amplitude of both spontaneous excitatory postsynaptic currents and miniature excitatory postsynaptic currents in laminar II neurons. Furthermore, 8-hydroxyguanine (8-OHG), an oxidative stress marker, was increased in the spinal dorsal horn. Spinal application of ROS scavenger, Phenyl-N-tert-butylnitrone (PBN), depressed the already established spinal LTP. Spinal application of H2O2, one ROS, induced LTP and bilateral mechanical allodynia, increased the frequency and amplitude of spontaneous excitatory postsynaptic currents in laminar II neurons, and phosphorylated GluN2B and p65 in the dorsal horn. This study provided electrophysiological and behavioral evidence that NOX2-derived ROS in the spinal cord contributed to persistent mirror-image pain by enhancing the synaptic transmission, which was mediated by increasing presynaptic glutamate release and activation of NMDA receptor and NF-κB in the spinal dorsal horn.
Collapse
|
6
|
Spinal Anesthesia Reduces Myocardial Ischemia-triggered Ventricular Arrhythmias by Suppressing Spinal Cord Neuronal Network Interactions in Pigs. Anesthesiology 2021; 134:405-420. [PMID: 33411921 DOI: 10.1097/aln.0000000000003662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cardiac sympathoexcitation leads to ventricular arrhythmias. Spinal anesthesia modulates sympathetic output and can be cardioprotective. However, its effect on the cardio-spinal reflexes and network interactions in the dorsal horn cardiac afferent neurons and the intermediolateral nucleus sympathetic neurons that regulate sympathetic output is not known. The authors hypothesize that spinal bupivacaine reduces cardiac neuronal firing and network interactions in the dorsal horn-dorsal horn and dorsal horn-intermediolateral nucleus that produce sympathoexcitation during myocardial ischemia, attenuating ventricular arrhythmogenesis. METHODS Extracellular neuronal signals from the dorsal horn and intermediolateral nucleus neurons were simultaneously recorded in Yorkshire pigs (n = 9) using a 64-channel high-density penetrating microarray electrode inserted at the T2 spinal cord. Dorsal horn and intermediolateral nucleus neural interactions and known markers of cardiac arrhythmogenesis were evaluated during myocardial ischemia and cardiac load-dependent perturbations with intrathecal bupivacaine. RESULTS Cardiac spinal neurons were identified based on their response to myocardial ischemia and cardiac load-dependent perturbations. Spinal bupivacaine did not change the basal activity of cardiac neurons in the dorsal horn or intermediolateral nucleus. After bupivacaine administration, the percentage of cardiac neurons that increased their activity in response to myocardial ischemia was decreased. Myocardial ischemia and cardiac load-dependent stress increased the short-term interactions between the dorsal horn and dorsal horn (324 to 931 correlated pairs out of 1,189 pairs, P < 0.0001), and dorsal horn and intermediolateral nucleus neurons (11 to 69 correlated pairs out of 1,135 pairs, P < 0.0001). Bupivacaine reduced this network response and augmentation in the interactions between dorsal horn-dorsal horn (931 to 38 correlated pairs out of 1,189 pairs, P < 0.0001) and intermediolateral nucleus-dorsal horn neurons (69 to 1 correlated pairs out of 1,135 pairs, P < 0.0001). Spinal bupivacaine reduced shortening of ventricular activation recovery interval and dispersion of repolarization, with decreased ventricular arrhythmogenesis during acute ischemia. CONCLUSIONS Spinal anesthesia reduces network interactions between dorsal horn-dorsal horn and dorsal horn-intermediolateral nucleus cardiac neurons in the spinal cord during myocardial ischemia. Blocking short-term coordination between local afferent-efferent cardiac neurons in the spinal cord contributes to a decrease in cardiac sympathoexcitation and reduction of ventricular arrhythmogenesis. EDITOR’S PERSPECTIVE
Collapse
|
7
|
Pain modulatory properties of Phoneutria nigriventer crude venom and derived peptides: A double-edged sword. Toxicon 2020; 185:120-128. [PMID: 32668276 DOI: 10.1016/j.toxicon.2020.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022]
Abstract
Phoneutria nigriventer venom (PNV) is a complex mixture of toxins exerting multiple pharmacological effects that ultimately result in severe local pain at the site of the bite. It has been proposed that the PNV-induced pain is mediated by both peripheral and central mechanisms. The nociception triggered by PNV is peripherally mediated by the activation of B2, 5-HT4, NMDA, AMPA, NK1, and NK2 receptors, as well as TTXS-Na+, ASIC, and TRPV1 channels. The activation of tachykinin, glutamate and CGRP receptors along with the production of inflammatory mediators are, at least partially, responsible for the central component of pain. Despite its well established pro-nociceptive properties, PNV contains some toxins with antinociceptive activity, which have been studied in the last few years. The toxins ω-CNTX-Pn4a, ω-CNTX-Pn2a, ω-CNTX-Pn3a, κ-CNTX-Pn1a, U7-CNTX-Pn1a, δ-CNTX-Pn1a, and Γ-CNTX-Pn1a from PNV, as well as the semi-synthetic peptide PnPP-19 have been tested in different experimental models of pain showing consistent antinociceptive properties. This review aims to discuss the pro- and antinociceptive actions of PNV and its toxins, highlighting possible mechanisms involved in these apparently dualistic properties.
Collapse
|
8
|
Ion Channels Involved in Substance P-Mediated Nociception and Antinociception. Int J Mol Sci 2019; 20:ijms20071596. [PMID: 30935032 PMCID: PMC6479580 DOI: 10.3390/ijms20071596] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/26/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Substance P (SP), an 11-amino-acid neuropeptide, has long been considered an effector of pain. However, accumulating studies have proposed a paradoxical role of SP in anti-nociception. Here, we review studies of SP-mediated nociception and anti-nociception in terms of peptide features, SP-modulated ion channels, and differential effector systems underlying neurokinin 1 receptors (NK1Rs) in differential cell types to elucidate the effect of SP and further our understanding of SP in anti-nociception. Most importantly, understanding the anti-nociceptive SP-NK1R pathway would provide new insights for analgesic drug development.
Collapse
|
9
|
Sandweiss AJ, McIntosh MI, Moutal A, Davidson-Knapp R, Hu J, Giri AK, Yamamoto T, Hruby VJ, Khanna R, Largent-Milnes TM, Vanderah TW. Genetic and pharmacological antagonism of NK 1 receptor prevents opiate abuse potential. Mol Psychiatry 2018; 23:1745-1755. [PMID: 28485408 PMCID: PMC5680162 DOI: 10.1038/mp.2017.102] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/03/2017] [Accepted: 03/16/2017] [Indexed: 11/29/2022]
Abstract
Development of an efficacious, non-addicting analgesic has been challenging. Discovery of novel mechanisms underlying addiction may present a solution. Here we target the neurokinin system, which is involved in both pain and addiction. Morphine exerts its rewarding actions, at least in part, by inhibiting GABAergic input onto substance P (SP) neurons in the ventral tegmental area (VTA), subsequently increasing SP release onto dopaminergic neurons. Genome editing of the neurokinin 1 receptor (NK1R) in the VTA renders morphine non-rewarding. Complementing our genetic approach, we demonstrate utility of a bivalent pharmacophore with dual activity as a μ/δ opioid agonist and NK1R antagonist in inhibiting nociception in an animal model of acute pain while lacking any positive reinforcement. These data indicate that dual targeting of the dopaminergic reward circuitry and pain pathways with a multifunctional opioid agonist-NK1R antagonist may be an efficacious strategy in developing future analgesics that lack abuse potential.
Collapse
MESH Headings
- Acute Pain/drug therapy
- Acute Pain/metabolism
- Analgesics/pharmacology
- Animals
- CRISPR-Cas Systems
- Disease Models, Animal
- Dopamine/metabolism
- Escherichia coli
- Gene Knockdown Techniques
- Male
- Mice, Inbred ICR
- Morphine/pharmacology
- Neurokinin-1 Receptor Antagonists/pharmacology
- Nociceptive Pain/drug therapy
- Nociceptive Pain/metabolism
- Opioid-Related Disorders/genetics
- Opioid-Related Disorders/metabolism
- Opioid-Related Disorders/prevention & control
- Rats, Sprague-Dawley
- Receptors, Neurokinin-1/genetics
- Receptors, Neurokinin-1/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Reward
- Substance P/metabolism
- Ventral Tegmental Area/drug effects
- Ventral Tegmental Area/metabolism
Collapse
Affiliation(s)
- A J Sandweiss
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - M I McIntosh
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - A Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R Davidson-Knapp
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - J Hu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - A K Giri
- Department of Chemistry, University of Arizona, Tucson, AZ, USA
| | - T Yamamoto
- Department of Chemistry, University of Arizona, Tucson, AZ, USA
| | - V J Hruby
- Department of Chemistry, University of Arizona, Tucson, AZ, USA
| | - R Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - T M Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - T W Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
10
|
Abstract
The mechanisms by which noxious stimuli produce the sensation of pain in animals are complex. Noxious stimuli are transduced at the periphery and transmitted to the CNS, where this information is subject to considerable modulation. Finally, the information is projected to the brain where it is perceived as pain. Additionally, plasticity can develop in the pain pathway and hyperalgesia and allodynia may develop through sensitisation both peripherally and centrally. A large number of different ion channels, receptors, and cell types are involved in pain perception, and it is hoped that through a better understanding of these, new and refined treatments for pain will result.
