1
|
Zheng W, Huang X, Wang J, Gao F, Chai Z, Zeng J, Li S, Yu C. The chronification mechanism of orofacial inflammatory pain: Facilitation by GPER1 and microglia in the rostral ventral medulla. Front Mol Neurosci 2023; 15:1078309. [PMID: 36683848 PMCID: PMC9853019 DOI: 10.3389/fnmol.2022.1078309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 01/08/2023] Open
Abstract
Background Chronic orofacial pain is a common and incompletely defined clinical condition. The role of G protein-coupled estrogen receptor 1 (GPER1) as a new estrogen receptor in trunk and visceral pain regulation is well known. Here, we researched the role of GPER1 in the rostral ventral medulla (RVM) during chronic orofacial pain. Methods and Results A pain model was established where rats were injected in the temporomandibular joint with complete Freund's adjuvant (CFA) to simulate chronic orofacial pain. Following this a behavioral test was performed to establish pain threshold and results showed that the rats injected with CFA had abnormal pain in the orofacial regions. Additional Immunostaining and blot analysis indicated that microglia were activated in the RVM and GPER1 and c-Fos were significantly upregulated in the rats. Conversely, when the rats were injected with G15 (a GPER1 inhibitor) the abnormal pain the CFA rats were experiencing was alleviated and microglia activation was prevented. In addition, we found that G15 downregulated the expression of phospholipase C (PLC) and protein kinase C (PKC), inhibited the expression of GluA1, restores aberrant synaptic plasticity and reduces the overexpression of the synapse-associated proteins PSD-95 and syb-2 in the RVM of CFA rats. Conclusion The findings indicate that GPER1 mediates chronic orofacial pain through modulation of the PLC-PKC signal pathway, sensitization of the RVM region and enhancement of neural plasticity. These results of this study therefore suggest that GPER1 may serve as a potential therapeutic target for chronic orofacial pain.
Collapse
Affiliation(s)
- Wenwen Zheng
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Xilu Huang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jing Wang
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Feng Gao
- The Sixth People’s Hospital of Chongqing, Anesthesiology, Chongqing, China
| | - Zhaowu Chai
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Jie Zeng
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Sisi Li
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China
| | - Cong Yu
- The Affiliated Hospital of Stomatology, Chongqing Medical University, Chongqing, China,*Correspondence: Cong Yu, ✉
| |
Collapse
|
2
|
Genaro K, Prado WA. The role of the anterior pretectal nucleus in pain modulation: A comprehensive review. Eur J Neurosci 2021; 54:4358-4380. [PMID: 33909941 DOI: 10.1111/ejn.15255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 11/27/2022]
Abstract
Descending pain modulation involves multiple encephalic sites and pathways that range from the cerebral cortex to the spinal cord. Behavioral studies conducted in the 1980s revealed that electrical stimulation of the pretectal area causes antinociception dissociation from aversive responses. Anatomical and physiological studies identified the anterior pretectal nucleus and its descending projections to several midbrain, pontine, and medullary structures. The anterior pretectal nucleus is morphologically divided into a dorsal part that contains a dense neuron population (pars compacta) and a ventral part that contains a dense fiber band network (pars reticulata). Connections of the two anterior pretectal nucleus parts are broad and include prominent projections to and from major encephalic systems associated with somatosensory processes. Since the first observation that acute or chronic noxious stimuli activate the anterior pretectal nucleus, it has been established that numerous mediators participate in this response through distinct pathways. Recent studies have confirmed that at least two pain inhibitory pathways are activated from the anterior pretectal nucleus. This review focuses on rodent anatomical, behavioral, molecular, and neurochemical data that have helped to identify mediators of the anterior pretectal nucleus and pathways related to its role in pain modulation.
Collapse
Affiliation(s)
- Karina Genaro
- Department of Anesthesiology, University of California, Irvine, CA, USA
| | - Wiliam A Prado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Costa AR, Carvalho P, Flik G, Wilson SP, Reguenga C, Martins I, Tavares I. Neuropathic Pain Induced Alterations in the Opioidergic Modulation of a Descending Pain Facilitatory Area of the Brain. Front Cell Neurosci 2019; 13:287. [PMID: 31316354 PMCID: PMC6610065 DOI: 10.3389/fncel.2019.00287] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/13/2019] [Indexed: 12/03/2022] Open
Abstract
Opioids play a major role at descending pain modulation but the effects of neuropathic pain on the brain opioidergic system remain understudied. Since descending facilitation is enhanced during neuropathic pain, we studied the opioidergic modulation of the dorsal reticular nucleus (DRt), a medullary pain facilitatory area, in the spared nerve injury (SNI) model of neuropathic pain. We first performed a series of behavioral experiments in naïve-animals to establish the role of μ-opioid receptor (MOR) in the effects of endogenous and exogenous opioids at the DRt. Specifically, we showed that lentiviral-mediated MOR-knockdown at the DRt increased sensitivity to thermal and mechanical stimuli while the MOR agonist DAMGO induced the opposite effects. Additionally, we showed that MOR-knockdown and the pharmacological blockade of MOR by CTAP at the DRt decreased and inhibited, respectively, the analgesic effects of systemic morphine. Then, we performed in vivo microdialysis to measure enkephalin peptides in the DRt and evaluated MOR expression in the DRt at mRNA, protein and phosphorylated form levels by quantitative real-time PCR and immunohistochemistry, respectively. SNI-animals, compared to sham control, showed higher levels of enkephalin peptides, lower MOR-labeled cells without alterations in MOR mRNA levels, and higher phosphorylated MOR-labeled cells. Finally, we performed behavioral studies in SNI animals to determine the potency of systemic morphine and the effects of the pharmacologic and genetic manipulation of MOR at the DRt. We showed a reduced potency of the antiallodynic effects of systemic morphine in SNI-animals compared to the antinociceptive effects in sham animals. Increasing MOR-cells at the DRt of SNI-animals by lentiviral-mediated MOR-overexpression produced no effects on mechanical allodynia. DAMGO induced anti-allodynia only after MOR-overexpression. These results show that MOR inhibits DRt pain facilitatory actions and that this action contributes to the analgesic effects of systemic opioids. We further show that the inhibitory function of MOR is impaired during neuropathic pain. This is likely due to desensitization and degradation of MOR which are adaptations of the receptor that can be triggered by MOR phosphorylation. Skipping counter-regulatory pathways involved in MOR adaptations might restore the opioidergic inhibition at pain facilitatory areas.
Collapse
Affiliation(s)
- Ana Rita Costa
- Departamento de Biomedicina - Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Paulina Carvalho
- Departamento de Biomedicina - Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Gunnar Flik
- Charles River Laboratories Den Bosch B.V., 's-Hertogenbosch, Netherlands
| | - Steven P Wilson
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Carlos Reguenga
- Departamento de Biomedicina - Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Isabel Martins
- Departamento de Biomedicina - Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Isaura Tavares
- Departamento de Biomedicina - Unidade de Biologia Experimental, Faculdade de Medicina da Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
4
|
Martínez-Navarro M, Maldonado R, Baños JE. Why mu-opioid agonists have less analgesic efficacy in neuropathic pain? Eur J Pain 2018; 23:435-454. [PMID: 30318675 DOI: 10.1002/ejp.1328] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/06/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022]
Abstract
Injury to peripheral nerves often leads to abnormal pain states (hyperalgesia, allodynia and spontaneous pain), which can remain long after the injury heals. Although opioid agonists remain the gold standard for the treatment of moderate to severe pain, they show reduced efficacy against neuropathic pain. In addition to analgesia, opioid use is also associated with hyperalgesia and analgesia tolerance, whose underlying mechanisms share some commonalities with nerve injury-induced hypersensitivity. Here, we reviewed up-to-day research exploring the contribution of mu-opioid receptor (MOR) on the pathophysiology of neuropathic pain and on analgesic opioid actions under these conditions. We focused on the specific contributions of MOR populations at peripheral, spinal and supraspinal level. Moreover, evidences of neuroplastic changes that may underlie the low efficacy of MOR agonists under neuropathic pain conditions are reviewed and discussed. Sensitization processes leading to pain hypersensitivity, molecular changes in signalling pathways triggered by MOR and glial activation are some of these mechanisms elicited by both nerve injury and opioid exposure. Nerve injury-induced pain hypersensitivity might be masking the initial analgesic effects of opioid agonists, and alternatively, sustained opioid treatment to individuals already suffering from neuropathic pain could aggravate their pathophysiological state. Finally, some combined therapies that can increase opioid analgesic effectiveness in neuropathic pain treatment are highlighted. SIGNIFICANCE: This review provides evidence of the low benefit of opioid monotherapy in neuropathic pain and analyses the reasons of this reduced effectiveness. Opioid agonists along with drugs targeted to block the sensitization processes induced by MOR stimulation might result in a better management of neuropathic pain.
