1
|
David ET, Yousuf MS, Mei HR, Jain A, Krishnagiri S, Elahi H, Venkatesan R, Srikanth KD, Dussor G, Dalva MB, Price TJ. ephrin-B2 promotes nociceptive plasticity and hyperalgesic priming through EphB2-MNK-eIF4E signaling in both mice and humans. Pharmacol Res 2024; 206:107284. [PMID: 38925462 DOI: 10.1016/j.phrs.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 06/28/2024]
Abstract
Ephrin-B-EphB signaling can promote pain through ligand-receptor interactions between peripheral cells, like immune cells expressing ephrin-Bs, and EphB receptors expressed by DRG neurons. Previous studies have shown increased ephrin-B2 expression in peripheral tissues like synovium of rheumatoid and osteoarthritis patients, indicating the clinical significance of this signaling. The primary goal of this study was to understand how ephrin-B2 acts on mouse and human DRG neurons, which express EphB receptors, to promote pain and nociceptor plasticity. We hypothesized that ephrin-B2 would promote nociceptor plasticity and hyperalgesic priming through MNK-eIF4E signaling, a critical mechanism for nociceptive plasticity induced by growth factors, cytokines and nerve injury. Both male and female mice developed dose-dependent mechanical hypersensitivity in response to ephrin-B2, and both sexes showed hyperalgesic priming when challenged with PGE2 injection either to the paw or the cranial dura. Acute nociceptive behaviors and hyperalgesic priming were blocked in mice lacking MNK1 (Mknk1 knockout mice) and by eFT508, a specific MNK inhibitor. Sensory neuron-specific knockout of EphB2 using Pirt-Cre demonstrated that ephrin-B2 actions require this receptor. In Ca2+-imaging experiments on cultured DRG neurons, ephrin-B2 treatment enhanced Ca2+ transients in response to PGE2 and these effects were absent in DRG neurons from MNK1-/- and EphB2-PirtCre mice. In experiments on human DRG neurons, ephrin-B2 increased eIF4E phosphorylation and enhanced Ca2+ responses to PGE2 treatment, both blocked by eFT508. We conclude that ephrin-B2 acts directly on mouse and human sensory neurons to induce nociceptor plasticity via MNK-eIF4E signaling, offering new insight into how ephrin-B signaling promotes pain.
Collapse
Affiliation(s)
- Eric T David
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Muhammad Saad Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Hao-Ruei Mei
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Ashita Jain
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Sharada Krishnagiri
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Hajira Elahi
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Rupali Venkatesan
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Kolluru D Srikanth
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA
| | - Matthew B Dalva
- Jefferson Synaptic Biology Center, Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA; Tulane Brain Institute, Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70124, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences, Department of Neuroscience, Center for Advanced Pain Studies, USA.
| |
Collapse
|
2
|
Muir WM, Lo CL, Bell RL, Zhou FC. Multi-animal-model study reveals mutations in neural plasticity and nociception genes linked to excessive alcohol drinking. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1478-1493. [PMID: 37336636 PMCID: PMC10728351 DOI: 10.1111/acer.15131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND The basis for familial alcohol use disorder (AUD) remains an enigma due to various biological and societal confounds. The present study used three of the most adopted and documented rat models, combining the alcohol-preferring/non-alcohol-preferring (P/NP) lines and high alcohol-drinking/low alcohol-drinking (HAD/LAD) replicated lines, of AUD as examined through the lens of whole genomic analyses. METHODS We used complete genome sequencing of the P/NP lines and previously published sequences of the HAD/LAD replicates to enhance the discovery of variants associated with AUD and to remove confounding with genetic background and random genetic drift. Specifically, we used high-order statistical methods to search for genetic variants whose frequency changes in whole sets of gene ontologies corresponded with phenotypic changes in the direction of selection, that is, ethanol-drinking preference. RESULTS Our first finding was that in addition to variants causing translational changes, the principal genetic changes associated with drinking predisposition were silent mutations and mutations in the 3' untranslated regions (3'UTR) of genes. Neither of these types of mutations alters the amino acid sequence of the translated protein but they influence both the rate and conformation of gene transcription, including its stability and posttranslational events that alter gene efficacy. This finding argues for refocusing human genomic studies on changes in gene efficacy. Among the key ontologies identified were the central genes associated with the Na+ voltage-gated channels of neurons and glia (including the Scn1a, Scn2a, Scn2b, Scn3a, Scn7a, and Scn9a subtypes) and excitatory glutamatergic secretion (including Grm2 and Myo6), both of which are essential in neuroplasticity. In addition, we identified "Nociception or Sensory Perception of Pain," which contained variants in nociception (Arrb1, Ccl3, Ephb1) and enlist sodium (Scn1a, Scn2a, Scn2b, Scn3a, Scn7a), pain activation (Scn9a), and potassium channel (Kcna1) genes. CONCLUSION The multi-model analyses used herein reduced the confounding effects of random drift and the "founders" genetic background. The most differentiated bidirectionally selected genes across all three animal models were Scn9a, Scn1a, and Kcna, all of which are annotated in the nociception ontology. The complexity of neuroplasticity and nociception adds strength to the hypothesis that neuroplasticity and pain (physical or psychological) are prominent phenotypes genetically linked to the development of AUD.
Collapse
Affiliation(s)
- William M. Muir
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Chiao-Ling Lo
- Indiana Alcohol Research Center, Indiana University School of Medicine
| | - Richard L. Bell
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Stark Neuroscience Research Institute, Indianapolis, Indiana, USA
| | - Feng C. Zhou
- Indiana Alcohol Research Center, Indiana University School of Medicine
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Stark Neuroscience Research Institute, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Washburn HR, Chander P, Srikanth KD, Dalva MB. Transsynaptic Signaling of Ephs in Synaptic Development, Plasticity, and Disease. Neuroscience 2023; 508:137-152. [PMID: 36460219 DOI: 10.1016/j.neuroscience.2022.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
Synapse formation between neurons is critical for proper circuit and brain function. Prior to activity-dependent refinement of connections between neurons, activity-independent cues regulate the contact and recognition of potential synaptic partners. Formation of a synapse results in molecular recognition events that initiate the process of synaptogenesis. Synaptogenesis requires contact between axon and dendrite, selection of correct and rejection of incorrect partners, and recruitment of appropriate pre- and postsynaptic proteins needed for the establishment of functional synaptic contact. Key regulators of these events are families of transsynaptic proteins, where one protein is found on the presynaptic neuron and the other is found on the postsynaptic neuron. Of these families, the EphBs and ephrin-Bs are required during each phase of synaptic development from target selection, recruitment of synaptic proteins, and formation of spines to regulation of synaptic plasticity at glutamatergic spine synapses in the mature brain. These roles also place EphBs and ephrin-Bs as important regulators of human neurological diseases. This review will focus on the role of EphBs and ephrin-Bs at synapses.