Collapse
Affiliation(s)
- A Bell
- School of Veterinary Medicine, University of Glasgow, Glasgow, G61 1QH, UK.
| |
Collapse
|
11
|
Fazzari J, Linher-Melville K, Singh G. Tumour-Derived Glutamate: Linking Aberrant Cancer Cell Metabolism to Peripheral Sensory Pain Pathways. Curr Neuropharmacol 2018; 15:620-636. [PMID: 27157265 PMCID: PMC5543678 DOI: 10.2174/1570159x14666160509123042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/16/2016] [Accepted: 04/17/2016] [Indexed: 01/22/2023] Open
Abstract
Background Chronic pain is a major symptom that develops in cancer patients, most commonly emerging during advanced stages of the disease. The nature of cancer-induced pain is complex, and the efficacy of current therapeutic interventions is restricted by the dose-limiting side-effects that accompany common centrally targeted analgesics. Methods This review focuses on how up-regulated glutamate production and export by the tumour converge at peripheral afferent nerve terminals to transmit nociceptive signals through the transient receptor cation channel, TRPV1, thereby initiating central sensitization in response to peripheral disease-mediated stimuli. Results Cancer cells undergo numerous metabolic changes that include increased glutamine catabolism and over-expression of enzymes involved in glutaminolysis, including glutaminase. This mitochondrial enzyme mediates glutaminolysis, producing large pools of intracellular glutamate. Up-regulation of the plasma membrane cystine/glutamate antiporter, system xc-, promotes aberrant glutamate release from cancer cells. Increased levels of extracellular glutamate have been associated with the progression of cancer-induced pain and we discuss how this can be mediated by activation of TRPV1. Conclusion With a growing population of patients receiving inadequate treatment for intractable pain, new targets need to be considered to better address this largely unmet clinical need for improving their quality of life. A better understanding of the mechanisms that underlie the unique qualities of cancer pain will help to identify novel targets that are able to limit the initiation of pain from a peripheral source–the tumour.
Collapse
Affiliation(s)
| | | | - Gurmit Singh
- Department of Pathology and Molecular Medicine; Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON. Canada
| |
Collapse
|
12
|
Abstract
Novel minimally invasive treatment options for the management of male lower urinary tract symptoms (LUTS) due to bladder outlet obstruction (BOO) aim to provide equal efficacy compared to standard techniques with a more favourable safety profile. The preservation of all aspects of male sexual function including antegrade ejaculation is increasingly important to patients. It should be ideally performed in an outpatient setting under local anesthesia to assure a short recovery time. Novel injectables for intraprostatic application (botulinum neurotoxin A, NX-1207, PRX302) have emerged and first phase III results could not confirm the promising initial data. For mechanical devices like Urolift® the early and mid-term benefits demonstrate a rapid and durable symptom relief without compromising sexual function. Novel innovative procedures like aquablation (AquaBeam®) are entering the scene, but their feasibility, efficacy and safety still need to be addressed in randomized controlled trials.
Collapse
|
13
|
Merkel Cell-Driven BDNF Signaling Specifies SAI Neuron Molecular and Electrophysiological Phenotypes. J Neurosci 2016; 36:4362-76. [PMID: 27076431 DOI: 10.1523/jneurosci.3781-15.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/07/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The extent to which the skin instructs peripheral somatosensory neuron maturation is unknown. We studied this question in Merkel cell-neurite complexes, where slowly adapting type I (SAI) neurons innervate skin-derived Merkel cells. Transgenic mice lacking Merkel cells had normal dorsal root ganglion (DRG) neuron numbers, but fewer DRG neurons expressed the SAI markers TrkB, TrkC, and Ret. Merkel cell ablation also decreased downstream TrkB signaling in DRGs, and altered the expression of genes associated with SAI development and function. Skin- and Merkel cell-specific deletion of Bdnf during embryogenesis, but not postnatal Bdnf deletion or Ntf3 deletion, reproduced these results. Furthermore, prototypical SAI electrophysiological signatures were absent from skin regions where Bdnf was deleted in embryonic Merkel cells. We conclude that BDNF produced by Merkel cells during a precise embryonic period guides SAI neuron development, providing the first direct evidence that the skin instructs sensory neuron molecular and functional maturation. SIGNIFICANCE STATEMENT Peripheral sensory neurons show incredible phenotypic and functional diversity that is initiated early by cell-autonomous and local environmental factors found within the DRG. However, the contribution of target tissues to subsequent sensory neuron development remains unknown. We show that Merkel cells are required for the molecular and functional maturation of the SAI neurons that innervate them. We also show that this process is controlled by BDNF signaling. These findings provide new insights into the regulation of somatosensory neuron development and reveal a novel way in which Merkel cells participate in mechanosensation.
Collapse
|
14
|
Huang D, Huang S, Gao H, Liu Y, Qi J, Chen P, Wang C, Scragg JL, Vakurov A, Peers C, Du X, Zhang H, Gamper N. Redox-Dependent Modulation of T-Type Ca(2+) Channels in Sensory Neurons Contributes to Acute Anti-Nociceptive Effect of Substance P. Antioxid Redox Signal 2016; 25:233-51. [PMID: 27306612 PMCID: PMC4971421 DOI: 10.1089/ars.2015.6560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 05/26/2016] [Accepted: 06/14/2016] [Indexed: 02/05/2023]
Abstract
AIMS Neuropeptide substance P (SP) is produced and released by a subset of peripheral sensory neurons that respond to tissue damage (nociceptors). SP exerts excitatory effects in the central nervous system, but peripheral SP actions are still poorly understood; therefore, here, we aimed at investigating these peripheral mechanisms. RESULTS SP acutely inhibited T-type voltage-gated Ca(2+) channels in nociceptors. The effect was mediated by neurokinin 1 (NK1) receptor-induced stimulation of intracellular release of reactive oxygen species (ROS), as it can be prevented or reversed by the reducing agent dithiothreitol and mimicked by exogenous or endogenous ROS. This redox-mediated T-type Ca(2+) channel inhibition operated through the modulation of CaV3.2 channel sensitivity to ambient zinc, as it can be prevented or reversed by zinc chelation and mimicked by exogenous zinc. Elimination of the zinc-binding site in CaV3.2 rendered the channel insensitive to SP-mediated inhibition. Importantly, peripherally applied SP significantly reduced bradykinin-induced nociception in rats in vivo; knock-down of CaV3.2 significantly reduced this anti-nociceptive effect. This atypical signaling cascade shared the initial steps with the SP-mediated augmentation of M-type K(+) channels described earlier. INNOVATION Our study established a mechanism underlying the peripheral anti-nociceptive effect of SP whereby this neuropeptide produces ROS-dependent inhibition of pro-algesic T-type Ca(2+) current and concurrent enhancement of anti-algesic M-type K(+) current. These findings will lead to a better understanding of mechanisms of endogenous analgesia. CONCLUSION SP modulates T-type channel activity in nociceptors by a redox-dependent tuning of channel sensitivity to zinc; this novel modulatory pathway contributes to the peripheral anti-nociceptive effect of SP. Antioxid. Redox Signal. 25, 233-251.
Collapse
Affiliation(s)
- Dongyang Huang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Haixia Gao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yani Liu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Pingping Chen
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Caixue Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Jason L. Scragg
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Alexander Vakurov
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Chris Peers
- Leeds Institute of Cardiovascular and Metabolic Medicine (LICAMM), Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, P.R. China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
15
|
Maixner DW, Yan X, Hooks SB, Weng HR. AMPKα1 knockout enhances nociceptive behaviors and spinal glutamatergic synaptic activities via production of reactive oxygen species in the spinal dorsal horn. Neuroscience 2016; 326:158-169. [PMID: 27058143 DOI: 10.1016/j.neuroscience.2016.03.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/15/2022]
Abstract
Emerging studies have shown that pharmacological activation of adenosine monophosphate-activated protein kinase (AMPK) produces potent analgesic effects in different animal pain models. Currently, the spinal molecular and synaptic mechanism by which AMPK regulates the pain signaling system remains unclear. To address this issue, we utilized the Cre-LoxP system to conditionally knockout the AMPKα1 gene in the nervous system of mice. We demonstrated that AMPKα1 is imperative for maintaining normal nociception, and mice deficient for AMPKα1 exhibit mechanical allodynia. This is concomitantly associated with increased glutamatergic synaptic activities in neurons located in the superficial spinal dorsal horn, which results from the increased glutamate release from presynaptic terminals and function of ligand-gated glutamate receptors at the postsynaptic neurons. Additionally, AMPKα1 knockout mice have increased activities of extracellular signal-regulated kinases (ERK) and p38 mitogen-activated protein kinases (p38), as well as elevated levels of interleukin-1β (IL-1β), reactive oxygen species (ROS), and heme oxygenase 1 (HO-1) in the spinal dorsal horn. Systemic administration of a non-specific ROS scavenger (phenyl-N-tert-butylnitrone, PBN) or a HO-1 activator (Cobalt protoporphyrin IX, CoPP) attenuated allodynia in AMPKα1 knockout mice. Bath-perfusion of the ROS scavenger or HO-1 activator effectively attenuated the increased ROS levels and glutamatergic synaptic activities in the spinal dorsal horn. Our findings suggest that ROS are the key down-stream signaling molecules mediating the behavioral hypersensitivity in AMPKα1 knockout mice. Thus, targeting AMPKα1 may represent an effective approach for the treatment of pathological pain conditions associated with neuroinflammation at the spinal dorsal horn.