Collapse
Affiliation(s)
- Miriam Martínez-Navarro
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| | - Rafael Maldonado
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| | - Josep-E Baños
- Department of Experimental and Health Sciences, Laboratory of Neuropharmacology, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
5
|
Bar-Shalita T, Livshitz A, Levin-Meltz Y, Rand D, Deutsch L, Vatine JJ. Sensory modulation dysfunction is associated with Complex Regional Pain Syndrome. PLoS One 2018; 13:e0201354. [PMID: 30091986 PMCID: PMC6084887 DOI: 10.1371/journal.pone.0201354] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 07/14/2018] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Complex Regional Pain Syndrome (CRPS), a chronic pain condition, develops mainly after limb trauma and severely inhibits function. While early diagnosis is essential, factors for CRPS onset are elusive. Therefore, identifying those at risk is crucial. Sensory modulation dysfunction (SMD), affects the capacity to regulate responses to sensory input in a graded and adaptive manner and was found associated with hyperalgesia in otherwise healthy individuals, suggestive of altered pain processing. AIM To test SMD as a potential risk factor for CRPS. METHODS In this cross-sectional study, forty-four individuals with CRPS (29.9±11 years, 27 men) and 204 healthy controls (27.4±3.7 years, 105 men) completed the Sensory Responsiveness Questionnaire-Intensity Scale (SRQ-IS). A physician conducted the CRPS Severity Score (CSS), testing individuals with CRPS. RESULTS Thirty-four percent of the individuals with CRPS and twelve percent of the healthy individuals were identified to have SMD (χ2 (1) = 11.95; p<0.001). Logistic regression modeling revealed that the risk of CRPS is 2.68 and 8.21 times higher in individuals with sensory over- and sensory under-responsiveness, respectively, compared to non-SMD individuals (p = 0.03 and p = 0.01, respectively). CONCLUSIONS SMD, particularly sensory under-responsiveness, might serve as a potential risk factor for CRPS and therefore screening for SMD is recommended. This study provides the risk index probability clinical tool a simple evaluation to be applied by clinicians in order to identify those at risk for CRPS immediately after injury. Further research is needed.
Collapse
Affiliation(s)
- Tami Bar-Shalita
- Department of Occupational Therapy, School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Anatoly Livshitz
- Center for Rehabilitation of Pain Syndromes, Reuth Rehabilitation Hospital, Tel Aviv, Israel
| | - Yulia Levin-Meltz
- Center for Rehabilitation of Pain Syndromes, Reuth Rehabilitation Hospital, Tel Aviv, Israel
| | - Debbie Rand
- Department of Occupational Therapy, School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lisa Deutsch
- BioStats Statistical Consulting Ltd, Modiin, Israel
| | - Jean-Jacques Vatine
- Center for Rehabilitation of Pain Syndromes, Reuth Rehabilitation Hospital, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Dias QM, Prado WA. The lesion of dorsolateral funiculus changes the antiallodynic effect of the intrathecal muscimol and baclofen in distinct phases of neuropathic pain induced by spinal nerve ligation in rats. Brain Res Bull 2016; 124:103-15. [DOI: 10.1016/j.brainresbull.2016.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/20/2022]
|
7
|
Abstract
Pain is more than merely nociception and response, but rather it encompasses emotional, behavioral and cognitive components that make up the pain experience. With the recent advances in imaging techniques, we now understand that nociceptive inputs can result in the activation of complex interactions among central sites, including cortical regions that are active in cognitive, emotional and reward functions. These sites can have a bimodal influence on the serotonergic and noradrenergic descending pain modulatory systems via communications among the periaqueductal gray, rostral ventromedial medulla and pontine noradrenergic nuclei, ultimately either facilitating or inhibiting further nociceptive inputs. Understanding these systems can help explain the emotional and cognitive influences on pain perception and placebo/nocebo effects, and can help guide development of better pain therapeutics.
Collapse
Affiliation(s)
- Milena De Felice
- The University of Sheffield, Academic Unit of Oral & Maxillofacial Medicine & Surgery, Sheffield, South Yorkshire, UK
| | - Michael H Ossipov
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ 85724-5050, USA
| |
Collapse
|
8
|
Patel R, Rutten K, Valdor M, Schiene K, Wigge S, Schunk S, Damann N, Christoph T, Dickenson AH. Electrophysiological characterization of activation state-dependent Ca(v)2 channel antagonist TROX-1 in spinal nerve injured rats. Neuroscience 2015; 297:47-57. [PMID: 25839150 PMCID: PMC4436437 DOI: 10.1016/j.neuroscience.2015.03.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/13/2015] [Accepted: 03/24/2015] [Indexed: 11/27/2022]
Abstract
TROX-1 exhibits activation state-dependent inhibition of Cav2.2 in vitro. TROX-1 selectively attenuates neuronal responses to mechanical stimulation. Anti-nociceptive effect of TROX-1 dependent on pathophysiological state.
Prialt, a synthetic version of Cav2.2 antagonist ω-conotoxin MVIIA derived from Conus magus, is the first clinically approved voltage-gated calcium channel blocker for refractory chronic pain. However, due to the narrow therapeutic window and considerable side effects associated with systemic dosing, Prialt is only administered intrathecally. N-triazole oxindole (TROX-1) is a novel use-dependent and activation state-selective small-molecule inhibitor of Cav2.1, 2.2 and 2.3 calcium channels designed to overcome the limitations of Prialt. We have examined the neurophysiological and behavioral effects of blocking calcium channels with TROX-1. In vitro, TROX-1, in contrast to state-independent antagonist Prialt, preferentially inhibits Cav2.2 currents in rat dorsal root ganglia (DRG) neurons under depolarized conditions. In vivo electrophysiology was performed to record from deep dorsal horn lamina V/VI wide dynamic range neurons in non-sentient spinal nerve-ligated (SNL) and sham-operated rats. In SNL rats, spinal neurons exhibited reduced responses to innocuous and noxious punctate mechanical stimulation of the receptive field following subcutaneous administration of TROX-1, an effect that was absent in sham-operated animals. No effect was observed on neuronal responses evoked by dynamic brushing, heat or cold stimulation in SNL or sham rats. The wind-up response of spinal neurons following repeated electrical stimulation of the receptive field was also unaffected. Spinally applied TROX-1 dose dependently inhibited mechanically evoked neuronal responses in SNL but not sham-operated rats, consistent with behavioral observations. This study confirms the pathological state-dependent actions of TROX-1 through a likely spinal mechanism and reveals a modality selective change in calcium channel function following nerve injury.
Collapse
Affiliation(s)
- R Patel
- University College London, Gower Street, Department of Neuroscience, Physiology and Pharmacology, London WC1E 6BT, UK.
| | - K Rutten
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - M Valdor
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - K Schiene
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - S Wigge
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - S Schunk
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - N Damann
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - T Christoph
- Grünenthal GmbH, Global Preclinical Research, 52078 Aachen, Germany
| | - A H Dickenson
- University College London, Gower Street, Department of Neuroscience, Physiology and Pharmacology, London WC1E 6BT, UK
| |
Collapse
|
9
|
Suardíaz M, Galan-Arriero I, Avila-Martin G, Estivill-Torrús G, de Fonseca FR, Chun J, Gómez-Soriano J, Bravo-Esteban E, Taylor J. Spinal cord compression injury in lysophosphatidic acid 1 receptor-null mice promotes maladaptive pronociceptive descending control. Eur J Pain 2015; 20:176-85. [PMID: 25820316 DOI: 10.1002/ejp.695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2015] [Indexed: 11/11/2022]
Abstract
BACKGROUND Although activation of the lysophosphatidic acid receptor 1 (LPA1) is known to mediate pronociceptive effects in peripheral pain models, the role of this receptor in the modulation of spinal nociception following spinal cord injury (SCI) is unknown. AIM In this study, LPA1 regulation of spinal excitability mediated by supraspinal descending antinociceptive control systems was assessed following SCI in both wild-type (WT) and maLPA1-null receptor mice. METHODS The effect of a T8 spinal compression in WT and maLPA1-null mice was assessed up to 1 month after SCI using histological, immunohistochemical and behavioural techniques analysis including electrophysiological recording of noxious toes-Tibialis Anterior (TA) stimulus-response reflex activity. The effect of a T3 paraspinal transcutaneous electrical conditioning stimulus on TA noxious reflex temporal summation was also assessed. RESULTS Histological analysis demonstrated greater dorsolateral funiculus damage after SCI in maLPA1-null mice, without a change in the stimulus-response function of the TA noxious reflex when compared to WT mice. While T3 conditioning stimulation in the WT group inhibited noxious TA reflex temporal summation after SCI, this stimulus strongly excited TA reflex temporal summation in maLPA1-null mice. The functional switch from descending inhibition to maladaptive facilitation of central excitability of spinal nociception demonstrated in maLPA1-null mice after SCI was unrelated to a general change in reflex activity. CONCLUSIONS These data suggest that the LPA1 receptor is necessary for inhibition of temporal summation of noxious reflex activity, partly mediated via long-tract descending modulatory systems acting at the spinal level.