Collapse
Affiliation(s)
- Halley R Washburn
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Praveen Chander
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Kolluru D Srikanth
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Matthew B Dalva
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA.
| |
Collapse
|
4
|
Gamble MC, Williams BR, Singh N, Posa L, Freyberg Z, Logan RW, Puig S. Mu-opioid receptor and receptor tyrosine kinase crosstalk: Implications in mechanisms of opioid tolerance, reduced analgesia to neuropathic pain, dependence, and reward. Front Syst Neurosci 2022; 16:1059089. [PMID: 36532632 PMCID: PMC9751598 DOI: 10.3389/fnsys.2022.1059089] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/31/2022] [Indexed: 07/30/2023] Open
Abstract
Despite the prevalence of opioid misuse, opioids remain the frontline treatment regimen for severe pain. However, opioid safety is hampered by side-effects such as analgesic tolerance, reduced analgesia to neuropathic pain, physical dependence, or reward. These side effects promote development of opioid use disorders and ultimately cause overdose deaths due to opioid-induced respiratory depression. The intertwined nature of signaling via μ-opioid receptors (MOR), the primary target of prescription opioids, with signaling pathways responsible for opioid side-effects presents important challenges. Therefore, a critical objective is to uncouple cellular and molecular mechanisms that selectively modulate analgesia from those that mediate side-effects. One such mechanism could be the transactivation of receptor tyrosine kinases (RTKs) via MOR. Notably, MOR-mediated side-effects can be uncoupled from analgesia signaling via targeting RTK family receptors, highlighting physiological relevance of MOR-RTKs crosstalk. This review focuses on the current state of knowledge surrounding the basic pharmacology of RTKs and bidirectional regulation of MOR signaling, as well as how MOR-RTK signaling may modulate undesirable effects of chronic opioid use, including opioid analgesic tolerance, reduced analgesia to neuropathic pain, physical dependence, and reward. Further research is needed to better understand RTK-MOR transactivation signaling pathways, and to determine if RTKs are a plausible therapeutic target for mitigating opioid side effects.
Collapse
Affiliation(s)
- Mackenzie C. Gamble
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Benjamin R. Williams
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Navsharan Singh
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Luca Posa
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
- Center for Systems Neuroscience, Boston University, Boston, MA, United States
| | - Stephanie Puig
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
5
|
Blockade of Erythropoietin-Producing Human Hepatocellular Carcinoma Receptor B1 in Spinal Dorsal Horn Alleviates Visceral Pain in Rats. Pain Res Manag 2021; 2021:7582494. [PMID: 33880135 PMCID: PMC8046573 DOI: 10.1155/2021/7582494] [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: 06/25/2020] [Revised: 02/19/2021] [Accepted: 03/10/2021] [Indexed: 11/24/2022]
Abstract
Objective This experiment was designed to determine whether erythropoietin-producing human hepatocellular carcinoma (Eph) receptors were involved in the development of visceral pain. Methods Adult male Sprague-Dawley rats were randomly divided into three groups receiving different treatments (n = 16 per group): intracolonic vehicle (control group), intracolonic 2, 4, 6-trinitrobenzene sulfonic acid (TNBS) (TNBS group), and intracolonic TNBS and intrathecal EphB1 receptor blocking reagent (TNBS + EphB2-Fc group). Visceral hyperalgesia was evaluated with quantification of visceral pain threshold induced by colorectal distention. The spinal expressions of EphB1 and ephrinB2 and levels of their phosphorylated forms (p-EphB1 and p-ephrinB2) were assessed by Western blotting and immunohistochemistry. Results The TNBS-treated rats developed significant visceral hyperalgesia. The spinal expressions of EphB1, p-EphB1, ephrinB2, and p-ephrinB2 were significantly increased in the TNBS group compared with the control group, but visceral hyperalgesia and elevation of spinal EphB1 and p-EphB1 expressions were evidently alleviated by intrathecal administration of EphB2-Fc in the TNBS + EphB2-Fc group. The number of EphB1- and p-EphB1-immunopositive cells, the average optical (AO) value of EphB1, and its phosphorylated form in the spinal dorsal horn were significantly increased in the TNBS group than in the control group, but they were obviously reduced by intrathecal administration of EphB2-Fc. There were no significant differences in the number of ephrinB2- and p-ephrinB2-immunopositive cells and the AO value of ephrinB2 and its phosphorylated form between the TNBS and TNBS + EphB2-Fc groups. Conclusion EphB1 receptors in the spinal dorsal horn play a pivotal role in the development of visceral pain and may be considered as a potential target for the treatment of visceral pain.
Collapse
|
6
|
Identification of tetracycline combinations as EphB1 tyrosine kinase inhibitors for treatment of neuropathic pain. Proc Natl Acad Sci U S A 2021; 118:2016265118. [PMID: 33627480 DOI: 10.1073/pnas.2016265118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Previous studies have demonstrated that the synaptic EphB1 receptor tyrosine kinase is a major mediator of neuropathic pain, suggesting that targeting the activity of this receptor might be a viable therapeutic option. Therefore, we set out to determine if any FDA-approved drugs can act as inhibitors of the EphB1 intracellular catalytic domain. An in silico screen was first used to identify a number of tetracycline antibiotics which demonstrated potential docking to the ATP-binding catalytic domain of EphB1. Kinase assays showed that demeclocycline, chlortetracycline, and minocycline inhibit EphB1 kinase activity at low micromolar concentrations. In addition, we cocrystallized chlortetracycline and EphB1 receptor, which confirmed its binding to the ATP-binding domain. Finally, in vivo administration of the three-tetracycline combination inhibited the phosphorylation of EphB1 in the brain, spinal cord, and dorsal root ganglion (DRG) and effectively blocked neuropathic pain in mice. These results indicate that demeclocycline, chlortetracycline, and minocycline can be repurposed for treatment of neuropathic pain and potentially for other indications that would benefit from inhibition of EphB1 receptor kinase activity.
Collapse
|
7
|
Theofanous SA, Florens MV, Appeltans I, Denadai Souza A, Wood JN, Wouters MM, Boeckxstaens GE. Ephrin-B2 signaling in the spinal cord as a player in post-inflammatory and stress-induced visceral hypersensitivity. Neurogastroenterol Motil 2020; 32:e13782. [PMID: 32004400 DOI: 10.1111/nmo.13782] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/07/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Ephrin-B2/EphB receptor signaling contributes to persistent pain states such as postinflammatory and neuropathic pain. Visceral hypersensitivity (VHS) is a major mechanism underlying abdominal pain in patients with irritable bowel syndrome (IBS) and inflammatory bowel diseases (IBD) in remission, but the underlying pathophysiology remains unclear. Here, we evaluated the spinal ephrin-B2/EphB pathway in VHS in 2 murine models of VHS, that is, postinflammatory TNBS colitis and maternal separation (MS). METHODS Wild-type (WT) mice and mice lacking ephrin-B2 in Nav 1.8 nociceptive neurons (cKO) were studied. VHS was induced by: 1. intracolonic instillation of TNBS or 2. water avoidance stress (WAS) in mice that underwent maternal separation (MS). VHS was assessed by quantifying the visceromotor response (VMRs) during colorectal distention. Colonic tissue and spinal cord were collected for histology, gene, and protein expression evaluation. KEY RESULTS In WT mice, but not cKO mice, TNBS induced VHS at day 14 after instillation, which returned to baseline perception from day 28 onwards. In MS WT mice, WAS induced VHS for up to 4 weeks. In cKO however, visceral pain perception returned to basal level by week 4. The development of VHS in WT mice was associated with significant upregulation of spinal ephrin-B2 and EphB1 mRNA expression or protein levels in the TNBS model and upregulation of spinal ephrin-B2 protein in the MS model. No changes were observed in cKO mice. VHS was not associated with persistent intestinal inflammation. CONCLUSIONS AND INFERENCES Overall, our data indicate that the ephrin-B2/EphB1 spinal signaling pathway is involved in VHS and may represent a novel therapeutic target.