Collapse
Affiliation(s)
- Dylan W Maixner
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| | - Xisheng Yan
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA.,Department of Cardiovascular Medicine, The Third Hospital of Wuhan, Wuhan 430074, Hubei Province, China.,Department of Endocrinology and Metabolism, Shanghai Tenth People's hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Shelley B Hooks
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| | - Han-Rong Weng
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, Georgia 30602, USA
| |
Collapse
|
16
|
Saidi M, Kamali S, Ruiz AO, Beaudry F. Tachykinins Processing is Significantly Impaired in PC1 and PC2 Mutant Mouse Spinal Cord S9 Fractions. Neurochem Res 2015; 40:2304-16. [DOI: 10.1007/s11064-015-1720-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 08/18/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
|
17
|
Liu M, Kay JC, Shen S, Qiao LY. Endogenous BDNF augments NMDA receptor phosphorylation in the spinal cord via PLCγ, PKC, and PI3K/Akt pathways during colitis. J Neuroinflammation 2015; 12:151. [PMID: 26289587 PMCID: PMC4545933 DOI: 10.1186/s12974-015-0371-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/06/2015] [Indexed: 12/18/2022] Open
Abstract
Background Spinal central sensitization is an important process in the generation and maintenance of visceral hypersensitivity. The release of brain-derived neurotrophic factor (BDNF) from the primary afferent neurons to the spinal cord contributes to spinal neuronal plasticity and increases neuronal activity and synaptic efficacy. The N-Methyl-D-aspartic acid (NMDA) receptor possesses ion channel properties, and its activity is modulated by phosphorylation of its subunits including the NMDA receptor 1 (NR1). Methods Colonic inflammation was induced by a single dose of intracolonic instillation of tri-nitrobenzene sulfonic acid (TNBS). NR1 phosphorylation by BDNF in vivo and in culture was examined by western blot and immunohistochemistry. Signal transduction was studied by direct examination and use of specific inhibitors. Results During colitis, the level of NR1 phospho-Ser896 was increased in the dorsal horn region of the L1 and S1 spinal cord; this increase was attenuated by injection of BDNF neutralizing antibody to colitic animals (36 μg/kg, intravenous (i.v.)) and was also reduced in BDNF+/− rat treated with TNBS. Signal transduction examination showed that the extracellular signal-regulated kinase (ERK) activation was not involved in BDNF-induced NR1 phosphorylation. In contrast, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway mediated BDNF-induced NR1 phosphorylation in vivo and in culture; this is an additional pathway to the phospholipase C-gamma (PLCγ) and the protein kinase C (PKC) that was widely considered to phosphorylate NR1 at Ser896. In spinal cord culture, the inhibitors to PLC (U73122), PKC (bisindolylmaleimide I), and PI3K (LY294002), but not MEK (PD98059) blocked BDNF-induced NR1 phosphorylation. In animals with colitis, treatment with LY294002 (50 μg/kg, i.v.) blocked the Akt activity as well as NR1 phosphorylation at Ser896 in the spinal cord. Conclusion BDNF participates in colitis-induced spinal central sensitization by up-regulating NR1 phosphorylation at Ser896. The PI3K/Akt pathway, in addition to PLCγ and PKC, mediates BDNF action in the spinal cord during colitis. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0371-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miao Liu
- Department of Physiology and Biophysics, Internal Medicine Gastroenterology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street MMRB 5046, Richmond, VA, 23298-0551, USA.
| | - Jarren C Kay
- Department of Physiology and Biophysics, Internal Medicine Gastroenterology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street MMRB 5046, Richmond, VA, 23298-0551, USA.
| | - Shanwei Shen
- Department of Physiology and Biophysics, Internal Medicine Gastroenterology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street MMRB 5046, Richmond, VA, 23298-0551, USA.
| | - Li-Ya Qiao
- Department of Physiology and Biophysics, Internal Medicine Gastroenterology, Virginia Commonwealth University School of Medicine, 1220 East Broad Street MMRB 5046, Richmond, VA, 23298-0551, USA.
| |
Collapse
|
18
|
West S, Bannister K, Dickenson A, Bennett D. Circuitry and plasticity of the dorsal horn – Toward a better understanding of neuropathic pain. Neuroscience 2015; 300:254-75. [DOI: 10.1016/j.neuroscience.2015.05.020] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 11/24/2022]
|
19
|
Magistro G, Stief CG, Gratzke C. New intraprostatic injectables and prostatic urethral lift for male LUTS. Nat Rev Urol 2015. [PMID: 26195444 DOI: 10.1038/nrurol.2015.169] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Treatment modalities for male lower urinary tract symptoms (LUTS) comprise a broad spectrum of medical and surgical options. Interest is growing in minimally invasive treatment options, which should ideally be performed in an outpatient setting and have a short recovery time, durable efficacy and a good safety profile. The preservation of all aspects of sexual function, including antegrade ejaculation, seems to be increasingly important for patients. Initial experimental data on new minimally invasive procedures-such as the intraprostatic injection of novel agents including botulinum neurotoxin A (BoNT-A), NX-1207 and PRX302-were promising, but clinical trials have not confirmed the findings. Trials of the mechanical prostatic urethral lift device-Urolift(®) (Neotract, Inc., USA)-have been positive, but further long-term results are needed to confirm its beneficial effects.
Collapse
Affiliation(s)
- Giuseppe Magistro
- Urologische Klinik und Poliklinik, Klinikum der Universität München-Campus Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Christian G Stief
- Urologische Klinik und Poliklinik, Klinikum der Universität München-Campus Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Christian Gratzke
- Urologische Klinik und Poliklinik, Klinikum der Universität München-Campus Grosshadern, Ludwig-Maximilians-Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| |
Collapse
|
20
|
Abstract
Morphine has been widely used for the treatment of acute, chronic, and cancer pain and is considered the strongest analgesic in clinical care. Conversely, morphine-induced analgesia may be accompanied by several side effects. Animal studies have demonstrated that low doses of morphine administered intrathecally can produce reliable analgesia for thermal, mechanical, and chemical nociceptive stimulation. On the other hand, high doses of morphine administered intrathecally may induce spontaneous nociceptive responses such as scratching, biting, and licking in mice as well as agitation and vocalization in rats. In addition, similar nociceptive responses including hyperalgesia, allodynia, and myoclonus have been observed in humans following intrathecal or systemic administration of high-dose morphine. It has been suggested that the spontaneous nociceptive behaviors evoked by high-dose morphine may be mediated by a non-opioid mechanism that is not yet fully understood. This review describes the mechanisms of spontaneous nociceptive behaviors evoked by high-dose morphine focusing on the neurotransmitters/neuromodulators released from primary afferent fibers.
Collapse
Affiliation(s)
- Chizuko Watanabe
- Department of Physiology and Anatomy, Tohoku Pharmaceutical University
| |
Collapse
|
21
|
Nakamura Y, Morioka N, Zhang FF, Hisaoka-Nakashima K, Nakata Y. Downregulation of connexin36 in mouse spinal dorsal horn neurons leads to mechanical allodynia. J Neurosci Res 2015; 93:584-91. [PMID: 25400139 DOI: 10.1002/jnr.23515] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/24/2014] [Accepted: 10/13/2014] [Indexed: 11/08/2022]
Abstract
Connexin36 (Cx36), a component of neuronal gap junctions, is crucial for interneuronal communication and regulation. Gap junction dysfunction underlies neurological disorders, including chronic pain. Following a peripheral nerve injury, Cx36 expression in the ipsilateral spinal dorsal horn was markedly decreased over time, which paralleled the time course of hind paw tactile allodynia. Intrathecal (i.t.) injection of Cx36 siRNA (1 and 5 pg) significantly reduced the expression of Cx36 protein in the lumbar spinal cord, peaking 3 days after the injection, which corresponded with the onset of hind paw tactile allodynia. It is possible that some of the tactile allodynia resulting from Cx36 downregulation could be mediated through excitatory neuromodulators, such as glutamate and substance P. The Cx36 knockdown-evoked tactile allodynia was significantly attenuated by i.t. treatment with the N-methyl-D-aspartate glutamate receptor antagonist MK-801 but not the substance P receptor antagonist CP96345. Immunohistochemistry showed that Cx36 was colocalized with glycine transporter-2, a marker for inhibitory glycinergic spinal interneurons, but not with glutamate decarboxylase 67, a marker for inhibitory GABAergic spinal interneurons. The results indicate that spinal inhibition through glycinergic interneurons is reduced, leading to increased glutamatergic neurotransmission, as a result of Cx36 downregulation. The current data suggest that gap junction dysfunction underlies neuropathic pain and further suggest a novel target for the development of analgesics.