Collapse
Affiliation(s)
- M Suardíaz
- Unidad de Gestión Clínica Intercentros de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Málaga, Spain
| | - I Galan-Arriero
- Sensorimotor Function Group, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - G Avila-Martin
- Sensorimotor Function Group, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - G Estivill-Torrús
- Unidad de Gestión Clínica Intercentros de Neurociencias, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Málaga, Spain
| | - F R de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospitales Universitarios Regional de Malaga y Virgen de la Victoria, Malaga, Spain
| | - J Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Centre The Scripps Research Institute, La Jolla, USA
| | - J Gómez-Soriano
- Grupo de Investigación en Fisioterapia Toledo (GIFTO), E.U.E. Fisioterapia de Toledo, Universidad de Castilla la Mancha, USA
| | - E Bravo-Esteban
- Neurorehabilitation Group, Instituto Cajal, Council for Scientific Research (CSIC), Madrid, Spain
| | - J Taylor
- Sensorimotor Function Group, Hospital Nacional de Parapléjicos, Toledo, Spain.,Stoke Mandeville Spinal Research, National Spinal Injuries Centre, Aylesbury, UK
| |
Collapse
|
10
|
Rossaneis AC, Prado WA. The ventral portion of the anterior pretectal nucleus controls descending mechanisms that initiate neuropathic pain in rats. Clin Exp Pharmacol Physiol 2015; 42:704-10. [PMID: 25809943 DOI: 10.1111/1440-1681.12391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 02/27/2015] [Accepted: 03/19/2015] [Indexed: 11/30/2022]
Abstract
Stimulating the dorsal anterior pretectal nucleus (dAPtN) in rats is more effective than stimulating the ventral APtN (vAPtN) at reducing tail-flick latency, whereas stimulation of the vAPtN is more effective at reducing postoperative pain behaviour. This study examines whether a cell lesion caused by injecting N-methyl-D-aspartate into the dAPtN or vAPtN changes the withdrawal threshold of a rat hind paw during different phases of the tactile hypersensitivity induced by a chronic constriction injury (CCI) of the contralateral sciatic nerve. The number of Fos immunoreactive cells in the APtN was also evaluated. The rats whose vAPtN was lesioned 2 days before CCI had more intense tactile hypersensitivity 2 days after CCI than that of the control group, but the groups were not different 7 days after the CCI. The rats whose vAPtN was lesioned 5 days after CCI had withdrawal thresholds that did not differ significantly 7 days after the CCI. The tactile hypersensitivity of the rats whose dAPtN was lesioned 2 days before or 5 days after CCI was not different from that of the control on the second and seventh days after the CCI. The number of Fos immunoreactive cells in the vAPtN and dAPtN increased 2 days after CCI, but did not differ from that in the control 7 days after CCI. We conclude that vAPtN and dAPtN cells are activated by nerve injury; the vAPtN exerts inhibitory control of the initial phase of neuropathic pain whereas the dAPtN does not appear to exert an inhibitory effect in neuropathic processing.
Collapse
Affiliation(s)
- Ana C Rossaneis
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Wiliam A Prado
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
11
|
Uchytilova E, Spicarova D, Palecek J. Single high-concentration capsaicin application prevents c-Fos expression in spinothalamic and postsynaptic dorsal column neurons after surgical incision. Eur J Pain 2015; 19:1496-505. [DOI: 10.1002/ejp.683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2015] [Indexed: 12/30/2022]
Affiliation(s)
- E. Uchytilova
- Department of Functional Morphology; Institute of Physiology; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - D. Spicarova
- Department of Functional Morphology; Institute of Physiology; Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - J. Palecek
- Department of Functional Morphology; Institute of Physiology; Academy of Sciences of the Czech Republic; Prague Czech Republic
| |
Collapse
|
12
|
Pineda-Farias JB, Velázquez-Lagunas I, Barragán-Iglesias P, Cervantes-Durán C, Granados-Soto V. 5-HT 2B Receptor Antagonists Reduce Nerve Injury-Induced Tactile Allodynia and Expression of 5-HT 2B Receptors. Drug Dev Res 2015; 76:31-39. [PMID: 25620128 DOI: 10.1002/ddr.21238] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/10/2014] [Indexed: 01/01/2023]
Abstract
Preclinical Research This work was performed to assess the effects of intrathecal serotonin 2B (5-HT2B ) receptor antagonists in rats with neuropathic pain. With RS-127445, its effect was also determined on 5-HT2B receptor expression. Neuropathic pain was induced by L5/L6 spinal nerve ligation. Western blotting was used to determine 5-HT2B receptor expression. Dose-response curves with the 5-HT2B receptor antagonists 2-amino-4-(4-fluoronaphth-1-yl)-6-isopropylpyridine (RS-127445, 1-100 nmol) and 1-[(2-chloro-3,4-dimethoxyphenyl)methyl]-2,3,4,9-tetrahydro-6-methyl-1H-pyrido[3,4-b]indole hydrochloride (LY-266097, 1-100 nmol) were performed in rats. Tactile allodynia of the left hind paw (ipsilateral) was assessed for 8 h after compound administration. Intrathecal injection of the 5-HT2B receptor antagonists RS-127445 and LY-266097 diminished spinal nerve ligation-induced allodynia. In contrast, intrathecal injection of the 5-HT2 receptor agonist (±)-2,5-dimethoxy-4-iodoamphetamine hydrochloride (DOI, 10 nmol) did not modify tactile allodynia induced by nerve ligation. L5/L6 nerve ligation increased expression of the 5-HT2B receptors in the ipsilateral, but not contralateral, dorsal root ganglia. Furthermore, nerve injury also enhanced 5-HT2B receptor expression in the ipsilateral dorsal part of the spinal cord. Intrathecal treatment with RS-127445 (100 nmol) diminished spinal nerve injury-induced increased expression of 5-HT2B receptors in dorsal root ganglia and spinal cord. Our results imply that spinal 5-HT2B receptors are present on sites related to nociception and participate in neuropathic pain. © 2014 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jorge B Pineda-Farias
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Sede Sur. México, D.F., México
| | | | | | | | | |
Collapse
|
13
|
Saadé NE, Barchini J, Tchachaghian S, Chamaa F, Jabbur SJ, Song Z, Meyerson BA, Linderoth B. The role of the dorsolateral funiculi in the pain relieving effect of spinal cord stimulation: a study in a rat model of neuropathic pain. Exp Brain Res 2014; 233:1041-52. [PMID: 25537469 DOI: 10.1007/s00221-014-4180-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/09/2014] [Indexed: 02/06/2023]
Abstract
Activation of the dorsal columns is relayed to supraspinal centers, involved in pain modulation, probably via the descending fibers in the dorsolateral funiculi (DLF). The present study examines the role of the DLF in the attenuation of pain-related signs by spinal cord stimulation (SCS). Several groups of rats were subjected to nerve injury and to chronic bilateral DLF lesions at C5-7 level. In each animal, two sets of miniature electrodes were implanted, a caudal system placed in the dorsal epidural space at low thoracic level and another implanted over the dorsal column nuclei, rostral to the lesions. Stimulation (50 Hz, 0.2 ms; 70 % of motor threshold) was applied for 5 min via either of the electrodes. Behavioral tests were used to assess the effects of SCS on the nerve injury-induced mechanical and cold hypersensitivity and heat hyperalgesia. Prior to application of SCS, antagonists to either of GABAA or B, 5-HT1 or 1-2 or α/β-adrenergic receptors were injected i.p. Both stimulations produced comparable decreases (80-90 % of the control) of neuropathic manifestations in rats with intact spinal cords. DLF lesions attenuated the effects of both types of stimulation by about 50 %. Pretreatment with receptor antagonists differentially counteracted the effects of rostral and caudal stimulation; the inhibition with rostral stimulation generally being more prominently influenced. These results provide further support to the notion of important involvement of brainstem pain modulating centers in the effects of SCS. A major component of the inhibitory spinal-supraspinal-spinal loop is mediated by fibers running in the DLF.
Collapse
Affiliation(s)
- N E Saadé
- Department of Anatomy, Cell Biology and Physiology, American University of Beirut, Riad El Solh Beirut, Beirut, 1107-2020, Lebanon,
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Rossaneis A, Genaro K, Dias Q, Guethe L, Fais R, Del Bel E, Prado W. Descending mechanisms activated by the anterior pretectal nucleus initiate but do not maintain neuropathic pain in rats. Eur J Pain 2014; 19:1148-57. [DOI: 10.1002/ejp.638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2014] [Indexed: 11/06/2022]
Affiliation(s)
- A.C. Rossaneis
- Department of Pharmacology; Ribeirão Preto Medical School; University of São Paulo; Ribeirão Preto Brazil
| | - K. Genaro
- Department of Pharmacology; Ribeirão Preto Medical School; University of São Paulo; Ribeirão Preto Brazil
| | - Q.M. Dias
- Oswaldo Cruz Foundation; Fiocruz Rondônia; Brazil
| | - L.M. Guethe
- Department of Pharmacology; Ribeirão Preto Medical School; University of São Paulo; Ribeirão Preto Brazil
| | - R.S. Fais
- Department of Pharmacology; Ribeirão Preto Medical School; University of São Paulo; Ribeirão Preto Brazil
| | - E.A. Del Bel
- Department of Morphology, Estomatology and Physiology; Faculty of Odontology of Ribeirão Preto; University of São Paulo; Ribeirão Preto Brazil
| | - W.A. Prado
- Department of Pharmacology; Ribeirão Preto Medical School; University of São Paulo; Ribeirão Preto Brazil
| |
Collapse
|
15
|
Garcia-Larrea L, Peyron R. Pain matrices and neuropathic pain matrices: A review. Pain 2013; 154 Suppl 1:S29-S43. [PMID: 24021862 DOI: 10.1016/j.pain.2013.09.001] [Citation(s) in RCA: 305] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 08/19/2013] [Accepted: 09/02/2013] [Indexed: 01/18/2023]
|
16
|
Aira Z, Buesa I, García del Caño G, Bilbao J, Doñate F, Zimmermann M, Azkue JJ. Transient, 5-HT2B receptor–mediated facilitation in neuropathic pain: Up-regulation of PKCγ and engagement of the NMDA receptor in dorsal horn neurons. Pain 2013; 154:1865-1877. [DOI: 10.1016/j.pain.2013.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 12/22/2022]
|
17
|
The rostroventromedial medulla is engaged in the effects of spinal cord stimulation in a rodent model of neuropathic pain. Neuroscience 2013; 247:134-44. [PMID: 23711584 DOI: 10.1016/j.neuroscience.2013.05.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 11/21/2022]
Abstract
The neurobiological mechanisms underlying the suppression of neuropathic pain by spinal cord stimulation (SCS) are still incompletely known. The present study aims at exploring whether the descending pain control system in the rostroventromedial medulla (RVM) exerts a role in the attenuation of neuropathic pain by SCS. Experiments were performed in the rat spared nerve injury (SNI) pain model. The effects of SCS on neuronal activity of pronociceptive ON-like, antinociceptive OFF-like, and neutral cells, including 5-HT-like cells, in the RVM were analyzed in SCS responding and SCS non-responding SNI animals as well as in naïve controls. Decreased spontaneous activities in OFF-like cells and increased spontaneous activities in ON-like cells were observed in SNI animals, whereas the spontaneous activities of 5-HT-like and neutral cells were unchanged. SCS produced a prominent increase in the discharge of OFF- and 5-HT-like cells in SCS responding, but not in non-responding SNI animals or controls. Discharge rates of ON-like and neutral cell were not affected by SCS. In awake SNI animals, microinjection of a GABAA receptor agonist, muscimol, into the RVM significantly attenuated the antihypersensitivity effect induced by SCS while a non-selective opioid receptor antagonist, naltrexone, was ineffective. It is concluded that SCS may shift the reciprocal inhibitory and facilitatory pain modulation balance controlled by the RVM in favor of inhibition. This increase in the descending antinociceptive effect operates in concert with segmental spinal mechanisms in producing pain relief.
