1
|
Dou YN, Liu Y, Ding WQ, Li Q, Zhou H, Li L, Zhao MT, Li ZYQ, Yuan J, Wang XF, Zou WY, Li A, Sun YG. Single-neuron projectome-guided analysis reveals the neural circuit mechanism underlying endogenous opioid antinociception. Natl Sci Rev 2024; 11:nwae195. [PMID: 39045468 PMCID: PMC11264302 DOI: 10.1093/nsr/nwae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/05/2024] [Accepted: 05/24/2024] [Indexed: 07/25/2024] Open
Abstract
Endogenous opioid antinociception is a self-regulatory mechanism that reduces chronic pain, but its underlying circuit mechanism remains largely unknown. Here, we showed that endogenous opioid antinociception required the activation of mu-opioid receptors (MORs) in GABAergic neurons of the central amygdala nucleus (CEA) in a persistent-hyperalgesia mouse model. Pharmacogenetic suppression of these CEAMOR neurons, which mimics the effect of MOR activation, alleviated the persistent hyperalgesia. Furthermore, single-neuron projection analysis revealed multiple projectome-based subtypes of CEAMOR neurons, each innervating distinct target brain regions. We found that the suppression of axon branches projecting to the parabrachial nucleus (PB) of one subtype of CEAMOR neurons alleviated persistent hyperalgesia, indicating a subtype- and axonal-branch-specific mechanism of action. Further electrophysiological analysis revealed that suppression of a distinct CEA-PB disinhibitory circuit controlled endogenous opioid antinociception. Thus, this study identified the central neural circuit that underlies endogenous opioid antinociception, providing new insight into the endogenous pain modulatory mechanisms.
Collapse
Affiliation(s)
- Yan-Nong Dou
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Department of Biology, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Lingang Laboratory, Shanghai 200031, China
| | - Wen-Qun Ding
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Li
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hua Zhou
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ling Li
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng-Ting Zhao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zheng-Yi-Qi Li
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jing Yuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Xiao-Fei Wang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wang-Yuan Zou
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215123, China
| | - Yan-Gang Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
2
|
Xie Y, Brynildsen JK, Windisch K, Blendy JA. Neural Network Connectivity Following Opioid Dependence is Altered by a Common Genetic Variant in the µ-Opioid Receptor, OPRM1 A118G. J Neurosci 2024; 44:e1492232023. [PMID: 38124015 PMCID: PMC10866092 DOI: 10.1523/jneurosci.1492-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
Opioid use disorder is a chronic, relapsing disease associated with persistent changes in brain plasticity. A common single nucleotide polymorphism (SNP) in the µ-opioid receptor gene, OPRM1 A118G, is associated with altered vulnerability to opioid addiction. Reconfiguration of neuronal connectivity may explain dependence risk in individuals with this SNP. Mice with the equivalent Oprm1 variant, A112G, demonstrate sex-specific alterations in the rewarding properties of morphine and heroin. To determine whether this SNP influences network-level changes in neuronal activity, we compared FOS expression in male and female mice that were opioid-naive or opioid-dependent. Network analyses identified significant differences between the AA and GG Oprm1 genotypes. Based on several graph theory metrics, including small-world analysis and degree centrality, we show that GG females in the opioid-dependent state exhibit distinct patterns of connectivity compared to other groups of the same genotype. Using a network control theory approach, we identified key cortical brain regions that drive the transition between opioid-naive and opioid-dependent brain states; however, these regions are less influential in GG females leading to sixfold higher average minimum energy needed to transition from the acute to the dependent state. In addition, we found that the opioid-dependent brain state is significantly less stable in GG females compared to other groups. Collectively, our findings demonstrate sex- and genotype-specific modifications in local, mesoscale, and global properties of functional brain networks following opioid exposure and provide a framework for identifying genotype differences in specific brain regions that play a role in opioid dependence.