Collapse
Affiliation(s)
| | - Morgane V Florens
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | | | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research (WIBR), University College London (UCL), London, UK
| | - Mira M Wouters
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Activation of EphB receptors contributes to primary sensory neuron excitability by facilitating Ca2+ influx directly or through Src kinase-mediated N-methyl-D-aspartate receptor phosphorylation. Pain 2020; 161:1584-1596. [DOI: 10.1097/j.pain.0000000000001855] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Goncalves MB, Moehlin J, Clarke E, Grist J, Hobbs C, Carr AM, Jack J, Mendoza-Parra MA, Corcoran JPT. RARβ Agonist Drug (C286) Demonstrates Efficacy in a Pre-clinical Neuropathic Pain Model Restoring Multiple Pathways via DNA Repair Mechanisms. iScience 2019; 20:554-566. [PMID: 31655065 PMCID: PMC6833472 DOI: 10.1016/j.isci.2019.09.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/06/2019] [Accepted: 09/12/2019] [Indexed: 01/05/2023] Open
Abstract
Neuropathic pain (NP) is associated with profound gene expression alterations within the nociceptive system. DNA mechanisms, such as epigenetic remodeling and repair pathways have been implicated in NP. Here we have used a rat model of peripheral nerve injury to study the effect of a recently developed RARβ agonist, C286, currently under clinical research, in NP. A 4-week treatment initiated 2 days after the injury normalized pain sensation. Genome-wide and pathway enrichment analysis showed that multiple mechanisms persistently altered in the spinal cord were restored to preinjury levels by the agonist. Concomitant upregulation of DNA repair proteins, ATM and BRCA1, the latter being required for C286-mediated pain modulation, suggests that early DNA repair may be important to prevent phenotypic epigenetic imprints in NP. Thus, C286 is a promising drug candidate for neuropathic pain and DNA repair mechanisms may be useful therapeutic targets to explore.
Collapse
Affiliation(s)
- Maria B Goncalves
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK.
| | - Julien Moehlin
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057 Evry, France
| | - Earl Clarke
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - John Grist
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Carl Hobbs
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Antony M Carr
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| | - Julian Jack
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Marco Antonio Mendoza-Parra
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057 Evry, France.
| | - Jonathan P T Corcoran
- The Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
10
|
Talebian A, Henkemeyer M. EphB2 receptor cell-autonomous forward signaling mediates auditory memory recall and learning-driven spinogenesis. Commun Biol 2019; 2:372. [PMID: 31633063 PMCID: PMC6789002 DOI: 10.1038/s42003-019-0625-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/16/2019] [Indexed: 01/09/2023] Open
Abstract
While ephrin-B ligands and EphB receptors are expressed to high levels in the learning centers of the brain, it remains largely unknown how their trans-synaptic interactions contribute to memory. We find that EphB2 forward signaling is needed for contextual and sound-evoked memory recall and that constitutive over-activation of the receptor's intracellular tyrosine kinase domain results in enhanced memory. Loss of EphB2 expression does not affect the number of neurons activated following encoding, although a reduction of neurons activated after the sound-cued retrieval test was detected in the auditory cortex and hippocampal CA1. Further, spine density and maturation was reduced in the auditory cortex of mutants especially in the neurons that were dual-activated during both encoding and retrieval. Our data demonstrates that trans-synaptic ephrin-B-EphB2 interactions and forward signaling facilitate neural activation and structural plasticity in learning-associated neurons involved in the generation of memories.
Collapse
Affiliation(s)
- Asghar Talebian
- Department of Neuroscience and Kent Waldrep Center for Basic Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Mark Henkemeyer
- Department of Neuroscience and Kent Waldrep Center for Basic Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| |
Collapse
|
11
|
Henderson NT, Dalva MB. EphBs and ephrin-Bs: Trans-synaptic organizers of synapse development and function. Mol Cell Neurosci 2018; 91:108-121. [PMID: 30031105 PMCID: PMC6159941 DOI: 10.1016/j.mcn.2018.07.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 12/31/2022] Open
Abstract
Synapses are specialized cell-cell junctions that underlie the function of neural circuits by mediating communication between neurons. Both the formation and function of synapses require tight coordination of signaling between pre- and post-synaptic neurons. Trans-synaptic organizing molecules are important mediators of such signaling. Here we discuss how the EphB and ephrin-B families of trans-synaptic organizing proteins direct synapse formation during early development and regulate synaptic function and plasticity at mature synapses. Finally, we highlight recent evidence linking the synaptic organizing role of EphBs and ephrin-Bs to diseases of maladaptive synaptic function and plasticity.
Collapse
Affiliation(s)
- Nathan T Henderson
- The Jefferson Synaptic Biology Center, Department of Neuroscience, The Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Jefferson Hospital for Neuroscience, Suite 463, 900 Walnut St., Philadelphia, PA 19107, United States
| | - Matthew B Dalva
- The Jefferson Synaptic Biology Center, Department of Neuroscience, The Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Jefferson Hospital for Neuroscience, Suite 463, 900 Walnut St., Philadelphia, PA 19107, United States.
| |
Collapse
|
12
|
Skupio U, Sikora M, Korostynski M, Wawrzczak-Bargiela A, Piechota M, Ficek J, Przewlocki R. Behavioral and transcriptional patterns of protracted opioid self-administration in mice. Addict Biol 2017; 22:1802-1816. [PMID: 27578564 DOI: 10.1111/adb.12449] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 12/31/2022]
Abstract
Chronic exposure to opioids induces adaptations in brain function that lead to the formation of the behavioral and physiological symptoms of drug dependence and addiction. Animal models commonly used to test these symptoms typically last less than two weeks, which is presumably too short to observe the alterations in the brain that accompany drug addiction. Here, we analyzed the phenotypic and molecular effects of nearly lifelong morphine or saccharin intake in C57BL/6J mice. We used multiple paradigms to evaluate the symptoms of compulsive drug intake: a progressive ratio schedule, intermittent access and a schedule involving a risk of punishment were programmed into an automated IntelliCage system. Gene expression profiles were evaluated in the striatum using whole-genome microarrays and further validated using quantitative polymerase chain reaction in the striatum and the prefrontal cortex. Mice voluntary self-administering morphine showed addiction-related behavioral pattern that included: higher motivation to work for a drug reward, increased reward seeking and increased craving. The analysis of molecular changes revealed a tolerance effect in the transcriptional response to morphine injection (20 mg/kg, ip), as well as some long-lasting alterations in gene expression profiles between the analyzed groups of animals. Interestingly, among the morphine-drinking animals, certain transcriptional profiles were found to be associated with alterations in behavior. In conclusion, our model represents a novel approach for investigating the behavioral and molecular mechanisms underlying opioid addiction. Prolonged morphine intake caused adaptive processes in the brain that manifested as altered behavior and transcriptional sensitivity to opioids.