Collapse
|
18
|
Pitcher GM, Ritchie J, Henry JL. Peripheral neuropathy induces cutaneous hypersensitivity in chronically spinalized rats. PAIN MEDICINE 2013; 14:1057-71. [PMID: 23855791 DOI: 10.1111/pme.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES The present study was aimed at the issue of whether peripheral nerve injury-induced chronic pain is maintained by supraspinal structures governing descending facilitation to the spinal dorsal horn, or whether altered peripheral nociceptive mechanisms sustain central hyperexcitability and, in turn, neuropathic pain. We examined this question by determining the contribution of peripheral/spinal mechanisms, isolated from supraspinal influence(s), in cutaneous hypersensitivity in an animal model of peripheral neuropathy. METHODS Adult rats were spinalized at T8-T9; 8 days later, peripheral neuropathy was induced by implanting a 2-mm polyethylene cuff around the left sciatic nerve. Hind paw withdrawal responses to mechanical or thermal plantar stimulation were evaluated using von Frey filaments or a heat lamp, respectively. RESULTS Spinalized rats without cuff implantation exhibited a moderate decrease in mechanical withdrawal threshold on ~day 10 (P < 0.05) and in thermal withdrawal threshold on ~day 18 (P < 0.05). However, cuff-implanted spinalized rats developed a more rapid and significant decrease in mechanical (~day 4; P < 0.001) and thermal (~day 10; P < 0.05) withdrawal thresholds that remained significantly decreased through the duration of the study. CONCLUSIONS Our findings demonstrate an aberrant peripheral/spinal mechanism that induces and maintains thermal and to a greater degree tactile cutaneous hypersensitivity in the cuff model of neuropathic pain, and raise the prospect that altered peripheral/spinal nociceptive mechanisms in humans with peripheral neuropathy may have a pathologically relevant role in both inducing and sustaining neuropathic pain.
Collapse
Affiliation(s)
- Graham M Pitcher
- Departments of Physiology and Psychiatry, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
19
|
Wang R, King T, De Felice M, Guo W, Ossipov MH, Porreca F. Descending facilitation maintains long-term spontaneous neuropathic pain. THE JOURNAL OF PAIN 2013; 14:845-53. [PMID: 23602267 DOI: 10.1016/j.jpain.2013.02.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 02/26/2013] [Accepted: 02/28/2013] [Indexed: 11/25/2022]
Abstract
UNLABELLED Neuropathic pain is frequently characterized by spontaneous pain (ie, pain at rest) and, in some cases, by cold- and touch-induced allodynia. Mechanisms underlying the chronicity of neuropathic pain are not well understood. Rats received spinal nerve ligation (SNL) and were monitored for tactile and thermal thresholds. While heat hypersensitivity returned to baseline levels within approximately 35 to 40 days, tactile hypersensitivity was still present at 580 days after SNL. Tactile hypersensitivity at post-SNL day 60 (D60) was reversed by microinjection of 1) lidocaine; 2) a cholecystokinin 2 receptor antagonist into the rostral ventromedial medulla; or 3) dorsolateral funiculus lesion. Rostral ventromedial medulla lidocaine at D60 or spinal ondansetron, a 5-hydroxytryptamine 3 antagonist, at post-SNL D42 produced conditioned place preference selectively in SNL-treated rats, suggesting long-lasting spontaneous pain. Touch-induced FOS was increased in the spinal dorsal horn of SNL rats at D60 and prevented by prior dorsolateral funiculus lesion, suggesting that long-lasting tactile hypersensitivity depends upon spinal sensitization, which is mediated in part by descending facilitation, in spite of resolution of heat hypersensitivity. PERSPECTIVE These data suggest that spontaneous pain is present for an extended period of time and, consistent with likely actions of clinically effective drugs, is maintained by descending facilitation.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Pharmacology, University of Arizona Health Science Center, Tucson, Arizona 85724, USA
| | | | | | | | | | | |
Collapse
|
20
|
Time-dependent cross talk between spinal serotonin 5-HT2A receptor and mGluR1 subserves spinal hyperexcitability and neuropathic pain after nerve injury. J Neurosci 2012; 32:13568-81. [PMID: 23015446 DOI: 10.1523/jneurosci.1364-12.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Emerging evidence implicates serotonergic descending facilitatory pathways from the brainstem to the spinal cord in the maintenance of pathologic pain. Upregulation of the serotonin receptor 2A (5-HT(2A)R) in dorsal horn neurons promotes spinal hyperexcitation and impairs spinal μ-opioid mechanisms during neuropathic pain. We investigated the involvement of spinal glutamate receptors, including metabotropic receptors (mGluRs) and NMDA, in 5-HT(2A)R-induced hyperexcitability after spinal nerve ligation (SNL) in rat. High-affinity 5-HT(2A)R agonist (4-bromo-3,6-dimethoxybenzocyclobuten-1-yl)methylamine hydrobromide (TCB-2) enhanced C-fiber-evoked dorsal horn potentials after SNL, which was prevented by mGluR1 antagonist AIDA [(RS)-1-aminoindan-1,5-dicarboxylic acid] but not by group II mGluR antagonist LY 341495 [(2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl)propanoic acid] or NMDA antagonist d-AP5 [D-(-)-2-amino-5-phosphonopentanoic acid]. 5-HT(2A)R and mGluR1 were found to be coexpressed in postsynaptic densities in dorsal horn neurons. In the absence of SNL, pharmacological stimulation of 5-HT(2A)R with TCB-2 both induced rapid bilateral upregulation of mGluR1 expression in cytoplasmic and synaptic fractions of spinal cord homogenates, which was attenuated by PKC inhibitor chelerythrine, and enhanced evoked potentials during costimulation of mGluR1 with 3,5-DHPG [(RS)-3,5-dihydroxyphenylglycine]. SNL was followed by bilateral upregulation of mGluR1 in 5-HT(2A)R-containing postsynaptic densities. Upregulation of mGluR1 in synaptic compartments was partially prevented by chronic administration of selective 5-HT(2A)R antagonist M100907 [(R)-(+)-α-(2,3-dimethoxyphenyl)-1-[2-(4-fluorophenyl)ethyl]-4-pipidinemethanol], confirming 5-HT(2A)R-mediated control of mGluR1 upregulation triggered by SNL. Changes in thermal and mechanical pain thresholds following SNL were increasingly reversed over the days after injury by chronic 5-HT(2A)R blockade. These results emphasize a role for 5-HT(2A)R in hyperexcitation and pain after nerve injury and support mGluR1 upregulation as a novel feedforward activation mechanism contributing to 5-HT(2A)R-mediated facilitation.