Collapse
Affiliation(s)
- Yihan Xie
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Julia K Brynildsen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Kyle Windisch
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics and Perelman School of Medicine, University of Pennsylvania, Philadelphia 19104, Pennsylvania
| |
Collapse
|
3
|
Hirota I, Koyama Y, Shimada S. Histochemical analysis of the biphasic properties of formalin pain-induced behavior. Biochem Biophys Rep 2023; 34:101467. [PMID: 37125080 PMCID: PMC10139972 DOI: 10.1016/j.bbrep.2023.101467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/24/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
The formalin test has been established as a method for evaluating mouse models of pain. Although there have been numerous reports of formalin-pain-induced behavior, few reports of a detailed histochemical analysis of the central nervous system focus on behavioral biphasic properties. To investigate the alternation of spinal neuronal activity with formalin-induced pain, we performed immunofluorescent staining with c-Fos antibodies as neuronal activity markers using acute pain model mice induced by 2% formalin stimulation. As a result, phase-specific expression patterns were observed. In the spinal dorsal horn region, there were many neural activities in the deep region (layers V-VII) in the behavioral first phase and those in the surface region (layers I-III) in the behavioral second phase. Furthermore, we conducted comparative studies using low concentrations (0.25%) of formalin and capsaicin, which did not show distinct behavioral biphasic properties. Neural activity was observed only in the spinal dorsal horn surface region for both stimuli. Our study suggested that the histochemical biphasic nature of formalin-induced pain was attributable to the activity of the deep region of the spinal cord. In the future, treatment strategies focusing on the deep region neuron will lead to the development of effective treatments for allodynia and intractable chronic pain.
Collapse
Affiliation(s)
- Ikuei Hirota
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
| | - Yoshihisa Koyama
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, 541-8567, Japan
- Corresponding author. Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Osaka University Graduate School of Medicine, Osaka, 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka, 541-8567, Japan
| |
Collapse
|
4
|
Sakloth F, Sanchez-Reyes OB, Ruiz A, Nicolais A, Serafini RA, Pryce KD, Bertherat F, Torres-Berrío A, Gomes I, Devi LA, Wacker D, Zachariou V. A Regional and Projection-Specific Role of RGSz1 in the Ventrolateral Periaqueductal Grey in the Modulation of Morphine Reward. Mol Pharmacol 2023; 103:1-8. [PMID: 36310031 PMCID: PMC11033942 DOI: 10.1124/molpharm.122.000528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023] Open
Abstract
Opioid analgesics exert their therapeutic and adverse effects by activating μ opioid receptors (MOPR); however, functional responses to MOPR activation are modulated by distinct signal transduction complexes within the brain. The ventrolateral periaqueductal gray (vlPAG) plays a critical role in modulation of nociception and analgesia, but the exact intracellular pathways associated with opioid responses in this region are not fully understood. We previously showed that knockout of the signal transduction modulator Regulator of G protein Signaling z1 (RGSz1) enhanced analgesic responses to opioids, whereas it decreased the rewarding efficacy of morphine. Here, we applied viral mediated gene transfer methodology and delivered adeno-associated virus (AAV) expressing Cre recombinase to the vlPAG of RGSz1fl\fl mice to demonstrate that downregulation of RGSz1 in this region decreases sensitivity to morphine in the place preference paradigm, under pain-free as well as neuropathic pain states. We also used retrograde viral vectors along with flippase-dependent Cre vectors to conditionally downregulate RGSz1 in vlPAG projections to the ventral tegmental area (VTA) and show that downregulation of RGSz1 prevents the development of place conditioning to low morphine doses. Consistent with the role for RGSz1 as a negative modulator of MOPR activity, RGSz1KO enhances opioid-induced cAMP inhibition in periaqueductal gray (PAG) membranes. Furthermore, using a new generation of bioluminescence resonance energy transfer (BRET) sensors, we demonstrate that RGSz1 modulates Gαz but not other Gαi family subunits and selectively impedes MOPR-mediated Gαz signaling events invoked by morphine and other opioids. Our work highlights a regional and circuit-specific role of the G protein-signaling modulator RGSz1 in morphine reward, providing insights on midbrain intracellular pathways that control addiction-related behaviors. SIGNIFICANCE STATEMENT: This study used advanced genetic mouse models to highlight the role of the signal transduction modulator named RGSz1 in responses to clinically used opioid analgesics. We show that RGSz1 controls the rewarding efficacy of opioids by actions in ventrolateral periaqueductal gray projections to the ventral tegmental area, a key component of the midbrain dopamine pathway. These studies highlight novel mechanisms by which pain-modulating structures control the rewarding efficacy of opioids.