Collapse
Affiliation(s)
- Urszula Skupio
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| | - Magdalena Sikora
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| | - Michal Korostynski
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| | | | - Marcin Piechota
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| | - Joanna Ficek
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| | - Ryszard Przewlocki
- Department of Molecular Neuropharmacology, Institute of Pharmacology; Polish Academy of Sciences; Poland
| |
Collapse
|
13
|
Hanamura K, Washburn HR, Sheffler-Collins SI, Xia NL, Henderson N, Tillu DV, Hassler S, Spellman DS, Zhang G, Neubert TA, Price TJ, Dalva MB. Extracellular phosphorylation of a receptor tyrosine kinase controls synaptic localization of NMDA receptors and regulates pathological pain. PLoS Biol 2017; 15:e2002457. [PMID: 28719605 PMCID: PMC5515392 DOI: 10.1371/journal.pbio.2002457] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 06/12/2017] [Indexed: 11/18/2022] Open
Abstract
Extracellular phosphorylation of proteins was suggested in the late 1800s when it was demonstrated that casein contains phosphate. More recently, extracellular kinases that phosphorylate extracellular serine, threonine, and tyrosine residues of numerous proteins have been identified. However, the functional significance of extracellular phosphorylation of specific residues in the nervous system is poorly understood. Here we show that synaptic accumulation of GluN2B-containing N-methyl-D-aspartate receptors (NMDARs) and pathological pain are controlled by ephrin-B-induced extracellular phosphorylation of a single tyrosine (p*Y504) in a highly conserved region of the fibronectin type III (FN3) domain of the receptor tyrosine kinase EphB2. Ligand-dependent Y504 phosphorylation modulates the EphB-NMDAR interaction in cortical and spinal cord neurons. Furthermore, Y504 phosphorylation enhances NMDAR localization and injury-induced pain behavior. By mediating inducible extracellular interactions that are capable of modulating animal behavior, extracellular tyrosine phosphorylation of EphBs may represent a previously unknown class of mechanism mediating protein interaction and function. The activity of proteins can be finely and reversibly tuned by post-translational modifications. The attachment of phosphate groups to tyrosine residues is one of such modifications. While the existence of extracellular phosphoproteins has been known, the functional significance of extracellular phosphorylation is poorly understood. Here we describe a single extracellular tyrosine whose inducible phosphorylation may represent an archetype for a new class of mechanism mediating protein—protein interaction and regulating protein function. We show that the interaction between EphB2—which occurs upon receptor activation by its ligand ephrin-B—and the N-methyl-D-aspartate receptor (NMDAR) depends on extracellular phosphorylation of EphB2. This interaction regulates the localization of the NMDA receptor to synaptic sites in neurons. In vivo, EphB2 is phosphorylated in response to injury, and the subsequent up-regulation of the interaction between EphB2 and NMDA receptors enhances injury-induced pain behavior and mechanical hypersensitivity in mice. Importantly, our study defines a specific extracellular phosphorylation event as a mechanism driving protein interaction and suggests that extracellular phosphorylation of proteins is an underappreciated mechanism contributing to the development and function of the nervous system and synapse.
Collapse
Affiliation(s)
- Kenji Hanamura
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi City, Gunma, Japan
| | - Halley R. Washburn
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
| | - Sean I. Sheffler-Collins
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
- Neuroscience Graduate Group, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Nan L. Xia
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
| | - Nathan Henderson
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
| | - Dipti V. Tillu
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, United States of America
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, United States of America
| | - Shayne Hassler
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, United States of America
| | - Daniel S. Spellman
- Department of Cell Biology and Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York, United States of America
| | - Guoan Zhang
- Department of Cell Biology and Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York, United States of America
| | - Thomas A. Neubert
- Department of Cell Biology and Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York, United States of America
| | - Theodore J. Price
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, Arizona, United States of America
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas, United States of America
| | - Matthew B. Dalva
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, Jefferson Hospital for Neuroscience, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Deng XT, Wu MZ, Xu N, Ma PC, Song XJ. Activation of ephrinB-EphB receptor signalling in rat spinal cord contributes to maintenance of diabetic neuropathic pain. Eur J Pain 2016; 21:278-288. [PMID: 27461472 DOI: 10.1002/ejp.922] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Diabetic neuropathic pain (DNP) is severe and intractable in clinic. The specific cellular and molecular mechanisms underlying DNP remain elusive and its treatment are limited. We investigated roles of EphB1 receptor in the development of DNP. METHODS Diabetic neuropathic pain was produced in male, adult, Sprague-Dawley rats by a single i.p. streptozotocin (STZ) or alloxan. Western blot analysis and immunohistochemistry were used to analyse expression of EphB1 receptor as well as the activation of the glial cells and the pro-inflammatory cytokines in the spinal cord. DNP manifested as mechanical allodynia, which was determined by measuring incidence of foot withdrawal in response to mechanical indentation of the hind paw by an electro von Frey filament. RESULTS Diabetic neuropathic pain and high blood glucose were exhibited simultaneously in around 70% of animals that received i.p. STZ or alloxan. Phosphorylation of EphB1, activation of the astrocytes and microglial cells, and level of tumour necrosis factor (TNF)-α and interleukin (IL)-1β in the spinal cord were significantly increased in rats with DNP. Spinal blocking EphB1 receptor activation in the late phase after STZ injection significantly suppressed the established mechanical allodynia as well as activation of the astrocytes and microglial cells and activity of TNF-α and IL-1β. However, spinal treatment of EphB1-Fc in the early phase after STZ injection did not prevent the induction of DNP. CONCLUSIONS EphB1 receptor activation in the spinal cord is critical to the maintenance, but not induction of diabetic pain. EphB1 receptor may be a potential target for relieving the established diabetic pain. SIGNIFICANCE Activation of EphB1 receptor in the spinal cord is critical to maintaining the established diabetic neuropathic pain, but not to diabetic pain induction. Spinal blocking EphB1 receptor activation suppresses ongoing diabetic neuropathic pain.
Collapse
Affiliation(s)
- X-T Deng
- Department of Neurobiology, School of Basic Medical Sciences, Neuroscience Research Institute, Peking University Health Science Center, Beijing, China
| | - M-Z Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Center for Anesthesiology & Pain Medicine, and Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - N Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Center for Anesthesiology & Pain Medicine, and Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - P-C Ma
- Department of Neurobiology, School of Basic Medical Sciences, Neuroscience Research Institute, Peking University Health Science Center, Beijing, China
| | - X-J Song
- Department of Neurobiology, School of Basic Medical Sciences, Neuroscience Research Institute, Peking University Health Science Center, Beijing, China.,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education of China), Center for Anesthesiology & Pain Medicine, and Department of Anesthesiology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
15
|
Bhatia S, Baig NA, Timofeeva O, Pasquale EB, Hirsch K, MacDonald TJ, Dritschilo A, Lee YC, Henkemeyer M, Rood B, Jung M, Wang XJ, Kool M, Rodriguez O, Albanese C, Karam SD. Knockdown of EphB1 receptor decreases medulloblastoma cell growth and migration and increases cellular radiosensitization. Oncotarget 2016; 6:8929-46. [PMID: 25879388 PMCID: PMC4496193 DOI: 10.18632/oncotarget.3369] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/11/2015] [Indexed: 02/03/2023] Open
Abstract
The expression of members of the Eph family of receptor tyrosine kinases and their ephrin ligands is frequently dysregulated in medulloblastomas. We assessed the expression and functional role of EphB1 in medulloblastoma cell lines and engineered mouse models. mRNA and protein expression profiling showed expression of EphB1 receptor in the human medulloblastoma cell lines DAOY and UW228. EphB1 downregulation reduced cell growth and viability, decreased the expression of important cell cycle regulators, and increased the percentage of cells in G1 phase of the cell cycle. It also modulated the expression of proliferation, and cell survival markers. In addition, EphB1 knockdown in DAOY cells resulted in significant decrease in migration, which correlated with decreased β1-integrin expression and levels of phosphorylated Src. Furthermore, EphB1 knockdown enhanced cellular radiosensitization of medulloblastoma cells in culture and in a genetically engineered mouse medulloblastoma model. Using genetically engineered mouse models, we established that genetic loss of EphB1 resulted in a significant delay in tumor recurrence following irradiation compared to EphB1-expressing control tumors. Taken together, our findings establish that EphB1 plays a key role in medulloblastoma cell growth, viability, migration, and radiation sensitivity, making EphB1 a promising therapeutic target.