Collapse
|
21
|
Saadé NE, Al Amin HA, Barchini J, Tchachaghian S, Shamaa F, Jabbur SJ, Atweh SF. Brainstem injection of lidocaine releases the descending pain-inhibitory mechanisms in a rat model of mononeuropathy. Exp Neurol 2012; 237:180-90. [DOI: 10.1016/j.expneurol.2012.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/11/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
|
22
|
Godínez-Chaparro B, López-Santillán FJ, Orduña P, Granados-Soto V. Secondary mechanical allodynia and hyperalgesia depend on descending facilitation mediated by spinal 5-HT₄, 5-HT₆ and 5-HT₇ receptors. Neuroscience 2012; 222:379-91. [PMID: 22796074 DOI: 10.1016/j.neuroscience.2012.07.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/03/2012] [Accepted: 07/03/2012] [Indexed: 12/26/2022]
Abstract
In the present study we determined the role of spinal 5-hydroxytriptamine (5-HT) and 5-HT(4/6/7) receptors in the long-term secondary mechanical allodynia and hyperalgesia induced by formalin in the rat. Formalin produced acute nociceptive behaviors (flinching and licking/lifting) followed by long-term secondary mechanical allodynia and hyperalgesia in both paws. In addition, formalin increased the tissue content of 5-HT in the ipsilateral, but not contralateral, dorsal part of the spinal cord compared to control animals. Intrathecal (i.t.) administration of 5,7-dihydroxytriptamine (5,7-DHT), a serotonergic neurotoxin, diminished tissue 5-HT content in the ipsilateral and contralateral dorsal parts of the spinal cord. Accordingly, i.t. 5,7-DHT prevented formalin-induced secondary allodynia and hyperalgesia in both paws. I.t. pre-treatment (-10 min) with ML-10302 (5-HT(4) agonist), EMD-386088 (5-HT(6) agonist) and LP-12 (5-HT(7) agonist) significantly increased secondary mechanical allodynia and hyperalgesia in both paws. In contrast, i.t. pre-treatment (-20 min) with GR-125487 (5-HT(4) antagonist), SB-258585 (5-HT(6) antagonist) and SB-269970 (5-HT(7) antagonist) significantly prevented formalin-induced long-term effects in both paws. In addition, these antagonists prevented the pro-nociceptive effect of ML-10302, EMD-386088 and LP-12, respectively. The i.t. post-treatment (6 days after formalin injection) with GR-125487, SB-258585 and SB-269970 reversed formalin-induced secondary allodynia and hyperalgesia in both paws. These results suggest that spinal 5-HT, released from the serotonergic projections in response to formalin injection, activates pre- or post-synaptic 5-HT(4/6/7) receptors at the dorsal root ganglion/spinal cord promoting the development and maintenance of secondary allodynia and hyperalgesia.
Collapse
Affiliation(s)
- B Godínez-Chaparro
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Sede Sur, México, DF, Mexico
| | | | | | | |
Collapse
|
23
|
Aira Z, Buesa I, Del Caño GG, Salgueiro M, Mendiable N, Mingo J, Aguilera L, Bilbao J, Azkue JJ. Selective impairment of spinal mu-opioid receptor mechanism by plasticity of serotonergic facilitation mediated by 5-HT2A and 5-HT2B receptors. Pain 2012; 153:1418-1425. [PMID: 22520172 DOI: 10.1016/j.pain.2012.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/19/2012] [Accepted: 03/14/2012] [Indexed: 01/11/2023]
Abstract
Opioid analgesia is compromised by intracellular mediators such as protein kinase C (PKC). The phosphatidylinositol hydrolysis-coupled serotonin receptor 5-HT2 is ideally suited to promote PKC activation. We test the hypothesis that 5-HT2A and 5-HT2B receptors, which have been previously shown to become pro-excitatory after spinal nerve ligation (SNL), can negatively influence the ability of opioids to depress spinal excitation evoked by noxious input. Spinal superfusion with (100 nM) mu-opioid receptor (MOR)-agonist DAMGO significantly depressed C fiber-evoked spinal field potentials. Simultaneous administration of subclinical 5-HT2AR antagonist 4F 4PP (100 nM) or 5-HT2BR antagonist SB 204741 (100 nM) significantly reduced the IC50 value for DAMGO in nerve-ligated rats (97.56 nM ± 1.51 and 1.20 nM ± 1.28 respectively, relative to 104 nM ± 1.08 at the baseline condition), but not in sham-operated rats. Both antagonists failed to alter depression induced by delta-opioid receptor (DOR)-agonist D-ala2-deltorphin II after SNL as well as in the sham condition. Western blot analysis of dorsal horn homogenates revealed bilateral upregulation of 5-HT2AR and 5-HT2BR protein band densities after SNL. As assessed from double immunofluorescence labeling for confocal laser scanning microscopy, scarce dorsal horn cell processes showed co-localization color overlay for 5-HT2AR/MOR, 5-HT2BR/MOR, 5-HT2AR/DOR, or 5-HT2BR/DOR in sham-operated rats. Intensity correlation-based analyses showed significant increases in 5-HT2AR/MOR and 5-HT2BR/MOR co-localizations after SNL. These results indicate that plasticity of spinal serotonergic neurotransmission can selectively reduce spinal MOR mechanisms via 5-HT2A and 5-HT2B receptors, including upregulation of the latter and increased expression in dorsal horn neurons containing MOR.
Collapse
Affiliation(s)
- Zigor Aira
- Department of Neurosciences, School of Medicine and Dentistry, University of the Basque Country, Leioa, Spain Department of Neurosciences, School of Pharmacy, University of the Basque Country, Vitoria-Gasteiz, Spain Department of Surgery, Radiology and Physical Medicine, University of the Basque Country, Bilbao, Spain Department of Preventive Medicine and Public Health, School of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Cell death has been reported in the CNS in models of neuropathic pain (Sugimoto et al., 1990; Whiteside and Munglani, 2001; Scholz et al., 2005; Fuccio et al., 2009). In our present study, we examined the effects of spinal nerve ligation (SNL) on the number of neurons in the rostral ventromedial medulla (RVM), a brainstem region involved in modulation of nociception. In rats receiving SNL, we found that the number of RVM neurons decreased by 23% in the side ipsilateral to the surgery. The loss of RVM neurons was also associated with a bilateral increase in the number of glia as well as bilateral activation of both astrocytes and microglia. Administration of tauroursodeoxycholic acid (TUDCA), which reportedly inhibits apoptosis, significantly reduced the loss of neurons, the increase in glia, and the mechanical hypersensitivity induced by SNL. Among RVM neurons, we found that serotonergic (5-hydroxytryptamine, 5-HT) neurons decreased by 35% ipsilateral to SNL. Consistent with these findings, the density of 5-HT-immunoreactive varicosities in the superficial dorsal horn of the spinal cord was 15-30% lower, ipsilateral to SNL. To test the function of the remaining 5-HT neurons, we administered the 5-HT neurotoxin, 5,7-dihydroxytryptamine (5,7-DHT). Interestingly, after 5,7-DHT, mechanical withdrawal thresholds increased significantly. We conclude that nerve injury induces death of antinociceptive RVM neurons that can be reduced or abolished by TUDCA. We propose that the loss of RVM neurons shifts the balance of descending control from pain inhibition to pain facilitation.
Collapse
|
25
|
Ruscheweyh R, Sandkühler J. Opioids and central sensitisation: II. Induction and reversal of hyperalgesia. Eur J Pain 2012; 9:149-52. [PMID: 15737805 DOI: 10.1016/j.ejpain.2004.05.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 05/17/2004] [Indexed: 11/19/2022]
Abstract
Opioids are powerful analgesics when used to treat acute pain and some forms of chronic pain. In addition, opioids can preempt some forms of central sensitization. Here we review evidence that opioids may also induce and perhaps reverse some forms of central sensitization.
Collapse
Affiliation(s)
- Ruth Ruscheweyh
- Department of Neurophysiology, Centre for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | | |
Collapse
|
26
|
Tessitore A, Russo A, Esposito F, Giordano A, Taglialatela G, De Micco R, Cirillo M, Conte F, d'Onofrio F, Cirillo S, Tedeschi G. Interictal cortical reorganization in episodic migraine without aura: an event-related fMRI study during parametric trigeminal nociceptive stimulation. Neurol Sci 2011; 32 Suppl 1:S165-7. [PMID: 21533737 DOI: 10.1007/s10072-011-0537-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of our study was to explore the pain processing network in patients with migraine during trigeminal nociceptive stimulation. Sixteen patients with episodic migraine without aura and 16 healthy controls performed functional magnetic resonance imaging during thermal stimuli (at 41, 51 and 53°C). Patients with migraine showed a greater activation in the perigenual part of anterior cingulate cortex at 51°C and less activation in the bilateral somatosensory cortex at 53°C compared to healthy controls. There were no differences in experimental pain perception between groups. Our findings demonstrate a functional reorganization of cerebral areas known to be involved in pain processing in patients with migraine.
Collapse
Affiliation(s)
- A Tessitore
- Department of Neurological Sciences, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Engagement of descending inhibition from the rostral ventromedial medulla protects against chronic neuropathic pain. Pain 2011; 152:2701-2709. [PMID: 21745713 DOI: 10.1016/j.pain.2011.06.008] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 05/25/2011] [Accepted: 06/08/2011] [Indexed: 11/23/2022]
Abstract
A puzzling observation is why peripheral nerve injury results in chronic pain in some, but not all, patients. We explored potential mechanisms that may prevent the expression of chronic pain. Sprague Dawley (SD) or Holtzman (HZ) rats showed no differences in baseline sensory thresholds or responses to inflammatory stimuli. However, spinal nerve ligation (SNL)-induced tactile allodynia occurred in approximately 85% of SD and 50% of HZ rats, respectively. No apparent differences were observed in a survey of dorsal root ganglion or spinal neuropathic markers after SNL regardless of allodynic phenotype. SNL-induced allodynia was reversed by administration of lidocaine within the rostral ventromedial medulla (RVM), a site that integrates descending pain modulation via pain inhibitory (ie, OFF) and excitatory (ie, ON) cells. However, in SD or HZ rats with SNL but without allodynia, RVM lidocaine precipitated allodynia. Additionally, RVM lidocaine produced conditioned place preference in allodynic SD or HZ rats but conditioned place aversion in nonallodynic HZ rats. Similarly, RVM U69,593 (kappa opioid agonist) or blockade of spinal α(2) adrenergic receptors precipitated allodynia in previously nonallodynic HZ rats with SNL. All rats showed an equivalent first-phase formalin responses. However, HZ rats had reduced second-phase formalin behaviors along with fewer RVM OFF cell pauses and RVM ON cell bursts. Thus, expression of nerve injury-induced pain may ultimately depend on descending modulation. Engagement of descending inhibition protects in the transition from acute to chronic pain. These unexpected findings might provide a mechanistic explanation for medications that engage descending inhibition or mimic its consequences.