Collapse
Affiliation(s)
- Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
22
|
Bansal P, Baduni N, Bhalla J, Mahawar B. A comparative evaluation of magnesium sulphate and nitroglycerine as potential adjuncts to lidocaine in intravenous regional anaesthesia. Int J Crit Illn Inj Sci 2015; 5:27-31. [PMID: 25810961 PMCID: PMC4366824 DOI: 10.4103/2229-5151.152324] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction: This randomized control trial was carried out to evaluate and compare the efficacy of magnesium sulphate and nitroglycerine (NTG) as adjuncts to lidocaine in intravenous regional anesthesia (IVRA). Materials and Methods: Seventy-five, ASA grade I and II patients, aged between 20–50 years, scheduled for hand and forearm surgery were selected and entered randomly into three study groups. Patients in group C received 3 mg/kg of preservative free lidocaine 2% diluted with saline to a total volume of 40 ml. Patients in group M received 3 mg/kg of preservative free lidocaine 2% mixed with 6 ml of 25% magnesium sulphate (1.5 g) diluted with saline to a total volume of 40 ml. Patients in group N received 3 mg/kg of preservative free lidocaine 2% mixed with 200 μg of nitroglycerine diluted with saline to a total volume of 40 ml. Sensory and motor block onset and recovery time, tourniquet pain onset time, intraoperative fentanyl requirement, the total number of patients requiring rescue analgesia and the time to first analgesia requirement, intra-operative and postoperative degree of analgesia were evaluated. Results: The sensory and motor block onset times were shorter in group M and N as compared to group C (P- = 0.004, 0.0036 for sensory block, 0.021, 0.038 for motor block. The mean time of onset of sensory block was earliest in group M and the mean time of onset of motor block was earliest in group N. Mean time of onset of tourniquet pain in the three groups was similar in groups M and N. The sensory and motor block recovery time were significantly prolonged in M and N group as compared to group C (P < 0.001). Intraoperative fentanyl requirement (P value– = 0.041), the total number of patients requiring rescue analgesia (P value = 0.009) and the time to first analgesia requirement (P value = 0.038) were lower in group M. Conclusion: The addition of both magnesium suphate and nitroglycerin (NTG) to lidocaine for intravenous regional anesthesia (IVRA) leads to early onset of sensory block and prolonged postoperative analgesia, with no side effects.
Collapse
Affiliation(s)
- Pooja Bansal
- Department of Anesthesiology and Intensive Care, Government Medical College, Jammu, Jammu and Kashmir, India
| | - Neha Baduni
- Department of Anesthesiology and Intensive Care, Employees' State Insurance Corporation, Rohini, New Delhi, India
| | - Jyoti Bhalla
- Department of Anesthesiology and Intensive Care, Employees' State Insurance Corporation, Rohini, New Delhi, India
| | - Bablesh Mahawar
- Department of Anesthesiology and Intensive Care, Employees' State Insurance Corporation, Rohini, New Delhi, India
| |
Collapse
|
23
|
|
24
|
Blockade of glutamate release by botulinum neurotoxin type A in humans: a dermal microdialysis study. Pain Res Manag 2015; 19:126-32. [PMID: 24851237 DOI: 10.1155/2014/410415] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND The analgesic action of botulinum neurotoxin type A (BoNTA) has been linked to the blockade of peripheral release of neuropeptides and neurotransmitters in animal models; however, there is no direct evidence of this in humans. OBJECTIVES To investigate the effect of BoNTA on glutamate release in humans, using an experimental model of pain and sensitization provoked by capsaicin plus mild heat. METHODS Twelve healthy volunteers (six men, six women) were pretreated with BoNTA (10 U) on the volar forearm and with a saline control on the contralateral side. Dermal microdialysis was applied one week later to collect interstitial samples before and after the application of a capsaicin patch (8%) plus mild heat (40°C⁄60 min) to provoke glutamate release, pain and vasodilation. Samples were collected every hour for 3 h using linear microdialysis probes (10 mm, 100 kD). Dialysate was analyzed for glutamate concentration. Pain intensity and skin vasomotor reactions (temperature and blood flow changes) were also recorded. RESULTS BoNTA significantly reduced glutamate release compared with saline (P<0.05). The provoked pain intensity was lower in the BoNTA-pretreated arm (P<0.01). The reduction in pain scores was not correlated with glutamate level. Cutaneous blood flow (P<0.05), but not cutaneous temperature (P≥0.05), was significantly reduced by BoNTA. There was a correlation between glutamate level and skin blood flow (r=0.58⁄P<0.05) but not skin temperature (P≥0.05). No differences according to sex were observed in any response. CONCLUSIONS The present study provided the first direct evidence supporting the inhibitory effect of BoNTA on glutamate release in human skin, which is potentially responsible for some of the analgesic action of BoNTA.
Collapse
|
25
|
Vaaga CE, Borisovska M, Westbrook GL. Dual-transmitter neurons: functional implications of co-release and co-transmission. Curr Opin Neurobiol 2014; 29:25-32. [PMID: 24816154 DOI: 10.1016/j.conb.2014.04.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/16/2014] [Accepted: 04/17/2014] [Indexed: 11/18/2022]
Abstract
Co-transmission, the ability of a neuron to release multiple transmitters, has long been recognized in selected circuits. However, the release of multiple primary neurotransmitters from a single neuron is only beginning to be appreciated. Here we consider recent examples of co-transmission as well as co-release-the packaging of multiple neurotransmitters into a single vesicle. The properties associated with each mode of release greatly enhance the possible action of such neurons within circuits. The functional importance of dual- (or multi-) transmitter neurons extends beyond actions on postsynaptic receptors, due in part to differential spatial and temporal profiles of each neurotransmitter. Recent evidence also suggests that the dual-transmitter phenotype can be dynamically regulated during development and following injury or disease.
Collapse
Affiliation(s)
- Christopher E Vaaga
- Neuroscience Graduate Program, Oregon Health and Science University, Portland, OR, USA; Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Maria Borisovska
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Gary L Westbrook
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
26
|
Trigeminal Medullary Dorsal Horn Neurons Activated by Nasal Stimulation Coexpress AMPA, NMDA, and NK1 Receptors. ISRN NEUROSCIENCE 2013; 2013:152567. [PMID: 24967301 PMCID: PMC4045565 DOI: 10.1155/2013/152567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/07/2013] [Indexed: 11/25/2022]
Abstract
Afferent information initiating the cardiorespiratory responses during nasal stimulation projects from the nasal passages to neurons within the trigeminal medullary dorsal horn (MDH) via the anterior ethmoidal nerve (AEN). Central AEN terminals are thought to release glutamate to activate the MDH neurons. This study was designed to determine which neurotransmitter receptors (AMPA, kainate, or NMDA glutamate receptor subtypes or the Substance P receptor NK1) are expressed by these activated MDH neurons. Fos was used as a neuronal marker of activated neurons, and immunohistochemistry combined with epifluorescent microscopy was used to determine which neurotransmitter receptor subunits were coexpressed by activated MDH neurons. Results indicate that, during nasal stimulation with ammonia vapors in urethane-anesthetized Sprague-Dawley rats, activated neurons within the superficial MDH coexpress the AMPA glutamate receptor subunits GluA1 (95.8%) and GluA2/3 (88.2%), the NMDA glutamate receptor subunits GluN1 (89.1%) and GluN2A (41.4%), and NK1 receptors (64.0%). It is therefore likely that during nasal stimulation the central terminals of the AEN release glutamate and substance P that then produces activation of these MDH neurons. The involvement of AMPA and NMDA receptors may mediate fast and slow neurotransmission, respectively, while NK1 receptor involvement may indicate activation of a nociceptive pathway.
Collapse
|
27
|
Chopra K, Arora V. An intricate relationship between pain and depression: clinical correlates, coactivation factors and therapeutic targets. Expert Opin Ther Targets 2013; 18:159-76. [PMID: 24295272 DOI: 10.1517/14728222.2014.855720] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION An apparent clinical relationship between pain and depression has long been recognized, which makes an enormous impact on the individual health care. At present, the practical implication of such overlapping symptomatology between pain and depression is not clear, but the prevalence estimates for depression are substantially inflated among patients with chronic pain and vice versa. This interaction has been labeled as the depression-pain syndrome or depression-pain dyad. AREAS COVERED This article discusses the neurobiological substrates and neuroanatomical pathways involved in pain-depression dyad along with newer therapeutic targets. EXPERT OPINION Several key themes emerged from our review of the relationship between depression and pain. First, the diagnosis of depression in pain or vice versa is particularly challenging, and the development of better diagnostic framework that involves both pain and depression is particularly required. Secondly, the entwined relationship between pain and depression supports the possibility of common coactivating factors that results in their neurophysiological overlap. A broad understanding of the role played by the central nervous system (CNS) in the processing of pain and depression may eventually lead to the introduction of triple reuptake inhibitors, agomelatine, vilazodone and ketamine with novel mechanism of action, hence appear to be of promising potential for pain with depression.
Collapse
Affiliation(s)
- Kanwaljit Chopra
- Panjab University, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Pharmacology Research Laboratory , Chandigarh-160 014 , India +91 172 2534105 ; +91 172 2541142 ;
| | | |
Collapse
|
28
|
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
|
29
|
Kay JC, Xia CM, Liu M, Shen S, Yu SJ, Chung C, Qiao LY. Endogenous PI3K/Akt and NMDAR act independently in the regulation of CREB activity in lumbosacral spinal cord in cystitis. Exp Neurol 2013; 250:366-75. [PMID: 24184018 DOI: 10.1016/j.expneurol.2013.10.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 02/08/2023]
Abstract
The integral interaction of signaling components in the regulation of visceral inflammation-induced central sensitization in the spinal cord has not been well studied. Here we report that phosphoinositide 3-kinase (PI3K)-dependent Akt activation and N-methyl-d-aspartic acid receptor (NMDAR) in lumbosacral spinal cord independently regulate the activation of cAMP response element-binding protein (CREB) in vivo in a rat visceral pain model of cystitis induced by intraperitoneal injection of cyclophosphamide (CYP). We demonstrate that suppression of endogenous PI3K/Akt activity with a potent PI3K inhibitor LY294002 reverses CYP-induced phosphorylation of CREB, however, it has no effect on CYP-induced phosphorylation of NR1 at Ser(897) and Ser(896); conversely, inhibition of NMDAR in vivo with MK801 fails to block CYP-induced Akt activation but significantly attenuates CYP-induced CREB phosphorylation in lumbosacral spinal cord. This novel interrelationship of PI3K/Akt, NMDAR, and CREB activation in lumbosacral spinal cord is further confirmed in an ex vivo spinal slice culture system exposed to an excitatory neurotransmitter calcitonin gene-related peptide (CGRP). Consistently we found that CGRP-triggered CREB activation can be blocked by both PI3K inhibitor LY294002 and NMDAR antagonists MK801 and D-AP5. However, CGRP-triggered Akt activation cannot be blocked by MK801 or D-AP5; vice versa, LY294002 pretreatment that suppresses the Akt activity fails to reverse CGRP-elicited NR1 phosphorylation. These results suggest that PI3K/Akt and NMDAR independently regulate spinal plasticity in visceral pain model, and target of a single pathway is necessary but not sufficient in treatment of visceral hypersensitivity.