Collapse
Affiliation(s)
- Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Nimrah A Baig
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Olga Timofeeva
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | - Kellen Hirsch
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Tobey J MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anatoly Dritschilo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Georgetown University Hospital, Washington, DC, USA
| | - Yi Chien Lee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Mark Henkemeyer
- Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brian Rood
- Children's National Medical Center, Washington DC, USA
| | - Mira Jung
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Xiao-Jing Wang
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Marcel Kool
- Division of Pediatric Neurooncology, German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Pathology, Georgetown University School of Medicine, Washington, DC, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
16
|
Bakhtazad A, Vousooghi N, Garmabi B, Zarrindast MR. CART peptide and opioid addiction: Expression changes in male rat brain. Neuroscience 2016; 325:63-73. [PMID: 26955782 DOI: 10.1016/j.neuroscience.2016.02.071] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 02/09/2016] [Accepted: 02/29/2016] [Indexed: 01/21/2023]
Abstract
Previous studies have shown the prominence of cocaine- and amphetamine-regulated transcript (CART) peptide in rewarding and reinforcing effects of drugs of abuse specially psychostimulants. The data regarding the effects of different stages of opioid addiction on CART expression and the interconnection between CART and opioids are not much available. Here we have studied the changes in the expression level of CART mRNA and protein in various parts of the brain reward pathway in different stages of opioid addiction. Groups of male rats received acute low-dose (10mg/kg), acute high-dose (80mg/kg) and chronic escalating doses of morphine. In addition, withdrawal and abstinence states were evaluated after injection of naloxone (1mg/kg) and long-term maintenance of addicted animals, respectively. Expression of CART mRNA in the brain was measured by real-time PCR method. Western blotting was used to quantify the protein level. CART mRNA and protein were both up-regulated in high-dose morphine-administered animals and also in the withdrawal group in the nucleus accumbens (NAc), striatum and prefrontal cortex (PFC). In the addicted group, CART mRNA and protein were both down-regulated in NAc and striatum. In the abstinent group, CART mRNA was down-regulated in NAc. In the hippocampus, the only observed change was the up-regulation of CART mRNA in the withdrawal group. We suggest that the modulatory role of CART peptide in rewarding and reinforcing effects of opioids weakens when opioids are used for a long time and is stimulated when acute stress such as naloxone-induced withdrawal syndrome or acute high-dose administration of morphine occurs to the animal.
Collapse
Affiliation(s)
- A Bakhtazad
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - N Vousooghi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - B Garmabi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - M R Zarrindast
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran; Genomics Center, School of Advanced Sciences, Tehran Medical Branch, Islamic Azad University, Tehran, Iran; School of Cognitive Sciences, Institute for Studies in Theoretical Physics and Mathematics, Tehran, Iran.
| |
Collapse
|
17
|
Huroy S, Kanawaty A, Magomedova L, Cummins CL, George SR, van der Kooy D, Henderson JT. EphB2 reverse signaling regulates learned opiate tolerance via hippocampal function. Behav Brain Res 2016; 300:85-96. [DOI: 10.1016/j.bbr.2015.09.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 11/27/2022]
|
18
|
Effect of circadian rhythm disturbance on morphine preference and addiction in male rats: Involvement of period genes and dopamine D1 receptor. Neuroscience 2016; 322:104-14. [PMID: 26892296 DOI: 10.1016/j.neuroscience.2016.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/16/2016] [Accepted: 02/10/2016] [Indexed: 12/17/2022]
Abstract
It is claimed that a correlation exists between disturbance of circadian rhythms by factors such as alteration of normal light-dark cycle and the development of addiction. However, the exact mechanisms involved in this relationship are not much understood. Here we have studied the effect of constant light on morphine voluntary consumption and withdrawal symptoms and also investigated the involvement of Per1, Per2 and dopamine D1 receptor in these processes. Male wistar rats were kept under standard (LD) or constant light (LL) conditions for one month. The plasma concentration of melatonin was evaluated by enzyme-linked immunosorbent assay (ELISA). Real-time PCR was used to determine the mRNA expression of Per1, Per2 and dopamine D1 receptor in the striatum and prefrontal cortex. Morphine preference (50mg/L) was evaluated in a two-bottle-choice paradigm for 10 weeks and withdrawal symptoms were recorded after administration of naloxone (3mg/kg). One month exposure to constant light resulted in a significant decrease of melatonin concentration in the LL group. In addition, mRNA levels of Per2 and dopamine D1 receptor were up-regulated in both the striatum and prefrontal cortex of the LL group. However, expression of Per1 gene was only up-regulated in the striatum of LL rats in comparison to LD animals. Furthermore, after one month exposure to constant light, morphine consumption and preference ratio and also severity of naloxone-induced withdrawal syndrome were significantly greater in LL animals. It is concluded that exposure to constant light by up-regulation of Per2 and dopamine D1 receptor in the striatum and prefrontal cortex and up-regulation of Per1 in the striatum and the possible involvement of melatonin makes animals vulnerable to morphine preference and addiction.
Collapse
|
19
|
Song XJ, Huang ZJ, Song WB, Song XS, Fuhr AF, Rosner AL, Ndtan H, Rupert RL. Attenuation Effect of Spinal Manipulation on Neuropathic and Postoperative Pain Through Activating Endogenous Anti-Inflammatory Cytokine Interleukin 10 in Rat Spinal Cord. J Manipulative Physiol Ther 2016; 39:42-53. [PMID: 26837229 DOI: 10.1016/j.jmpt.2015.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate roles of the anti-inflammatory cytokine interleukin (IL) 10 and the proinflammatory cytokines IL-1β and tumor necrosis factor α (TNF-α) in spinal manipulation-induced analgesic effects of neuropathic and postoperative pain. METHODS Neuropathic and postoperative pain were mimicked by chronic compression of dorsal root ganglion (DRG) (CCD) and decompression (de-CCD) in adult, male, Sprague-Dawley rats. Behavioral pain after CCD and de-CCD was determined by the increased thermal and mechanical hypersensitivity of the affected hindpaw. Hematoxylin and eosin staining, whole-cell patch clamp electrophysiological recordings, immunohistochemistry, and enzyme-linked immunosorbent assay were used to examine the neural inflammation, neural excitability, and expression of c-Fos and PKC as well as levels of IL-1β, TNF-α, and IL-10 in blood plasma, DRG, or the spinal cord. We used the activator adjusting instrument, a chiropractic spinal manipulative therapy tool, to deliver force to the spinous processes of L5 and L6. RESULTS After CCD and de-CCD treatments, the animals exhibited behavioral and neurochemical signs of neuropathic pain manifested as mechanical allodynia and thermal hyperalgesia, DRG inflammation, DRG neuron hyperexcitability, induction of c-Fos, and the increased expression of PKCγ in the spinal cord as well as increased level of IL-1β and TNF-α in DRG and the spinal cord. Repetitive Activator-assisted spinal manipulative therapy significantly reduced simulated neuropathic and postoperative pain, inhibited or reversed the neurochemical alterations, and increased the anti-inflammatory IL-10 in the spinal cord. CONCLUSION These findings show that spinal manipulation may activate the endogenous anti-inflammatory cytokine IL-10 in the spinal cord and thus has the potential to alleviate neuropathic and postoperative pain.