Collapse
|
28
|
Formalin-induced long-term secondary allodynia and hyperalgesia are maintained by descending facilitation. Pharmacol Biochem Behav 2011; 98:417-24. [DOI: 10.1016/j.pbb.2011.02.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 02/14/2011] [Indexed: 12/23/2022]
|
29
|
Mase H, Sakai A, Sakamoto A, Suzuki H. A subset of μ-opioid receptor-expressing cells in the rostral ventromedial medulla contribute to thermal hyperalgesia in experimental neuropathic pain. Neurosci Res 2011; 70:35-43. [DOI: 10.1016/j.neures.2011.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 12/29/2010] [Accepted: 01/05/2011] [Indexed: 01/08/2023]
|
30
|
Ossipov MH, Dussor GO, Porreca F. Central modulation of pain. J Clin Invest 2010; 120:3779-87. [PMID: 21041960 DOI: 10.1172/jci43766] [Citation(s) in RCA: 725] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
It has long been appreciated that the experience of pain is highly variable between individuals. Pain results from activation of sensory receptors specialized to detect actual or impending tissue damage (i.e., nociceptors). However, a direct correlation between activation of nociceptors and the sensory experience of pain is not always apparent. Even in cases in which the severity of injury appears similar, individual pain experiences may vary dramatically. Emotional state, degree of anxiety, attention and distraction, past experiences, memories, and many other factors can either enhance or diminish the pain experience. Here, we review evidence for "top-down" modulatory circuits that profoundly change the sensory experience of pain.
Collapse
Affiliation(s)
- Michael H Ossipov
- Department of Pharmacology, University of Arizona, Tucson, Arizona 85724, USA
| | | | | |
Collapse
|
31
|
Aira Z, Buesa I, Salgueiro M, Bilbao J, Aguilera L, Zimmermann M, Azkue JJ. Subtype-specific changes in 5-HT receptor-mediated modulation of C fibre-evoked spinal field potentials are triggered by peripheral nerve injury. Neuroscience 2010; 168:831-41. [PMID: 20412834 DOI: 10.1016/j.neuroscience.2010.04.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Revised: 03/18/2010] [Accepted: 04/15/2010] [Indexed: 10/19/2022]
Abstract
Neurotransmitter serotonin (5-HT) released from descending pain modulation pathways to the dorsal horn is crucial to spinal nociception processing. This study sought to gain insight into the modulatory roles of specific serotonin receptor subtypes in experimentally induced neuropathic pain. In rats subjected to spinal nerve ligation (SNL) surgery, we recorded field potentials evoked in the spinal dorsal horn by C fibre-input, during spinal superfusion with subtype-selective drugs. In neuropathic rats, subtype 5-HT1A agonist 8-OH-DPAT (100 nM) was found to potently depress evoked field potentials, as opposed to 5-HT2A or 5-HT2B subtype agonists TCB-2 (100 nM) or BW 723C86 (1 microM), respectively, which consistently enhanced evoked potentials. All three failed to alter spinal field potentials in sham operated rats. CP 94253 (1 microM), WAY 161503 (1 mM) or SR 57227 (at 1 microM in SNL rats, and 100 microM in sham rats), selective agonists for 5-HT1B, 5-HT2C and 5-HT3 receptors, respectively, significantly depressed evoked field potentials in both animal groups. The 5-HT4 agonist RS 67333 (1 microM) was depressant only in sham operated animals. Only after SNL, spinal superfusion with 5-HT1A- or 5-HT1B receptor-antagonists (S)-WAY 100135 (100 microM) or SB 224289 (100 microM), respectively, disinhibited C fibre-evoked potentials, whereas 5-HT2A or 5-HT2B receptor-antagonists 4F 4PP (100 microM) or SB 204741 (100 microM) depressed evoked potentials, suggesting tonic activity of all four subtypes as a consequence of experimental nerve injury. The present findings reveal profound subtype-specific changes in the functional modulatory activities of spinal serotonin receptors following peripheral nerve injury. In particular, spinal hyperexcitation promoted by receptors 5-HT2A and 5-HT2B is suggested as a novel pathogenic pathway contributing to neuropathic pain.
Collapse
Affiliation(s)
- Z Aira
- Department of Neurosciences, School of Medicine and Dentistry, Basque Country University, Barrio Sarriena s/n, 48940 Leioa, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Saadé NE, Al Amin H, Tchachaghian S, Jabbur SJ, Atweh SF. Alteration of GABAergic and glycinergic mechanisms by lidocaine injection in the rostral ventromedial medulla of neuropathic rats. Pain 2010; 149:89-99. [DOI: 10.1016/j.pain.2010.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 01/15/2010] [Accepted: 01/15/2010] [Indexed: 11/28/2022]
|
33
|
Song Z, Ultenius C, Meyerson BA, Linderoth B. Pain relief by spinal cord stimulation involves serotonergic mechanisms: An experimental study in a rat model of mononeuropathy. Pain 2009; 147:241-8. [DOI: 10.1016/j.pain.2009.09.020] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 09/15/2009] [Accepted: 09/18/2009] [Indexed: 01/03/2023]
|
34
|
Abstract
The chronicity of pain is the feature of pain that is least understood and most directly linked with our inability to effectively manage pain. Acute pain is relatively responsive to our current pharmacologic and interventional armamentarium. However, as pain persists, our ability to treat effectively diminishes and the patient's frustration and resource utilization increases. This article explores our current understanding of the factors linked to pain duration and the transition from acute to chronic pain in both human and animal models, and across a spectrum of human chronic pain conditions.
Collapse
|
35
|
Curtin C, Carroll I. Cutaneous neuroma physiology and its relationship to chronic pain. J Hand Surg Am 2009; 34:1334-6. [PMID: 19481362 PMCID: PMC2935247 DOI: 10.1016/j.jhsa.2009.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 04/07/2009] [Indexed: 02/02/2023]
Affiliation(s)
- Catherine Curtin
- Division of Plastic Surgery, Stanford University, Palo Alto, CA 94304, USA.
| | | |
Collapse
|
36
|
Sanoja R, Tortorici V, Fernandez C, Price TJ, Cervero F. Role of RVM neurons in capsaicin-evoked visceral nociception and referred hyperalgesia. Eur J Pain 2009; 14:120.e1-9. [PMID: 19443247 DOI: 10.1016/j.ejpain.2009.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 04/07/2009] [Accepted: 04/07/2009] [Indexed: 12/11/2022]
Abstract
Most forms of visceral pain generate intense referred hyperalgesia but the mechanisms of this enhanced visceral hypersensitivity are not known. The on-cells of the rostral ventromedial medulla (RVM) play an important role in descending nociceptive facilitation and can be sensitized to somatic mechanical stimulation following peripheral nerve injury or hindpaw inflammation. Here we have tested the hypothesis that visceral noxious stimulation sensitizes RVM ON-like cells, thus promoting an enhanced descending facilitation that can lead to referred visceral hyperalgesia. Intracolonic capsaicin instillation (ICI) was applied to rats in order to create a hyperalgesic state dependent on noxious visceral stimulation. This instillation produced acute pain-related behaviors and prolonged referred hyperalgesia that were prevented by the RVM microinjection of AP5, an NMDA selective antagonist. In electrophysiological experiments, ON-like RVM neurons showed ongoing spontaneous activity following ICI that lasted for approximately 20 min and an enhanced responsiveness to von Frey and heat stimulation of the hindpaw and to colorectal distention (CRD) that lasted for at least 50 min post capsaicin administration. Moreover, ON-like cells acquired a novel response to CRD and responded to heat stimulation in the innocuous range. OFF-like neurons responded to capsaicin administration with a brief (<5 min) inhibition of activity followed by an enhanced inhibition to noxious stimulation and a novel inhibition to innocuous stimulation (CRD and heat) at early time points (10 min post capsaicin). These results support the hypothesis that noxious visceral stimulation may cause referred hypersensitivity by promoting long-lasting sensitization of RVM ON-like cells.
Collapse
Affiliation(s)
- Raul Sanoja
- Instituto Venezolano de Investigaciones Cientificas (IVIC), Apartado 20632, Caracas 1020A, Venezuela
| | | | | | | | | |
Collapse
|
37
|
Fairbanks CA, Stone LS, Wilcox GL. Pharmacological profiles of alpha 2 adrenergic receptor agonists identified using genetically altered mice and isobolographic analysis. Pharmacol Ther 2009; 123:224-38. [PMID: 19393691 DOI: 10.1016/j.pharmthera.2009.04.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 04/09/2009] [Indexed: 12/24/2022]
Abstract
Endogenous, descending noradrenergic fibers impose analgesic control over spinal afferent circuitry mediating the rostrad transmission of pain signals. These fibers target alpha 2 adrenergic receptors (alpha(2)ARs) on both primary afferent terminals and secondary neurons, and their activation mediates substantial inhibitory control over this transmission, rivaling that of opioid receptors which share a similar pattern of distribution. The terminals of primary afferent nociceptive neurons and secondary spinal dorsal horn neurons express alpha(2A)AR and alpha(2C)AR subtypes, respectively. Spinal delivery of these agents serves to reduce their side effects, which are mediated largely at supraspinal sites, by concentrating the drugs at the spinal level. Targeting these spinal alpha(2)ARs with one of five selective therapeutic agonists, clonidine, dexmedetomidine, brimonidine, ST91 and moxonidine, produces significant antinociception that can work in concert with opioid agonists to yield synergistic antinociception. Application of several genetically altered mouse lines had facilitated identification of the primary receptor subtypes that likely mediate the antinociceptive effects of these agents. This review provides first an anatomical description of the localization of the three subtypes in the central nervous system, second a detailed account of the pharmacological history of each of the six primary agonists, and finally a comprehensive report of the specific interactions of other GPCR agonists with each of the six principal alpha(2)AR agonists featured.