Collapse
Affiliation(s)
- Farhana Sakloth
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Omar B Sanchez-Reyes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Anne Ruiz
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Andrew Nicolais
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Randal A Serafini
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Kerri D Pryce
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Feodora Bertherat
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Angélica Torres-Berrío
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Ivone Gomes
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Lakshmi A Devi
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Daniel Wacker
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute (F.S., A.R., A.N., R.A.S., K.D.P., F.B., A.T.-B., L.A.D., D.W., V.Z.) and Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York (O.B.S.R., I.G., L.A.D., D.W., V.Z.)
| |
Collapse
|
5
|
Tinnermann A, Sprenger C, Büchel C. Opioid analgesia alters corticospinal coupling along the descending pain system in healthy participants. eLife 2022; 11:74293. [PMID: 35471139 PMCID: PMC9042228 DOI: 10.7554/elife.74293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Opioids are potent analgesic drugs with widespread cortical, subcortical, and spinal targets. In particular, the central pain system comprising ascending and descending pain pathways has high opioid receptor densities and is thus crucial for opioid analgesia. Here, we investigated the effects of the opioid remifentanil in a large sample (n = 78) of healthy male participants using combined corticospinal functional MRI. This approach offers the possibility to measure BOLD responses simultaneously in the brain and spinal cord, allowing us to investigate the role of corticospinal coupling in opioid analgesia. Our data show that opioids altered activity in regions involved in pain processing such as somatosensory regions, including the spinal cord and pain modulation such as prefrontal regions. Moreover, coupling strength along the descending pain system, that is, between the anterior cingulate cortex, periaqueductal gray, and spinal cord, was stronger in participants who reported stronger analgesia during opioid treatment while participants that received saline showed reduced coupling when experiencing less pain. These results indicate that coupling along the descending pain pathway is a potential mechanism of opioid analgesia and can differentiate between opioid analgesia and unspecific reductions in pain such as habituation.
Collapse
Affiliation(s)
- Alexandra Tinnermann
- Department for Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Max Planck School of Cognition, Leipzig, Germany
| | - Christian Sprenger
- Department for Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Büchel
- Department for Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Max Planck School of Cognition, Leipzig, Germany
| |
Collapse
|
6
|
Hyporesponsivity to mu-opioid receptor agonism in the Wistar-Kyoto rat model of altered nociceptive responding associated with negative affective state. Pain 2021; 162:405-420. [PMID: 32826755 DOI: 10.1097/j.pain.0000000000002039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/03/2020] [Indexed: 11/25/2022]
Abstract
ABSTRACT Chronic pain is often comorbid with anxiety and depression, altering the level of perceived pain, which negatively affects therapeutic outcomes. The role of the endogenous mu-opioid receptor (MOP) system in pain-negative affect interactions and the influence of genetic background thereon are poorly understood. The inbred Wistar-Kyoto (WKY) rat, which mimics aspects of anxiety and depression, displays increased sensitivity (hyperalgesia) to noxious stimuli, compared with Sprague-Dawley (SD) rats. Here, we report that WKY rats are hyporesponsive to the antinociceptive effects of systemically administered MOP agonist morphine in the hot plate and formalin tests, compared with SD counterparts. Equivalent plasma morphine levels in the 2 rat strains suggested that these differences in morphine sensitivity were unlikely to be due to strain-related differences in morphine pharmacokinetics. Although MOP expression in the ventrolateral periaqueductal gray (vlPAG) did not differ between WKY and SD rats, the vlPAG was identified as a key locus for the hyporesponsivity to MOP agonism in WKY rats in the formalin test. Moreover, morphine-induced effects on c-Fos (a marker of neuronal activity) in regions downstream of the vlPAG, namely, the rostral ventromedial medulla and lumbar spinal dorsal horn, were blunted in the WKY rats. Together, these findings suggest that a deficit in the MOP-induced recruitment of the descending inhibitory pain pathway may underlie hyperalgesia to noxious inflammatory pain in the WKY rat strain genetically predisposed to negative affect.