Collapse
Affiliation(s)
- Jarren C Kay
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chun-Mei Xia
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Miao Liu
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Shanwei Shen
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Sharon J Yu
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chulwon Chung
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Li-Ya Qiao
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
30
|
Somers DL, Clemente RF. The neurophysiological basis of peripheral electrical nerve stimulation for the treatment of causalgia. PHYSICAL THERAPY REVIEWS 2013. [DOI: 10.1179/ptr.1996.1.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
31
|
Yan X, 严 喜, Weng HR, 翁 汉. Endogenous interleukin-1β in neuropathic rats enhances glutamate release from the primary afferents in the spinal dorsal horn through coupling with presynaptic N-methyl-D-aspartic acid receptors. J Biol Chem 2013; 288:30544-30557. [PMID: 24003233 DOI: 10.1074/jbc.m113.495465] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Excessive activation of glutamate receptors and overproduction of proinflammatory cytokines, including interleukin-1β (IL-1β) in the spinal dorsal horn, are key mechanisms underlying the development and maintenance of neuropathic pain. In this study, we investigated the mechanisms by which endogenous IL-1β alters glutamatergic synaptic transmission in the spinal dorsal horn in rats with neuropathic pain induced by ligation of the L5 spinal nerve. We demonstrated that endogenous IL-1β in neuropathic rats enhances glutamate release from the primary afferent terminals and non-NMDA glutamate receptor activities in postsynaptic neurons in the spinal dorsal horn. Myeloid differentiation primary response protein 88 (MyD88) is a mediator used by IL-1β to enhance non-NMDA glutamate receptor activities in postsynaptic neurons in the spinal dorsal horn. Presynaptic NMDA receptors are effector receptors used by the endogenous IL-1β to enhance glutamate release from the primary afferents in neuropathic rats. This is further supported by the fact that NMDA currents recorded from small neurons in the dorsal root ganglion of normal rats are potentiated by exogenous IL-1β. Furthermore, we provided evidence that functional coupling between IL-1β receptors and presynaptic NMDA receptors at the primary afferent terminals is mediated by the neutral sphingomyelinase/ceramide signaling pathway. Hence, functional coupling between IL-1β receptors and presynaptic NMDA receptors at the primary afferent terminals is a crucial mechanism leading to enhanced glutamate release and activation of non-NMDA receptors in the spinal dorsal horn neurons in neuropathic pain conditions. Interruption of such functional coupling could be an effective approach for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Xisheng Yan
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, Georgia 30602
| | - 喜胜 严
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, Georgia 30602
| | - Han-Rong Weng
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, Georgia 30602
| | - 汉荣 翁
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, Georgia 30602.
| |
Collapse
|
32
|
Evidence for glutamate as a neuroglial transmitter within sensory ganglia. PLoS One 2013; 8:e68312. [PMID: 23844184 PMCID: PMC3699553 DOI: 10.1371/journal.pone.0068312] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 05/28/2013] [Indexed: 11/19/2022] Open
Abstract
This study examines key elements of glutamatergic transmission within sensory ganglia of the rat. We show that the soma of primary sensory neurons release glutamate when depolarized. Using acute dissociated mixed neuronal/glia cultures of dorsal root ganglia (DRG) or trigeminal ganglia and a colorimetric assay, we show that when glutamate uptake by satellite glial cells (SGCs) is inhibited, KCl stimulation leads to simultaneous increase of glutamate in the culture medium. With calcium imaging we see that the soma of primary sensory neurons and SGCs respond to AMPA, NMDA, kainate and mGluR agonists, and selective antagonists block this response. Using whole cell patch-clamp technique, inward currents were recorded from small diameter (<30 µm) DRG neurons from intact DRGs (ex-vivo whole ganglion preparation) in response to local application of the above glutamate receptor agonists. Following a chronic constriction injury (CCI) of either the inferior orbital nerve or the sciatic nerve, glutamate expression increases in the trigeminal ganglia and DRG respectively. This increase occurs in neurons of all diameters and is present in the somata of neurons with injured axons as well as in somata of neighboring uninjured neurons. These data provides additional evidence that glutamate can be released within the sensory ganglion, and that the somata of primary sensory neurons as well as SGCs express functional glutamate receptors at their surface. These findings, together with our previous gene knockdown data, suggest that glutamatergic transmission within the ganglion could impact nociceptive threshold.
Collapse
|
33
|
Acute colitis induces neurokinin 1 receptor internalization in the rat lumbosacral spinal cord. PLoS One 2013; 8:e59234. [PMID: 23555638 PMCID: PMC3605455 DOI: 10.1371/journal.pone.0059234] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 02/13/2013] [Indexed: 11/19/2022] Open
Abstract
Substance P (SP) and its receptor, the neurokinin 1 receptor (NK1R), play important roles in transmitting and regulating somatosensory nociceptive information. However, their roles in visceral nociceptive transmission and regulation remain to be elucidated. In the previous study, moderate SP immunoreactive (SP-ir) terminals and NK1R-ir neurons were observed in the dorsal commissural nucleus (DCN) of the lumbosacral spinal cord. Thus we hypothesized that the SP-NK1R system is involved in visceral pain transmission and control within the DCN. The acute visceral pain behaviors, the colon histological changes and the temporal and spatial changes of NK1R-ir structures and Fos expression in the neurons of the DCN were observed in rats following lower colon instillation with 5% formalin. The formalin instillation induced significant acute colitis as revealed by the histological changes in the colon. NK1R internalization in the DCN was obvious at 8 min. It reached a peak (75.3%) at 30 min, began to decrease at 90 min (58.1%) and finally reached the minimum (19.7%) at 3 h after instillation. Meanwhile, formalin instillation induced a biphasic visceral pain response as well as a strong expression of Fos protein in the nuclei of neurons in the DCN. Finally, intrathecal treatment with the NK1R antagonist L732138 attenuated the NK1R internalization, Fos expression and visceral nociceptive responses. The present results suggest that the visceral nociceptive information arising from inflamed pelvic organs, such as the lower colon, might be mediated by the NK1R-ir neurons in the DCN of the lumbosacral spinal cord.
Collapse
|
34
|
Yan X, Jiang E, Gao M, Weng HR. Endogenous activation of presynaptic NMDA receptors enhances glutamate release from the primary afferents in the spinal dorsal horn in a rat model of neuropathic pain. J Physiol 2013; 591:2001-19. [PMID: 23359671 DOI: 10.1113/jphysiol.2012.250522] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Activation of N-methyl-D-aspartate (NMDA) receptors (NMDARs) is a crucial mechanism underlying the development and maintenance of pain. Traditionally, the role of NMDARs in the pathogenesis of pain is ascribed to their activation and signalling cascades in postsynaptic neurons. In this study, we determined if presynaptic NMDARs in the primary afferent central terminals play a role in synaptic plasticity of the spinal first sensory synapse in a rat model of neuropathic pain induced by spinal nerve ligation. Excitatory postsynaptic currents (EPSCs) were recorded from superficial dorsal horn neurons of spinal slices taken from young adult rats. We showed that increased glutamate release from the primary afferents contributed to the enhanced amplitudes of EPSCs evoked by input from the primary afferents in neuropathic rats. Endogenous activation of presynaptic NMDARs increased glutamate release from the primary afferents in neuropathic rats. Presynaptic NMDARs in neuropathic rats were mainly composed of NR2B receptors. The action of presynaptic NMDARs in neuropathic rats was enhanced by exogenous D-serine and/or NMDA and dependent on activation of protein kinase C. In contrast, glutamate release from the primary afferents in sham-operated rats was not regulated by presynaptic NMDARs. We demonstrated that the lack of NMDAR-mediated regulation of glutamate release in sham-operated rats was not attributable to low extracellular levels of the NMDAR agonist and/or coagonist (D-serine), but rather was due to the insufficient function and/or number of presynaptic NMDARs. This was supported by an increase of NR2B receptor protein expression in both the dorsal root ganglion and spinal dorsal horn ipsilateral to the injury site in neuropathic rats. Hence, suppression of the presynaptic NMDAR activity in the primary sensory afferents is an effective approach to attenuate the enhanced glutamatergic response in the spinal first sensory synapse induced by peripheral nerve injury, and presynaptic NMDARs might be a novel target for the development of analgesics.