Collapse
Affiliation(s)
- Carolyn A Fairbanks
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
38
|
|
39
|
Moulton EA, Burstein R, Tully S, Hargreaves R, Becerra L, Borsook D. Interictal dysfunction of a brainstem descending modulatory center in migraine patients. PLoS One 2008; 3:e3799. [PMID: 19030105 PMCID: PMC2582961 DOI: 10.1371/journal.pone.0003799] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 11/06/2008] [Indexed: 01/13/2023] Open
Abstract
Background The brainstem contains descending circuitry that can modulate nociceptive processing (neural signals associated with pain) in the dorsal horn of the spinal cord and the medullary dorsal horn. In migraineurs, abnormal brainstem function during attacks suggest that dysfunction of descending modulation may facilitate migraine attacks, either by reducing descending inhibition or increasing facilitation. To determine whether a brainstem dysfunction could play a role in facilitating migraine attacks, we measured brainstem function in migraineurs when they were not having an attack (i.e. the interictal phase). Methods and Findings Using fMRI (functional magnetic resonance imaging), we mapped brainstem activity to heat stimuli in 12 episodic migraine patients during the interictal phase. Separate scans were collected to measure responses to 41°C and noxious heat (pain threshold+1°C). Stimuli were either applied to the forehead on the affected side (as reported during an attack) or the dorsum of the hand. This was repeated in 12 age-gender-matched control subjects, and the side tested corresponded to that in the matched migraine patients. Nucleus cuneiformis (NCF), a component of brainstem pain modulatory circuits, appears to be hypofunctional in migraineurs. 3 out of the 4 thermal stimulus conditions showed significantly greater NCF activation in control subjects than the migraine patients. Conclusions Altered descending modulation has been postulated to contribute to migraine, leading to loss of inhibition or enhanced facilitation resulting in hyperexcitability of trigeminovascular neurons. NCF function could potentially serve as a diagnostic measure in migraine patients, even when not experiencing an attack. This has important implications for the evaluation of therapies for migraine.
Collapse
Affiliation(s)
- Eric A. Moulton
- P.A.I.N. Group, Brain Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rami Burstein
- Anaesthesia & Critical Care, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Shannon Tully
- P.A.I.N. Group, Brain Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Richard Hargreaves
- Imaging, Merck & Co. Inc., West Point, Pennsylvania, United States of America
| | - Lino Becerra
- P.A.I.N. Group, Brain Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Athinoula Martinos Center for Bioengineering, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - David Borsook
- P.A.I.N. Group, Brain Imaging Center, McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Radiology, Athinoula Martinos Center for Bioengineering, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
40
|
Governing role of primary afferent drive in increased excitation of spinal nociceptive neurons in a model of sciatic neuropathy. Exp Neurol 2008; 214:219-28. [PMID: 18773893 DOI: 10.1016/j.expneurol.2008.08.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 07/31/2008] [Accepted: 08/10/2008] [Indexed: 01/07/2023]
Abstract
Previously we reported that the cuff model of peripheral neuropathy, in which a 2 mm polyethylene tube is implanted around the sciatic nerve, exhibits aspects of neuropathic pain behavior in rats similar to those in humans and causes robust hyperexcitation of spinal nociceptive dorsal horn neurons. The mechanisms mediating this increased excitation are not known and remain a key unresolved question in models of peripheral neuropathy. In anesthetized adult male Sprague-Dawley rats 2-6 weeks after cuff implantation we found that elevated discharge rate of single lumbar (L(3-4)) wide dynamic range (WDR) neurons persists despite acute spinal transection (T9) but is reversed by local conduction block of the cuff-implanted sciatic nerve; lidocaine applied distal to the cuff (i.e. between the cuff and the cutaneous receptive field) decreased spontaneous baseline discharge of WDR dorsal horn neurons approximately 40% (n=18) and when applied subsequently proximal to the cuff, i.e. between the cuff and the spinal cord, it further reduced spontaneous discharge by approximately 60% (n=19; P<0.05 proximal vs. distal) to a level that was not significantly different from that of naive rats. Furthermore, in cuff-implanted rats WDR neurons (n=5) responded to mechanical cutaneous stimulation with an exaggerated afterdischarge which was reversed entirely by proximal nerve conduction block. These results demonstrate that the hyperexcited state of spinal dorsal horn neurons observed in this model of peripheral neuropathy is not maintained by tonic descending facilitatory mechanisms. Rather, on-going afferent discharges originating from the sciatic nerve distal to, at, and proximal to the cuff maintain the synaptically-mediated gain in discharge of spinal dorsal horn WDR neurons and hyperresponsiveness of these neurons to cutaneous stimulation. Our findings reveal that ectopic afferent activity from multiple regions along peripheral nerves may drive CNS changes and the symptoms of pain associated with peripheral neuropathy.
Collapse
|
41
|
Affiliation(s)
- William J. Martin
- Department of Pharmacology, Merck Research Laboratories, Rahway, New Jersey
| |
Collapse
|
42
|
Nociceptive behavior in animal models for peripheral neuropathy: spinal and supraspinal mechanisms. Prog Neurobiol 2008; 86:22-47. [PMID: 18602968 DOI: 10.1016/j.pneurobio.2008.06.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Revised: 04/08/2008] [Accepted: 06/11/2008] [Indexed: 02/01/2023]
Abstract
Since the initial description by Wall [Wall, P.D., 1967. The laminar organization of dorsal horn and effects of descending impulses. J. Neurophysiol. 188, 403-423] of tonic descending inhibitory control of dorsal horn neurons, several studies have aimed to characterize the role of various brain centers in the control of nociceptive input to the spinal cord. The role of brainstem centers in pain inhibition has been well documented over the past four decades. Lesion to peripheral nerves results in hypersensitivity to mild tactile or cold stimuli (allodynia) and exaggerated response to nociceptive stimuli (hyperalgesia), both considered as cardinal signs of neuropathic pain. The increased interest in animal models for peripheral neuropathy has raised several questions concerning the rostral conduction of the neuropathic manifestations and the role of supraspinal centers, especially brainstem, in the inhibitory control or in the abnormal contribution to the maintenance and facilitation of neuropathic-like behavior. This review aims to summarize the data on the ascending and descending modulation of neuropathic manifestations and discusses the recent experimental data on the role of supraspinal centers in the control of neuropathic pain. In particular, the review emphasizes the importance of the reciprocal interconnections between the analgesic areas of the brainstem and the pain-related areas of the forebrain. The latter includes the cerebral limbic areas, the prefrontal cortex, the intralaminar thalamus and the hypothalamus and play a critical role in the control of pain considered as part of an integrated behavior related to emotions and various homeostatic regulations. We finally speculate that neuropathic pain, like extrapyramidal motor syndromes, reflects a disorder in the processing of somatosensory information.
Collapse
|
43
|
Garcia-Larrea L, Magnin M. Physiopathologie de la douleur neuropathique : revue des modèles expérimentaux et des mécanismes proposés. Presse Med 2008; 37:315-40. [DOI: 10.1016/j.lpm.2007.07.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Accepted: 07/02/2007] [Indexed: 01/22/2023] Open
|
44
|
Saadé NE, Al Amin H, Chalouhi S, Baki SA, Jabbur SJ, Atweh SF. Spinal pathways involved in supraspinal modulation of neuropathic manifestations in rats. Pain 2006; 126:280-93. [PMID: 16945485 DOI: 10.1016/j.pain.2006.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Revised: 06/21/2006] [Accepted: 07/17/2006] [Indexed: 11/22/2022]
Abstract
Controversial results have been recently reported on the role of supraspinal centers in the modulation of nociceptive behavior in animal models of mononeuropathy. Our aim was to investigate the role of the various spinal pathways in the modulation of the neuropathic manifestations. Several groups of rats were subjected to selective spinal-tract lesions, either 2-3 weeks before or 2-3 weeks after the induction of mononeuropathy following the chronic constriction injury (CCI) or the spared nerve injury (SNI) models. Tactile and cold allodynias were assessed by Von Frey filaments and the acetone drops test, respectively. Thermal hyperalgesia was assessed by the paw withdrawal and the hot plate tests. The effects of unilateral and bilateral lesions of the dorso-lateral funiculus (DLF), the anterolateral column (ALC) or hemisection were tested over a period of 4-8 weeks. All spinal tract lesions produced reversible, but significant decrease of allodynia and hyperalgesia over a period of 1-3 weeks. The most pronounced effects were observed with bilateral lesions. The stronger attenuation was observed on thermal hyperalgesia, assessed by the paw withdrawal test, while cold allodynia was the least affected. Spinal lesions performed before the induction of neuropathy did not produce significant alterations in the temporal development of neuropathic manifestations. The present results allow the conclusion that all spinal tracts can be involved in the rostral transmission and the descending modulation of neuropathic manifestations. The recovery of symptoms following spinal lesions provides illustration on the plasticity of the neural network involved in the processing of the neuropathic syndromes.