Collapse
Affiliation(s)
- Xue-Jun Song
- Professor, Parker University, Parker Research Institute, Dallas, TX.
| | - Zhi-Jiang Huang
- Research Scientist, Parker University, Parker Research Institute, Dallas, TX
| | - William B Song
- (Volunteer) Research Assistant, Parker University, Parker Research Institute, Dallas, TX
| | - Xue-Song Song
- Research Scientist, Parker University, Parker Research Institute, Dallas, TX
| | - Arlan F Fuhr
- Professor, Parker University, Parker Research Institute, Dallas, TX
| | - Anthony L Rosner
- Research Consultant, Parker University, Parker Research Institute, Dallas, TX
| | - Harrison Ndtan
- Associate Professor, Parker University, Parker Research Institute, Dallas, TX
| | - Ronald L Rupert
- Research Consultant, Parker University, Parker Research Institute, Dallas, TX
| |
Collapse
|
20
|
Dines M, Lamprecht R. The Role of Ephs and Ephrins in Memory Formation. Int J Neuropsychopharmacol 2015; 19:pyv106. [PMID: 26371183 PMCID: PMC4851260 DOI: 10.1093/ijnp/pyv106] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/10/2015] [Indexed: 12/22/2022] Open
Abstract
The ability to efficiently store memories in the brain is a fundamental process and its impairment is associated with multiple human mental disorders. Evidence indicates that long-term memory formation involves alterations of synaptic efficacy produced by modifications in neural transmission and morphology. The Eph receptors and their cognate ephrin ligands have been shown to be involved in these key neuronal processes by regulating events such as presynaptic transmitter release, postsynaptic glutamate receptor conductance and trafficking, synaptic glutamate reuptake, and dendritic spine morphogenesis. Recent findings show that Ephs and ephrins are needed for memory formation in different organisms. These proteins participate in the formation of various types of memories that are subserved by different neurons and brain regions. Ephs and ephrins are involved in brain disorders and diseases with memory impairment symptoms, including Alzheimer's disease and anxiety. Drugs that agonize or antagonize Ephs/ephrins signaling have been developed and could serve as therapeutic agents to treat such diseases. Ephs and ephrins may therefore induce cellular alterations mandatory for memory formation and serve as a target for pharmacological intervention for treatment of memory-related brain diseases.
Collapse
Affiliation(s)
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Israel.
| |
Collapse
|
21
|
Zhou XL, Wang Y, Zhang CJ, Yu LN, Cao JL, Yan M. COX-2 is required for the modulation of spinal nociceptive information related to ephrinB/EphB signalling. Eur J Pain 2015; 19:1277-87. [PMID: 25919495 DOI: 10.1002/ejp.657] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2014] [Indexed: 01/09/2023]
Affiliation(s)
- X.-L. Zhou
- Department of Anesthesiology; School of Medicine; The Second Affiliated Hospital; Zhejiang University; Hangzhou China
| | - Y. Wang
- Jiangsu Province Key Laboratory of Anesthesilogy; Xuzhou Medical College; China
| | - C.-J. Zhang
- Department of Gastroenterology; School of Medicine; The Second Affiliated Hospital; Zhejiang University; Hangzhou China
| | - L.-N. Yu
- Department of Anesthesiology; School of Medicine; The Second Affiliated Hospital; Zhejiang University; Hangzhou China
| | - J.-L. Cao
- Jiangsu Province Key Laboratory of Anesthesilogy; Xuzhou Medical College; China
| | - M. Yan
- Department of Anesthesiology; School of Medicine; The Second Affiliated Hospital; Zhejiang University; Hangzhou China
- Jiangsu Province Key Laboratory of Anesthesilogy; Xuzhou Medical College; China
| |
Collapse
|
22
|
PKA is required for the modulation of spinal nociceptive information related to ephrinB-EphB signaling in mice. Neuroscience 2014; 284:546-554. [PMID: 25453775 DOI: 10.1016/j.neuroscience.2014.10.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 09/20/2014] [Accepted: 10/15/2014] [Indexed: 01/06/2023]
Abstract
EphB receptors and their ephrinB ligands are implicated in modulating of spinal nociceptive information processing. Here, we investigated whether protein kinase A (PKA), acts as a downstream effector, participates in the modulation spinal nociceptive information related to ephrinB-EphB signaling. Intrathecal injection of ephrinB2-Fc caused thermal hyperalgesia and mechanical allodynia, which were accompanied by increased expression of spinal PKA catalytic subunit (PKAca) and phosphorylated cAMP-response element-binding protein (p-CREB). Pre-treatment with H89, a PKA inhibitor, prevented the activation of CREB by ephrinB2-Fc. Inhibition of spinal PKA signaling prevented and reversed pain behaviors induced by the intrathecal injection of ephrinB2-Fc. Furthermore, blockade of the EphB receptors by intrathecal injection of EphB2-Fc reduced formalin-induced inflammatory, chronic constrictive injury (CCI)-induced neuropathic, and tibia bone cavity tumor cell implantation (TCI)-induced bone cancer pain behaviors, which were accompanied by decreased expression of spinal PKAca and p-CREB. Overall, these results confirmed the important involvement of PKA in the modulation of spinal nociceptive information related to ephrinBs-EphBs signaling. This finding may have important implications for exploring the roles and mechanisms of ephrinB-EphB signaling in physiologic and pathologic pain.
Collapse
|
23
|
Liu S, Liu YP, Song WB, Song XJ. EphrinB-EphB receptor signaling contributes to bone cancer pain via Toll-like receptor and proinflammatory cytokines in rat spinal cord. Pain 2013; 154:2823-2835. [DOI: 10.1016/j.pain.2013.08.017] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 12/29/2022]
|
24
|
Logan SM, Romero MI, Nguyen DH, Benson MD. Ephrin-B2 expression in the proprioceptive sensory system. Neurosci Lett 2013; 545:69-74. [PMID: 23623938 PMCID: PMC3867235 DOI: 10.1016/j.neulet.2013.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 04/04/2013] [Accepted: 04/10/2013] [Indexed: 11/17/2022]
Abstract
Recent studies have shown that ephrin-B2 on sensory afferent fibers from the dorsal root ganglia (DRG) controls transmission of pain sensation to the spinal cord. We examined ephrin-B2 expression in mouse DRG and spinal cord using an ephrin-B2/ß-galactosidase chimeric allele. We found that ephrin-B2 is expressed exclusively in proprioceptive neurons and fibers in neonates, while expression in lamina III and IV of the adult spinal cord was observed in addition to that in the deeper laminae. We confirmed that ephrin-B2 protein causes co-clustering of EphB2 and glutamate receptors in spinal cord neurons. Our data are consistent with a role for ephrin-B2 in transmission of positional information to the CNS, and thus suggest a role in synaptic plasticity of spinal cord locomotor circuits that are known to be sensitive to proprioceptive sensory input after spinal cord injury.