Collapse
Affiliation(s)
- Xisheng Yan
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, Athens, GA 30602, USA
| | | | | | | |
Collapse
|
35
|
Reactive oxygen species are second messengers of neurokinin signaling in peripheral sensory neurons. Proc Natl Acad Sci U S A 2012; 109:E1578-86. [PMID: 22586118 DOI: 10.1073/pnas.1201544109] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Substance P (SP) is a prominent neuromodulator, which is produced and released by peripheral damage-sensing (nociceptive) neurons; these neurons also express SP receptors. However, the mechanisms of peripheral SP signaling are poorly understood. We report a signaling pathway of SP in nociceptive neurons: Acting predominantly through NK1 receptors and G(i/o) proteins, SP stimulates increased release of reactive oxygen species from the mitochondrial electron transport chain. Reactive oxygen species, functioning as second messengers, induce oxidative modification and augment M-type potassium channels, thereby suppressing excitability. This signaling cascade requires activation of phospholipase C but is largely uncoupled from the inositol 1,4,5-trisphosphate sensitive Ca(2+) stores. In rats SP causes sensitization of TRPV1 and produces thermal hyperalgesia. However, the lack of coupling between SP signaling and inositol 1,4,5-trisphosphate sensitive Ca(2+) stores, together with the augmenting effect on M channels, renders the SP pathway ineffective to excite nociceptors acutely and produce spontaneous pain. Our study describes a mechanism for neurokinin signaling in sensory neurons and provides evidence that spontaneous pain and hyperalgesia can have distinct underlying mechanisms within a single nociceptive neuron.
Collapse
|
36
|
Steagall RJ, Sipe AL, Williams CA, Joyner WL, Singh K. Substance P release in response to cardiac ischemia from rat thoracic spinal dorsal horn is mediated by TRPV1. Neuroscience 2012; 214:106-19. [PMID: 22525132 DOI: 10.1016/j.neuroscience.2012.04.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/11/2012] [Accepted: 04/12/2012] [Indexed: 11/24/2022]
Abstract
Spinal cord stimulation (SCS) inhibits substance P (SP) release and decreases the expression of the transient receptor potential vanilloid 1 (TRPV1) in the spinal cord at thoracic 4 (T4) during cardiac ischemia in rat models (Ding et al., 2007). We hypothesized that activation of TRPV1 in the T4 spinal cord segment by intermittent occlusion of the left anterior descending coronary artery (CoAO) mediates spinal cord SP release. Experiments were conducted in urethane-anesthetized Sprague-Dawley male rats using SP antibody-coated microprobes to measure SP release at the central terminal endings of cardiac ischemic-sensitive afferent neurons (CISAN) in the spinal T4 dorsal horns. Vehicle, capsaicin (CAP; TRPV1 agonist) and capsazepine (CZP; TRPV1 antagonist) were injected into the left T4 prior to stimulation of CISAN by intermittent CoAO (with or without upper cervical SCS). CAP induced endogenous SP release from laminae I and II in the T4 spinal cord above baseline. Conversely, CZP injections significantly inhibited SP release from laminae I-VII in the T4 spinal cord segment below baseline. CZP also attenuated CoAO-induced SP release, while T4 injections of CZP with SCS completely restored SP release to basal levels during CoAO activation. CAP increased the number of c-Fos (a marker for CISAN activation) positive T4 dorsal horn neurons compared to sham-operated animals, while CZP (alone or during CoAO and SCS+CoAO) significantly reduced the number of c-Fos positive neurons. These results suggest that spinal release of the putative nociceptive transmitter SP occurs, at least in part, via a TRPV1 mechanism.
Collapse
Affiliation(s)
- R J Steagall
- Department of Physiology, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614-1708, United States.
| | | | | | | | | |
Collapse
|
37
|
Neural acupuncture unit: a new concept for interpreting effects and mechanisms of acupuncture. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:429412. [PMID: 22474503 PMCID: PMC3310280 DOI: 10.1155/2012/429412] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 11/30/2011] [Accepted: 12/01/2011] [Indexed: 12/12/2022]
Abstract
When an acupuncture needle is inserted into a designated point on the body and
mechanical or electrical stimulation is delivered, various neural and neuroactive
components are activated. The collection of the activated neural and neuroactive
components distributed in the skin, muscle, and connective tissues surrounding the
inserted needle is defined as a neural acupuncture unit (NAU). The traditionally defined
acupoints represent an anatomical landmark system that indicates local sites where NAUs
may contain relatively dense and concentrated neural and neuroactive components, upon
which acupuncture stimulation would elicit a more efficient therapeutic response. The
NAU-based local mechanisms of biochemical and biophysical reactions play an important
role in acupuncture-induced analgesia. Different properties of NAUs are associated with
different components of needling sensation. There exist several central pathways to
convey NAU-induced acupuncture signals, Electroacupuncture (EA) frequency-specific
neurochemical effects are related to different peripheral and central pathways transmitting
afferent signals from different frequency of NAU stimulation. More widespread and intense
neuroimaging responses of brain regions to acupuncture may be a consequence of more
efficient NAU stimulation modes. The introduction of the conception of NAU provides a
new theoretical approach to interpreting effects and mechanisms of acupuncture in
modern biomedical knowledge framework.
Collapse
|
38
|
Dmitrieva N, Rodríguez-Malaver AJ, Pérez J, Hernández L. Differential release of neurotransmitters from superficial and deep layers of the dorsal horn in response to acute noxious stimulation and inflammation of the rat paw. Eur J Pain 2012; 8:245-52. [PMID: 15109975 DOI: 10.1016/j.ejpain.2003.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2003] [Accepted: 09/01/2003] [Indexed: 11/29/2022]
Abstract
Experimental evidence suggests that release of neurotransmitters in response to acute noxious stimulation and inflammation can differ in superficial and deeper dorsal horn (DH) laminae. Using two different microdialysis probes, we studied changes in levels of glutamate, aspartate, arginine and GABA in dialysates collected from the surface of the spinal cord and within the DH induced by pinching the paw or paw inflammation. In penthotal anaesthetized rats, a flexible microdialysis probe was placed on the dorsal surface of the L4-L5 or L6-S2 spinal segments. In other rats, a rigid microdialysis probe was implanted within the DH of the same segments. Samples were collected every minute before, during and after pinching the hind paw (acute pain), and every half an hour after injecting either carrageenan or saline into the same paw (inflammation-induced pain). Amino acids were measured by capillary zone electrophoresis with laser-induced fluorescence detection (CZE-LIFD). Pinching the paw induced a significant but short lasting increase in extracellular glutamate and aspartate in dialysates from the surface of the DH. Carrageenan, but not saline, injected into the paw significantly increased concentrations of glutamate, aspartate and arginine both on the surface and within the DH of L4-L5 and also within the DH of the L6-S2 segments. The GABA level was significantly increased following carrageenan only within the DH. The maximum increase on the surface was detected 60-120 min after the onset of inflammation whereas the response within the DH reached a maximum between 150 and 180 min after carrageenan. These results indicate that unlike acute mechanical noxious stimulation which enhances amino acid neurotransmitters in surface dialysate, inflammation induced neurotransmitter release in all layers of the DH suggesting sensitization of the DH.
Collapse
Affiliation(s)
- Natalia Dmitrieva
- Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA.
| | | | | | | |
Collapse
|
39
|
Brumovsky PR, Robinson DR, La JH, Seroogy KB, Lundgren KH, Albers KM, Kiyatkin ME, Seal RP, Edwards RH, Watanabe M, Hökfelt T, Gebhart GF. Expression of vesicular glutamate transporters type 1 and 2 in sensory and autonomic neurons innervating the mouse colorectum. J Comp Neurol 2012; 519:3346-66. [PMID: 21800314 DOI: 10.1002/cne.22730] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) have been extensively studied in various neuronal systems, but their expression in visceral sensory and autonomic neurons remains to be analyzed in detail. Here we studied VGLUTs type 1 and 2 (VGLUT(1) and VGLUT(2) , respectively) in neurons innervating the mouse colorectum. Lumbosacral and thoracolumbar dorsal root ganglion (DRG), lumbar sympathetic chain (LSC), and major pelvic ganglion (MPG) neurons innervating the colorectum of BALB/C mice were retrogradely traced with Fast Blue, dissected, and processed for immunohistochemistry. Tissue from additional naïve mice was included. Previously characterized antibodies against VGLUT(1) , VGLUT(2) , and calcitonin gene-related peptide (CGRP) were used. Riboprobe in situ hybridization, using probes against VGLUT(1) and VGLUT(2) , was also performed. Most colorectal DRG neurons expressed VGLUT(2) and often colocalized with CGRP. A smaller percentage of neurons expressed VGLUT(1) . VGLUT(2) -immunoreactive (IR) neurons in the MPG were rare. Abundant VGLUT(2) -IR nerves were detected in all layers of the colorectum; VGLUT(1) -IR nerves were sparse. A subpopulation of myenteric plexus neurons expressed VGLUT2 protein and mRNA, but VGLUT1 mRNA was undetectable. In conclusion, we show 1) that most colorectal DRG neurons express VGLUT(2) , and to a lesser extent, VGLUT(1) ; 2) abundance of VGLUT2-IR fibers innervating colorectum; and 3) a subpopulation of myenteric plexus neurons expressing VGLUT(2). Altogether, our data suggests a role for VGLUT(2) in colorectal glutamatergic neurotransmission, potentially influencing colorectal sensitivity and motility.