Collapse
Affiliation(s)
- Nayef E Saadé
- Department of Human Morphology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | | | | | | | | | | |
Collapse
|
45
|
Vera-Portocarrero LP, Zhang ET, Ossipov MH, Xie JY, King T, Lai J, Porreca F. Descending facilitation from the rostral ventromedial medulla maintains nerve injury-induced central sensitization. Neuroscience 2006; 140:1311-20. [PMID: 16650614 DOI: 10.1016/j.neuroscience.2006.03.016] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 03/06/2006] [Accepted: 03/12/2006] [Indexed: 11/20/2022]
Abstract
Nerve injury can produce hypersensitivity to noxious and normally innocuous stimulation. Injury-induced central (i.e. spinal) sensitization is thought to arise from enhanced afferent input to the spinal cord and to be critical for expression of behavioral hypersensitivity. Descending facilitatory influences from the rostral ventromedial medulla have been suggested to also be critical for the maintenance, though not the initiation, of experimental neuropathic pain. The possibility that descending facilitation from the rostral ventromedial medulla is required for the maintenance of central sensitization was examined by determining whether ablation of mu-opioid receptor-expressing cells within the rostral ventromedial medulla prevented the enhanced expression of repetitive touch-evoked FOS within the spinal cord of animals with spinal nerve ligation injury as well as nerve injury-induced behavioral hypersensitivity. Rats received a single microinjection of vehicle, saporin, dermorphin or dermorphin-saporin into the rostral ventromedial medulla and 28 days later, underwent either sham or spinal nerve ligation procedures. Animals receiving rostral ventromedial medulla pretreatment with vehicle, dermorphin or saporin that were subjected to spinal nerve ligation demonstrated both thermal and tactile hypersensitivity, and showed significantly increased expression of touch-evoked FOS in the dorsal horn ipsilateral to nerve injury compared with sham-operated controls at days 3, 5 or 10 post-spinal nerve ligation. In contrast, nerve-injured animals pretreated with dermorphin-saporin showed enhanced behaviors and touch-evoked FOS expression in the spinal dorsal horn at day 3, but not days 5 and 10, post-spinal nerve ligation when compared with sham-operated controls. These results indicate the presence of nerve injury-induced behavioral hypersensitivity associated with nerve injury-induced central sensitization. Further, the results demonstrate the novel concept that once initiated, maintenance of nerve injury-induced central sensitization in the spinal dorsal horn requires descending pain facilitation mechanisms arising from the rostral ventromedial medulla.
Collapse
Affiliation(s)
- L P Vera-Portocarrero
- Department of Pharmacology, University of Arizona, Health Sciences Center, 1501 North Campbell Avenue, Tucson, AZ 85724, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Rahman W, Suzuki R, Webber M, Hunt SP, Dickenson AH. Depletion of endogenous spinal 5-HT attenuates the behavioural hypersensitivity to mechanical and cooling stimuli induced by spinal nerve ligation. Pain 2006; 123:264-274. [PMID: 16644129 DOI: 10.1016/j.pain.2006.02.033] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2005] [Revised: 02/15/2006] [Accepted: 02/27/2006] [Indexed: 11/15/2022]
Abstract
There is compelling evidence for a strong facilitatory drive modulating spinal nociceptive transmission. This is in part via serotonergic pathways and originates from the rostroventral medulla. We previously demonstrated that neuropathic pain is associated with an enhanced descending facilitatory drive onto the mechanical evoked responses of dorsal horn neurones, mediated by 5-HT acting at spinal 5-HT3 receptors. Furthermore, depletion of spinal 5-HT has been shown to reduce the at-level mechanical allodynia that follows spinal cord injury. To further clarify the role and direction of effect of endogenous 5-HT, we investigated the effects of depleting spinal 5-HT, via intrathecal injection of 5,7di-hydroxytryptamine (5,7DHT), on pain behaviours after spinal nerve ligation (SNL). Depletion of spinal 5-HT in normal animals leads to reductions in mechanical and thermal evoked responses of deep dorsal horn neurones implying that spinal 5-HT has a predominant facilitatory function. After nerve injury, the frequency of paw withdrawals to low intensity mechanical and cooling stimulation of the ipsilateral hindpaw in the SNL-5,7DHT group was significantly attenuated when compared with the SNL-saline group from day seven post-nerve injury. Sham-5,7DHT and sham-saline animals showed very little response sensitivity on either hindpaw. This 5-HT-mediated difference in behaviour was independent of both the up-regulation of the NK1 receptor and spinal microglial activation produced by nerve injury. These data suggest that supraspinal serotonergic influences under these conditions are facilitatory and are implicated in the maintenance of spinal cord neuronal events leading to the behavioural hypersensitivity manifested after peripheral nerve damage.
Collapse
Affiliation(s)
- Wahida Rahman
- Department of Pharmacology, University College London, Gower Street, London WC1E 6BT, UK Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
47
|
de Leeuw R, Albuquerque R, Okeson J, Carlson C. The contribution of neuroimaging techniques to the understanding of supraspinal pain circuits: implications for orofacial pain. ACTA ACUST UNITED AC 2006; 100:308-14. [PMID: 16122658 DOI: 10.1016/j.tripleo.2004.11.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Revised: 10/29/2004] [Accepted: 11/09/2004] [Indexed: 11/22/2022]
Abstract
The aim of this article was to give an overview of the current knowledge of supraspinal pain mechanisms derived from neuroimaging studies, and to present data related to chronic orofacial pain disorders. The available studies implied that the anterior cingulate cortex plays a role in the emotional-affective component of pain, as well as in pain-related attention and anxiety. The somatosensory cortices may be involved in encoding spatial, temporal, and intensity aspects of noxious input. The insula may mediate both affective and sensory-discriminative aspects of the pain experience. The thalamus appears to be a multifunctional relay system. The prefrontal cortex has been implied in the pain-related attention processing; it does not have intensity encoding properties. Chronic pain conditions were associated with increased activity in the somatosensory cortices, anterior cingulate cortex, and the prefrontal cortex, and with decreased activity in the thalamus. Few neuroimaging studies used experimental stimuli to the trigeminal system or included orofacial pain patients. However, the available studies appeared to be in agreement with those using stimuli to other body parts and those concerning other chronic pain conditions. Overall, the available data suggest that chronic (orofacial) pain states may be related to a dysfunctional brain network and may involve a compromised descending inhibitory control system. The somatosensory cortices, anterior cingulate cortex, thalamus, and prefrontal cortex may play a vital role in the pathophysiology of chronic pain and should be the main focus of future neuroimaging studies in chronic pain patients.
Collapse
Affiliation(s)
- Reny de Leeuw
- Orofacial Pain Center, University of Kentucky, Lexington, KY 40536-0297, USA.
| | | | | | | |
Collapse
|
48
|
|
49
|
Ossipov MH, Porreca F. Chapter 14 Descending excitatory systems. HANDBOOK OF CLINICAL NEUROLOGY 2006; 81:193-210. [PMID: 18808836 DOI: 10.1016/s0072-9752(06)80018-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Vanegas H, Schaible HG. Descending control of persistent pain: inhibitory or facilitatory? ACTA ACUST UNITED AC 2005; 46:295-309. [PMID: 15571771 DOI: 10.1016/j.brainresrev.2004.07.004] [Citation(s) in RCA: 351] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2004] [Indexed: 11/30/2022]
Abstract
The periaqueductal gray matter (PAG) and the nucleus raphe magnus and adjacent structures of the rostral ventromedial medulla (RVM), with their projections to the spinal dorsal horn, constitute the "efferent channel" of a pain-control system that "descends" from the brain onto the spinal cord. Considerable evidence has recently emerged regarding participation of this system in persistent pain conditions such as inflammation and neuropathy. Herein, this evidence is reviewed and organized to support the idea that persistent nociception simultaneously triggers descending facilitation and inhibition. In models of inflammation, descending inhibition predominates over facilitation in pain circuits with input from the inflamed tissue, and thus attenuates primary hyperalgesia, while descending facilitation predominates over inhibition in pain circuits with input from neighboring tissues, and thus facilitates secondary hyperalgesia. Both descending facilitation and inhibition mainly stem from RVM. The formalin-induced primary hyperalgesia, although considered a model for inflammation, is mainly facilitated from RVM. Also, formalin-induced secondary hyperalgesia is facilitated by RVM. Again, formalin triggers a concomitant but concealed descending inhibition. The (primary) hyperalgesia and allodynia of the neuropathic syndrome are also facilitated from RVM. Simultaneously, there is an inhibition of secondary neuronal pools that is partly supported from the PAG. Because in all these models of peripheral damage descending facilitation and inhibition are triggered simultaneously, it will be important to elucidate why inhibition predominates in some neuronal pools and facilitation in others. Therapies that enhance descending inhibition and/or attenuate descending facilitation are furthermore an important target for research in the future.
Collapse
Affiliation(s)
- Horacio Vanegas
- Instituto Venezolano de Investigaciones Cientificas (IVIC), Apartado 21827, Caracas 1020A, Venezuela.
| | | |
Collapse
|