Collapse
Affiliation(s)
- Shaun M. Logan
- Department of Biomedical Sciences, Texas A&M Health Science Center Baylor College of Dentistry, 3302 Gaston Ave., Dallas, TX, 75246
| | - Mario I. Romero
- Department of Bioengineering, The University of Texas Arlington, 500 UTA Blvd., Arlington, TX, 76019
| | - Dianna H. Nguyen
- Department of Bioengineering, The University of Texas Arlington, 500 UTA Blvd., Arlington, TX, 76019
| | - M. Douglas Benson
- Department of Biomedical Sciences, Texas A&M Health Science Center Baylor College of Dentistry, 3302 Gaston Ave., Dallas, TX, 75246
| |
Collapse
|
25
|
Zhang YK, Huang ZJ, Liu S, Liu YP, Song AA, Song XJ. WNT signaling underlies the pathogenesis of neuropathic pain in rodents. J Clin Invest 2013; 123:2268-86. [PMID: 23585476 DOI: 10.1172/jci65364] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 02/14/2013] [Indexed: 12/24/2022] Open
Abstract
Treating neuropathic pain is a major clinical challenge, and the underlying mechanisms of neuropathic pain remain elusive. We hypothesized that neuropathic pain-inducing nerve injury may elicit neuronal alterations that recapitulate events that occur during development. Here, we report that WNT signaling, which is important in developmental processes of the nervous system, plays a critical role in neuropathic pain after sciatic nerve injury and bone cancer in rodents. Nerve injury and bone cancer caused a rapid-onset and long-lasting expression of WNTs, as well as activation of WNT/frizzled/β-catenin signaling in the primary sensory neurons, the spinal dorsal horn neurons, and astrocytes. Spinal blockade of WNT signaling pathways inhibited the production and persistence of neuropathic pain and the accompanying neurochemical alterations without affecting normal pain sensitivity and locomotor activity. WNT signaling activation stimulated production of the proinflammatory cytokines IL-18 and TNF-α and regulated the NR2B glutamate receptor and Ca2+-dependent signals through the β-catenin pathway in the spinal cord. These findings indicate a critical mechanism underlying the pathogenesis of neuropathic pain and suggest that targeting the WNT signaling pathway may be an effective approach for treating neuropathic pain, including bone cancer pain.
Collapse
Affiliation(s)
- Yan-Kai Zhang
- Department of Neurobiology, Parker University Research Institute, Dallas, Texas 75229, USA
| | | | | | | | | | | |
Collapse
|
26
|
Involvement of EphB1 receptors signalling in models of inflammatory and neuropathic pain. PLoS One 2013; 8:e53673. [PMID: 23341972 PMCID: PMC3547059 DOI: 10.1371/journal.pone.0053673] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 12/03/2012] [Indexed: 12/12/2022] Open
Abstract
EphB receptors tyrosine kinases and ephrinB ligands were first identified as guidance molecules involved in the establishment of topographical mapping and connectivity in the nervous system during development. Later in development and into adulthood their primary role would switch from guidance to activity-dependent modulation of synaptic efficacy. In sensory systems, they play a role in both the onset of inflammatory and neuropathic pain, and in the establishment of central sensitisation, an NMDA-mediated form of synaptic plasticity thought to underlie most forms of chronic pain. We studied wild type and EphB1 knockout mice in a range of inflammatory and neuropathic pain models to determine 1), whether EphB1 expression is necessary for the onset and/or maintenance of persistent pain, regardless of origin; 2), whether in these models cellular and molecular changes, e.g. phosphorylation of the NR2B subunit of the NMDA receptor, increased c-fos expression or microglial activation, associated with the onset of pain, are affected by the lack of functional EphB1 receptors. Differences in phenotype were examined behaviourally, anatomically, biochemically and electrophysiologically. Our results establish firstly, that functional EphB1 receptors are not essential for the development of normal nociception, thermal or mechanical sensitivity. Secondly, they demonstrate a widespread involvement of EphB1 receptors in chronic pain. NR2B phosphorylation, c-fos expression and microglial activation are all reduced in EphB1 knockout mice. This last finding is intriguing, since microglial activation is supposedly triggered directly by primary afferents, therefore it was not expected to be affected. Interestingly, in some models of long-term pain (days), mechanical and thermal hyperalgesia develop both in wild type and EphB1 knockout mice, but recovery is faster in the latter, indicating that in particular models these receptors are required for the maintenance, rather than the onset of, thermal and mechanical hypersensitivity. This potentially makes them an attractive target for analgesic strategies.
Collapse
|
27
|
Song XJ. Hyperbaric oxygen therapy attenuates chronic pain and inhibits nerve injury-induced activation of ephrinB-EphB receptor signalling in the spinal cord: In response to the Letter to the Editor from Dr. Shailendra Kapoor. Eur J Pain 2012. [DOI: 10.1002/j.1532-2149.2012.00191.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Xue-Jun Song
- Department of Neurobiology; Parker University Research Institute; Dallas; TX; USA
| |
Collapse
|
28
|
Sheffler-Collins SI, Dalva MB. EphBs: an integral link between synaptic function and synaptopathies. Trends Neurosci 2012; 35:293-304. [PMID: 22516618 DOI: 10.1016/j.tins.2012.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/08/2012] [Accepted: 03/08/2012] [Indexed: 11/19/2022]
Abstract
The assembly and function of neuronal circuits rely on selective cell-cell interactions to control axon targeting, generate pre- and postsynaptic specialization and recruit neurotransmitter receptors. In neurons, EphB receptor tyrosine kinases mediate excitatory synaptogenesis early during development, and then later coordinate synaptic function by controlling synaptic glutamate receptor localization and function. EphBs direct synapse formation and function to regulate cellular morphology through downstream signaling mechanisms and by interacting with glutamate receptors. In humans, defective EphB-dependent regulation of NMDA receptor (NMDAR) localization and function is associated with neurological disorders, including neuropathic pain, anxiety disorders and Alzheimer's disease (AD). Here, we propose that EphBs act as a central organizer of excitatory synapse formation and function, and as a key regulator of diseases linked to NMDAR dysfunction.
Collapse
Affiliation(s)
- Sean I Sheffler-Collins
- Department of Neuroscience and Farber Institute for Neurosciences, Thomas Jefferson University, 900 Walnut Street, Suite 462, Philadelphia, PA 19107, USA
| | | |
Collapse
|
29
|
Zhao J, Yuan G, Cendan CM, Nassar MA, Lagerström MC, Kullander K, Gavazzi I, Wood JN. Nociceptor-expressed ephrin-B2 regulates inflammatory and neuropathic pain. Mol Pain 2010; 6:77. [PMID: 21059214 PMCID: PMC2992507 DOI: 10.1186/1744-8069-6-77] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2010] [Accepted: 11/08/2010] [Indexed: 01/14/2023] Open
Abstract
Background EphB receptors and their ephrin-B ligands play an important role in nervous system development, as well as synapse formation and plasticity in the adult brain. Recent studies show that intrathecal treatment with EphB-receptor activator ephrinB2-Fc induced thermal hyperalgesia and mechanical allodynia in rat, indicating that ephrin-B2 in small dorsal root ganglia (DRG) neurons and EphB receptors in the spinal cord modulate pain processing. To examine the role of ephrin-B2 in peripheral pain pathways, we deleted ephrin-B2 in Nav1.8+ nociceptive sensory neurons with the Cre-loxP system. Sensory neuron numbers and terminals were examined using neuronal makers. Pain behavior in acute, inflammatory and neuropathic pain models was assessed in the ephrin-B2 conditional knockout (CKO) mice. We also investigated the c-Fos expression and NMDA receptor NR2B phosphorylation in ephrin-B2 CKO mice and littermate controls. Results The ephrin-B2 CKO mice were healthy with no sensory neuron loss. However, pain-related behavior was substantially altered. Although acute pain behavior and motor co-ordination were normal, inflammatory pain was attenuated in ephrin-B2 mutant mice. Complete Freund's adjuvant (CFA)-induced mechanical hyperalgesia was halved. Formalin-induced pain behavior was attenuated in the second phase, and this correlated with diminished tyrosine phosphorylation of N-methyl-D-aspartic acid (NMDA) receptor subunit NR2B in the dorsal horn. Thermal hyperalgesia and mechanical allodynia were significantly reduced in the Seltzer model of neuropathic pain. Conclusions Presynaptic ephrin-B2 expression thus plays an important role in regulating inflammatory pain through the regulation of synaptic plasticity in the dorsal horn and is also involved in the pathogenesis of some types of neuropathic pain.