Collapse
Affiliation(s)
- Pablo R Brumovsky
- Pittsburgh Center for Pain Research, Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Clarke JN, Anderson RL, Haberberger RV, Gibbins IL. Non-peptidergic small diameter primary afferents expressing VGluT2 project to lamina I of mouse spinal dorsal horn. Mol Pain 2011; 7:95. [PMID: 22152428 PMCID: PMC3264520 DOI: 10.1186/1744-8069-7-95] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 12/08/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Unmyelinated primary afferent nociceptors are commonly classified into two main functional types: those expressing neuropeptides, and non-peptidergic fibers that bind the lectin IB4. However, many small diameter primary afferent neurons neither contain any known neuropeptides nor bind IB4. Most express high levels of vesicular glutamate transporter 2 (VGluT2) and are assumed to be glutamatergic nociceptors but their terminations within the spinal cord are unknown. We used in vitro anterograde axonal tracing with Neurobiotin to identify the central projections of these putative glutamatergic nociceptors. We also quantitatively characterised the spatial arrangement of these terminals with respect to those that expressed the neuropeptide, calcitonin gene-related peptide (CGRP). RESULTS Neurobiotin-labeled VGluT2-immunoreactive (IR) terminals were restricted to lamina I, with a medial-to-lateral distribution similar to CGRP-IR terminals. Most VGluT2-IR terminals in lateral lamina I were not labeled by Neurobiotin implying that they arose mainly from central neurons. 38 ± 4% of Neurobiotin-labeled VGluT2-IR terminals contained CGRP-IR. Conversely, only 17 ± 4% of Neurobiotin-labeled CGRP-IR terminals expressed detectable VGluT2-IR. Neurobiotin-labeled VGluT2-IR or CGRP-IR terminals often aggregated into small clusters or microdomains partially surrounding intrinsic lamina I neurons. CONCLUSIONS The central terminals of primary afferents which express high levels of VGluT2-IR but not CGRP-IR terminate mainly in lamina I. The spatial arrangement of VGluT2-IR and CGRP-IR terminals suggest that lamina I neurons receive convergent inputs from presumptive nociceptors that are primarily glutamatergic or peptidergic. This reveals a previously unrecognized level of organization in lamina I consistent with the presence of multiple nociceptive processing pathways.
Collapse
Affiliation(s)
- Jennifer N Clarke
- Anatomy and Histology, and Centre for Neuroscience, Flinders University, GPO Box 2100, Adelaide, SA, 5001, Australia
| | | | | | | |
Collapse
|
41
|
Carozzi VA, Zoia CP, Maggioni D, Verga E, Marmiroli P, Ferrarese C, Cavaletti G. Expression, distribution and glutamate uptake activity of high affinity-excitatory aminoacid transporters in in vitro cultures of embryonic rat dorsal root ganglia. Neuroscience 2011; 192:275-84. [PMID: 21726604 DOI: 10.1016/j.neuroscience.2011.05.065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 05/02/2011] [Accepted: 05/25/2011] [Indexed: 12/13/2022]
Abstract
Glutamate is the major mediator of excitatory signalling in the mammalian central nervous system, but it has recently been shown to play a role in the transduction of sensory input at the periphery and in peripheral neuropathies. New advances in research have demonstrated that rat peripheral sensory terminals and dorsal root ganglia (DRG) express molecules involved in glutamate signalling, including high-affinity membrane-bound glutamate transporters (GLAST [glutamate aspartate transporter], GLT1 [glutamate transporter 1], EAAC1 [excitatory aminoacid transporter 1]) and that alterations in their expression and/or functionality can be implicated in several models of peripheral neuropathy, neuropathic pain and hyperalgesia. Here we describe, through immunoblotting, immunofluorescence assays and β-counter analysis of [(3)H] l-glutamate uptake, the expression, distribution and activity of the glutamate transporters in in vitro cultures of embryonic dorsal root ganglia sensory neurons, sensory neurons+satellite cells and satellite cells. In this work we demonstrated that glutamate transporters are expressed in all cultures with a peculiar pattern of distribution. Even if GLAST is strongly detected in satellite cells, it is slightly expressed also in sensory neurons. GLT1 immunostaining is very weak in DRG neurons, but it was evident in the satellite cells. Finally, EAAC1 is localized in the soma and in the neuritis of sensory neurons, while it is not detectable in satellite cells. Moreover, all the cell cultures showed a strong sodium-energy-dependent glutamate uptake activity and it is more marked in neurons alone or in co-culture with satellite cells compared to satellite cells alone. Finally, we show that the complete or partial pharmacological inhibition of glutamate transporters virtually completely or partially abolish glutamate uptake in all cell culture. These results, that demonstrate that functionally active glutamate transporters can be studied in dorsal root ganglia cell cultures, provide further evidence for a role of glutamatergic transport in the peripheral nervous system and will be useful for testing whether any changes occur in in vitro models of peripheral nervous system damage.
Collapse
Affiliation(s)
- V A Carozzi
- Department of Neuroscience and Biomedical Technologies, University of Milano-Bicocca, Via Cadore 48, 20900 Monza (MB), Italy.
| | | | | | | | | | | | | |
Collapse
|
42
|
Quintero L, Cardenas R, Suarez-Roca H. Stress-induced hyperalgesia is associated with a reduced and delayed GABA inhibitory control that enhances post-synaptic NMDA receptor activation in the spinal cord. Pain 2011; 152:1909-1922. [PMID: 21636214 DOI: 10.1016/j.pain.2011.04.017] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 04/10/2011] [Accepted: 04/11/2011] [Indexed: 12/30/2022]
Abstract
GABA and glutamate are both affected by stress and are involved in nociception. Thus, we determined whether stress-induced enhancement of inflammatory hyperalgesia is mediated by an imbalance between glutamate and GABA neurotransmission. Male rats were subjected daily to 10 to 20 minutes per day of either forced swimming (FS) or sham swimming for 3 consecutive days; nonconditioned rats served as controls. Some rats were treated i.p. with ketamine (5 mg/kg), diazepam (2 mg/kg), flumazenil (0.1 mg/kg), or vehicle (0.9% NaCl), 30 to 60 minutes before each conditioning session or nociception assessment. Pain behavior, spinal nociceptive neuronal activation and GABA and glutamate release were respectively evaluated by the formalin test, the expression of c-Fos and in vivo microdialysis of superficial laminae of the lumbar spinal cord, 48 hours after the last conditioning session. Nitric oxide metabolites (NO(x)) were determined as markers of post-synaptic NMDA receptor activation. FS stress enhanced formalin-induced hyperalgesia, increased pain-elicited c-Fos expression, decreased basal and delayed pain-induced GABA release, and increased basal and induced glutamate release. Hyperalgesia and c-Fos overexpression were blocked only by prestress treatment with diazepam and post-stress treatment with ketamine, whereas changes in GABA and glutamate release were reversed by prestress treatment with diazepam. Diazepam effects were blocked by flumazenil. NO(x) increased in lumbar spinal cord of FS rats by a mechanism antagonized by ketamine. Thus, stress-induced hyperalgesia is initiated by a decreased and delayed GABA release and GABA-A receptor activation, whereas it is maintained by increased glutamate release and NMDA glutamate receptor activation at the spinal level.
Collapse
Affiliation(s)
- Luis Quintero
- Section of Neuropharmacology and Neuroscience, Instituto de Investigaciones Clínicas, Facultad de Medicina, University of Zulia, Maracaibo, Venezuela
| | | | | |
Collapse
|
43
|
A sensory subpopulation depends on vesicular glutamate transporter 2 for mechanical pain, and together with substance P, inflammatory pain. Proc Natl Acad Sci U S A 2011; 108:5789-94. [PMID: 21415372 DOI: 10.1073/pnas.1013602108] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ablating or functionally compromising sets of sensory neurons has provided important insights into peripheral modality-specific wiring in the somatosensory system. Inflammatory hyperalgesia, cold pain, and noxious mechanosensation have all been shown to depend upon Na(v)1.8-positive sensory neurons. The release of fast-acting neurotransmitters, such as glutamate, and more slowly released neuropeptides, such as substance P (SP), contribute to the diversified responses to external stimuli. Here we show that deleting Vglut2 in Na(v)1.8(Cre)-positive neurons compromised mechanical pain and NGF-induced thermal hyperalgesia, whereas tactile-evoked sensation, thermal, formalin-evoked, and chronic neuropathic pain were normal. However, when Vglut2(f/f);Na(v)1.8(Cre) mice were injected with a SP antagonist before the formalin test, the second phase pain response was nearly completely abolished, whereas in control mice, the pain response was unaffected. Our results suggest that VGLUT2-dependent signaling originating from Na(v)1.8-positive neurons is a principal sensing mechanism for mechanical pain and, together with SP, inflammatory pain. These data define sets of primary afferents associated with specific modalities and provide useful genetic tools with which to analyze the pathways that are activated by functionally distinct neuronal populations and transmitters.