Collapse
Affiliation(s)
- Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Cruciform Building, University College London, London WC1E 6BT, UK
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Spinal matrix metalloproteinase-9 contributes to physical dependence on morphine in mice. J Neurosci 2010; 30:7613-23. [PMID: 20519536 DOI: 10.1523/jneurosci.1358-10.2010] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Preventing and reversing opioid dependence continues to be a clinical challenge and underlying mechanisms of opioid actions remain elusive. We report that matrix metalloproteinase-9 (MMP-9) in the spinal cord contributes to development of physical dependence on morphine. Chronic morphine exposure and naloxone-precipitated withdrawal increase activity of spinal MMP-9. Spinal inhibition or targeted mutation of MMP-9 suppresses behavioral signs of morphine withdrawal and the associated neurochemical alterations. The increased MMP-9 activity is mainly distributed in the superficial dorsal horn and colocalized primarily with neurons and small numbers of astrocytes and microglia. Morphine exposure and withdrawal increase phosphorylation of NR1 and NR2B receptors, ERK1/2, calmodulin-dependent kinase II, and cAMP response element binding proteins; and such phosphorylation is suppressed by either spinal inhibition or targeted mutation of MMP-9. Further, spinal administration of exogenous MMP-9 induces morphine withdrawal-like behavioral signs and mechanical allodynia, activates NR1 and NR2 receptors, and downregulates integrin-beta1, while a function-neutralizing antibody against integrin-beta1 suppresses MMP-9-induced phosphorylation of NR1 and NR2B. Morphine withdrawal-induced MMP-9 activity is also reduced by an nNOS inhibitor. Thus, we hypothesize that spinal MMP-9 may contribute to the development of morphine dependence primarily through neuronal activation and interaction with NR1 and NR2B receptors via integrin-beta1 and NO pathways. The other gelatinase, MMP-2, is not involved in morphine dependence. Inhibiting spinal MMP-9 or MMP-2 reduces chronic and/or acute morphine tolerance. This study suggests a novel therapeutic approach for preventing, minimizing, or reversing opioid dependence and tolerance.
Collapse
|
31
|
Pain research in China. SCIENCE CHINA-LIFE SCIENCES 2010; 53:356-362. [DOI: 10.1007/s11427-010-0065-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 02/24/2010] [Indexed: 12/29/2022]
|
32
|
Hoschouer EL, Basso MD, Jakeman LB. Aberrant sensory responses are dependent on lesion severity after spinal cord contusion injury in mice. Pain 2009; 148:328-342. [PMID: 20022699 DOI: 10.1016/j.pain.2009.11.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 10/07/2009] [Accepted: 11/23/2009] [Indexed: 11/28/2022]
Abstract
Following spinal cord injury (SCI), individuals lose normal sensation and often develop debilitating neuropathic pain. Basic research has helped to elucidate many of the underlying mechanisms, but unanswered questions remain concerning how sensation changes after SCI and potential negative consequences of regenerative therapies. Mouse models provide an opportunity to explore these questions using genetic markers and manipulations. However, despite the increasing use of mice in pain and sensory research, the responses to sensory stimuli after SCI are poorly characterized in this species. This study evaluated behavioral responses to mechanical and nociceptive stimuli applied to the hindlimbs and the dorsal trunk in C57BL/6 mice after mid-thoracic SCI. Adult mice were subjected to laminectomy, contusion injuries of different severities, or complete transections to test the hypothesis that the patterns of sensory pathology depend on the extent of tissue damage at the injury site. In the hind paws, hyper-responsiveness to a heat stimulus developed independent of injury severity, while mechanical sensitivity decreased, except after the most severe contusion injuries sparing less than 2% of the white matter at the injury site, when enhanced sensitivity was observed. On the trunk, mechanical and pin prick testing revealed diminished sensitivity at and below the injury level, while responses above the level of the injury were unchanged. The contrast in injury severity threshold for thermal and mechanical hypersensitivity in the hind paws suggests that these responses have different underlying mechanisms. These results establish essential baseline information for murine studies of pain and changes in sensation after SCI.
Collapse
Affiliation(s)
- Emily L Hoschouer
- Dept of Physiology and Cell Biology, The Ohio State University College of Medicine, 1645 Neil Ave., Columbus, OH 43210, USA Dept of Neuroscience, The Ohio State University College of Medicine, 333 W. 10th Ave., Columbus, OH 43210, USA Division of Physical Therapy, The Ohio State University School of Allied Medical Professions, 453 W. 10th Ave., Columbus, OH 43210, USA Neuroscience Graduate Studies Program, The Ohio State University, Columbus, OH 43210, USA Center for Brain and Spinal Cord Repair, The Ohio State University College of Medicine, 460 W. 12th Ave., Columbus, OH 43210, USA
| | | | | |
Collapse
|
33
|
Abstract
This paper is the 31st consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2008 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration and thermoregulation (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd, Flushing, NY 11367, United States.
| |
Collapse
|
34
|
Liu WT, Han Y, Li HC, Adams B, Zheng JH, Wu YP, Henkemeyer M, Song XJ. An in vivo mouse model of long-term potentiation at synapses between primary afferent C-fibers and spinal dorsal horn neurons: essential role of EphB1 receptor. Mol Pain 2009; 5:29. [PMID: 19523204 PMCID: PMC2704201 DOI: 10.1186/1744-8069-5-29] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 06/12/2009] [Indexed: 11/10/2022] Open
Abstract
Background Long-term potentiation (LTP), a much studied cellular model of synaptic plasticity, has not been demonstrated at synapses between primary afferent C-fibers and spinal dorsal horn (DH) neurons in mice in vivo. EphrinB-EphB receptor signaling plays important roles in synaptic connection and plasticity in the nervous system, but its role in spinal synaptic plasticity remains unclear. Results This study characterizes properties of LTP at synapses of C-fibers onto neurons in the superficial DH following high-frequency stimulation (HFS) of a peripheral nerve at an intensity that activates C-fibers and examines associated activation of Ca2+/calmodulin-activated protein kinase II (p-CaMKII), extracellular signal-regulated kinase (p-ERK) and the cyclic AMP response element binding protein (p-CREB) and expression of c-Fos, and it investigates further roles for the EphB1 receptor in LTP. HFS induced LTP within 5 min and lasts for 3–8 h during the period of recording and resulted in upregulation of p-CaMKII, p-ERK and p-CREB protein levels in the spinal cord and expression of c-Fos in DH. Intrathecal pretreatment of MK-801 or EphB2-Fc prevented LTP and significantly reduced upregulation of p-CaMKII, p-ERK, p-CREB and c-Fos. Further, targeted mutation of EphB1 receptor prevented induction of LTP and associated increases in phosphorylation of CaMKII, ERK, and CREB. Conclusion This study provides an in vivo mouse model of LTP at synapses of C-fibers onto the superficial DH neurons that will be valuable for studying the DH neuron excitability and their synaptic plasticity and hyperalgesia. It further takes advantage of examining functional implications of a specific gene targeted mice and demonstrates that the EphB1 receptor is essential for development of LTP.
Collapse
Affiliation(s)
- Wen-Tao Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu, PR China.
| | | | | | | | | | | | | | | |
Collapse
|