Collapse
|
44
|
Kaufman MP, Forster HV. Reflexes Controlling Circulatory, Ventilatory and Airway Responses to Exercise. Compr Physiol 2011. [DOI: 10.1002/cphy.cp120110] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Wang Z, Ma W, Chabot JG, Quirion R. Morphological evidence for the involvement of microglial p38 activation in CGRP-associated development of morphine antinociceptive tolerance. Peptides 2010; 31:2179-84. [PMID: 20804799 DOI: 10.1016/j.peptides.2010.08.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 08/19/2010] [Accepted: 08/19/2010] [Indexed: 01/18/2023]
Abstract
Calcitonin gene-related peptide (CGRP) exerts an effect on the development of morphine antinociceptive tolerance, which may in part involve the activation of p38 kinase. In the present study, we investigated the temporal expression and spatial distribution of p38 phosphorylation as well as their possible regulation by CGRP receptor signaling following chronic morphine treatment. A 7-day intrathecal treatment with morphine (15 μg/day) produced tolerance to its analgesic effects as well as a rightward shift in the dose-response curve to its antinociceptive effects. This treatment time-dependently increased p38 phosphorylation in the spinal dorsal horn, as shown by phosphorylated p38-immunoreactive (p-p38-ir) cell counts. The increased phosphorylation occurred first in superficial layers of the spinal dorsal horn and then extended to deeper laminae. Interestingly, accompanying the development of morphine tolerance, p-p38-ir cells, identified as microglia, displayed hypertrophy and increased number of branched processes, suggesting their activation. These various behavioral and morphological changes were blocked by the intrathecal treatment with BIBN4096BS, a non-peptide CGRP receptor antagonist. These data provide additional morphological evidence in support of a role for CGRP in the development of morphine antinociceptive tolerance, possibly by regulating the expression and distribution of p38 phosphorylation in microglia.
Collapse
Affiliation(s)
- Zhiyong Wang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, 6875 LaSalle Boulevard, Montréal, Québec, Canada H4H 1R3
| | | | | | | |
Collapse
|
46
|
Allen CE, Worsley MA, King AE, Boissonade FM. Fos expression induced by activation of NMDA and neurokinin-1 receptors in the trigeminal subnucleus caudalis in vitro: role of protein kinases. Brain Res 2010; 1368:19-27. [PMID: 20977893 DOI: 10.1016/j.brainres.2010.10.072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 10/17/2010] [Accepted: 10/19/2010] [Indexed: 11/16/2022]
Abstract
Activity-induced neuronal plasticity is partly facilitated by the expression of the immediate-early gene c-fos and the resulting transcription factor Fos. Expression of Fos is associated with nociceptive afferent activation, but a detailed stimulation-transcription pathway for Fos expression has not yet been determined in the trigeminal system. This study utilized a novel in vitro model to determine whether Fos expression can be induced in trigeminal subnucleus caudalis by NMDA or neurokinin-1 receptor activation, and whether inhibition of intracellular kinases has any effect on Fos expression induced by activation of these receptors. Brainstems of male Wistar rats were excised and maintained in artificial cerebrospinal fluid at 37°C. NMDA or the specific neurokinin-1 receptor agonist [Sar(9),Met(O(2))(11)]-SP was applied. These agonists were subsequently tested in the presence of the protein kinase A inhibitor Rp-cAMP or protein kinase C inhibitor chelerythrine chloride. In all experiments the sodium channel blocker tetrodotoxin was used to prevent indirect neuronal activation. Brainstems were processed immunocytochemically for Fos expression, and positive cells were counted in the trigeminal subnucleus caudalis. NMDA and [Sar(9),Met(O(2))(11)]-SP significantly increased Fos expression, but these increases could be prevented by chelerythrine chloride. Rp-cAMP had no effect on Fos induced by NMDA but caused a significant reduction in Fos induced by [Sar(9),Met(O(2))(11)]-SP. These data demonstrate that in trigeminal subnucleus caudalis activation of either NK1 or NMDA receptors alone induces Fos expression; protein kinases A and C are involved in NK1R-induced Fos while protein kinase A is not required for NMDA receptor-induced Fos.
Collapse
Affiliation(s)
- Ceri E Allen
- Department of Oral and Maxillofacial Medicine and Surgery, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
47
|
Hegarty DM, Tonsfeldt K, Hermes SM, Helfand H, Aicher SA. Differential localization of vesicular glutamate transporters and peptides in corneal afferents to trigeminal nucleus caudalis. J Comp Neurol 2010; 518:3557-69. [PMID: 20593358 DOI: 10.1002/cne.22414] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Trigeminal afferents convey nociceptive information from the corneal surface of the eye to the trigeminal subnucleus caudalis (Vc). Trigeminal afferents, like other nociceptors, are thought to use glutamate and neuropeptides as neurotransmitters. The current studies examined whether corneal afferents contain both neuropeptides and vesicular glutamate transporters. Corneal afferents to the Vc were identified by using cholera toxin B (CTb). Corneal afferents project in two clusters to the rostral and caudal borders of the Vc, regions that contain functionally distinct nociceptive neurons. Thus, corneal afferents projecting to these two regions were examined separately. Dual immunocytochemical studies combined CTb with either calcitonin gene-related peptide (CGRP), substance P (SP), vesicular glutamate transporter 1 (VGluT1), or VGluT2. Corneal afferents were more likely to contain CGRP than SP, and corneal afferents projecting to the rostral region were more likely to contain CGRP than afferents projecting caudally. Overall, corneal afferents were equally likely to contain VGluT1 or VGluT2. Together, 61% of corneal afferents contained either VGluT1 or VGluT2, suggesting that some afferents lack a VGluT. Caudal corneal afferents were more likely to contain VGluT2 than VGluT1, whereas rostral corneal afferents were more likely to contain VGluT1 than VGluT2. Triple-labeling studies combining CTb, CGRP, and VGluT2 showed that very few corneal afferents contain both CGRP and VGluT2, caudally (1%) and rostrally (2%). These results suggest that most corneal afferents contain a peptide or a VGluT, but rarely both. Our results are consistent with a growing literature suggesting that glutamatergic and peptidergic sensory afferents may be distinct populations.
Collapse
Affiliation(s)
- Deborah M Hegarty
- Department of Physiology and Pharmacology, Oregon Health & Science University, Beaverton, Oregon 97006, USA
| | | | | | | | | |
Collapse
|
48
|
Heinzlmann A, Tóth ZE, Köves K. Secretin mRNA in the subdivision of primary sensory neurons in the trigeminal ganglion of rats. J Mol Neurosci 2010; 43:101-8. [PMID: 20582488 DOI: 10.1007/s12031-010-9395-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 05/19/2010] [Indexed: 11/26/2022]
Abstract
The primary sensory neurons use glutamate as a major neurotransmitter. Several neuropeptides are also found in these neurons. In our laboratory we demonstrated secretin-like immunoreactivity in primary sensory neurons of several species including human, rat and cat. In the present experiment utilizing in situ hybridization, we have demonstrated for the first time that secretin is not only immunostained but is also expressed in the primary sensory neurons of the trigeminal ganglion of male rats. In intact rats, secretin mRNA was not observed; we had to use intracerebroventricular colchicine administration to induce the expression of secretin. Secretin was expressed in about 5% of the cells in all the three subdivisions of the trigeminal ganglion. The secretin-synthetizing cells were large and medium sized, and their mean diameter was about 50 μm. When we compared the percentage and the size of secretin to that of calcitonin gene-related peptide (CGRP), substance-P (SP) and vasoactive intestinal polypeptide (VIP) cells, it was found that CGRP, SP and VIP are present in about 15-20% of the cells and their mean diameter is about 20-25 μm. The morphometric data indicate that secretin is present in a subdivision of neurons that is different from the subdivision of the CGRP, SP and VIP cells. It is suggested that secretin may modulate the function of the primary neurotransmitter.
Collapse
Affiliation(s)
- Andrea Heinzlmann
- Department of Human Morphology and Developmental Biology, Semmelweis University, Tűzoltó u. 58, Budapest 1094, Hungary.
| | | | | |
Collapse
|
49
|
Wu SX, Wang W, Li H, Wang YY, Feng YP, Li YQ. The synaptic connectivity that underlies the noxious transmission and modulation within the superficial dorsal horn of the spinal cord. Prog Neurobiol 2010; 91:38-54. [DOI: 10.1016/j.pneurobio.2010.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 12/10/2009] [Accepted: 01/14/2010] [Indexed: 01/27/2023]
|
50
|
Contralateral electroacupuncture pretreatment suppresses carrageenan-induced inflammatory pain via the opioid-mu receptor. Rheumatol Int 2010; 31:725-30. [PMID: 20130880 DOI: 10.1007/s00296-010-1364-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Accepted: 09/13/2009] [Indexed: 10/19/2022]
Abstract
Acupuncture has been used to treat various clinical diseases in Eastern medicine. To investigate the analgesic effect of electroacupuncture (EA) pretreatment on carrageenan-induced inflammatory pain, we studied on the effect of EA parameters on an animal model of acute arthritic pain. Pretreatment with 1 mA, 10 Hz EA prior to carrageenan injection under halothane anesthesia suppressed carrageenan-induced pain. Interestingly, EA stimulation of the 'Zu-San-Li' (ST36) acupuncture point (1 mA, 10 Hz) contralateral to the site of the carrageenan injection in the rat synovial cavity produced significantly greater improvement of the weight-bearing force compared with EA stimulation of the 'San-Yin-Jiao' acupuncture point. To determine how ST36 EA treatment suppresses carrageenan-induced inflammatory pain, we examined the effect of a mu opioid receptor antagonist on ST36 EA-induced analgesia. The selective antagonist of the mu opioid receptor (OR) significantly suppressed contralateral ST36 EA-induced analgesia against carrageenan-induced inflammation. These results suggested that the analgesic effect mediated by the mu OR during low-frequency contralateral EA pretreatment has an anti-nociceptive action against inflammatory pain and that it may provide a potential strategy to treat inflammatory arthritic pain.
Collapse
|