Collapse
|
7
|
Cortical Modulation of Nociception. Neuroscience 2021; 458:256-270. [PMID: 33465410 DOI: 10.1016/j.neuroscience.2021.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/28/2020] [Accepted: 01/03/2021] [Indexed: 02/06/2023]
Abstract
Nociception is the neuronal process of encoding noxious stimuli and could be modulated at peripheral, spinal, brainstem, and cortical levels. At cortical levels, several areas including the anterior cingulate cortex (ACC), prefrontal cortex (PFC), ventrolateral orbital cortex (VLO), insular cortex (IC), motor cortex (MC), and somatosensory cortices are involved in nociception modulation through two main mechanisms: (i) a descending modulatory effect at spinal level by direct corticospinal projections or mostly by activation of brainstem structures (i.e. periaqueductal grey matter (PAG), locus coeruleus (LC), the nucleus of raphe (RM) and rostroventral medulla (RVM)); and by (ii) cortico-cortical or cortico-subcortical interactions. This review summarizes evidence related to the participation of the aforementioned cortical areas in nociception modulation and different neurotransmitters or neuromodulators that have been studied in each area. Besides, we point out the importance of considering intracortical neuronal populations and receptors expression, as well as, nociception-induced cortical changes, both functional and connectional, to better understand this modulatory effect. Finally, we discuss the possible mechanisms that could potentiate the use of cortical stimulation as a promising procedure in pain alleviation.
Collapse
|
8
|
Zhang XY, Dou YN, Yuan L, Li Q, Zhu YJ, Wang M, Sun YG. Different neuronal populations mediate inflammatory pain analgesia by exogenous and endogenous opioids. eLife 2020; 9:55289. [PMID: 32519950 PMCID: PMC7311172 DOI: 10.7554/elife.55289] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Mu-opioid receptors (MORs) are crucial for analgesia by both exogenous and endogenous opioids. However, the distinct mechanisms underlying these two types of opioid analgesia remain largely unknown. Here, we demonstrate that analgesic effects of exogenous and endogenous opioids on inflammatory pain are mediated by MORs expressed in distinct subpopulations of neurons in mice. We found that the exogenous opioid-induced analgesia of inflammatory pain is mediated by MORs in Vglut2+ glutamatergic but not GABAergic neurons. In contrast, analgesia by endogenous opioids is mediated by MORs in GABAergic rather than Vglut2+ glutamatergic neurons. Furthermore, MORs expressed at the spinal level is mainly involved in the analgesic effect of morphine in acute pain, but not in endogenous opioid analgesia during chronic inflammatory pain. Thus, our study revealed distinct mechanisms underlying analgesia by exogenous and endogenous opioids, and laid the foundation for further dissecting the circuit mechanism underlying opioid analgesia.
Collapse
Affiliation(s)
- Xin-Yan Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Nong Dou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Lei Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qing Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yan-Jing Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Meng Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science & Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
9
|
Campos ACP, Berzuino MB, Hernandes MS, Fonoff ET, Pagano RL. Monoaminergic regulation of nociceptive circuitry in a Parkinson's disease rat model. Exp Neurol 2019; 318:12-21. [DOI: 10.1016/j.expneurol.2019.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/11/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
|
10
|
Lopes PSS, Campos ACP, Fonoff ET, Britto LRG, Pagano RL. Motor cortex and pain control: exploring the descending relay analgesic pathways and spinal nociceptive neurons in healthy conscious rats. Behav Brain Funct 2019; 15:5. [PMID: 30909927 PMCID: PMC6432755 DOI: 10.1186/s12993-019-0156-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 03/14/2019] [Indexed: 01/02/2023] Open
Abstract
Motor cortex stimulation (MCS) is an effective therapy for refractory neuropathic pain. MCS increases the nociceptive threshold in healthy rats via endogenous opioids, inhibiting thalamic nuclei and activating the periaqueductal gray. It remains unclear how the motor cortex induces top-down modulation of pain in the absence of persistent pain. Here, we investigated the main nuclei involved in the descending analgesic pathways and the spinal nociceptive neurons in rats that underwent one session of MCS and were evaluated with the paw pressure nociceptive test. The pattern of neuronal activation in the dorsal raphe nucleus (DRN), nucleus raphe magnus (NRM), locus coeruleus (LC), and dorsal horn of the spinal cord (DHSC) was assessed by immunoreactivity (IR) for Egr-1 (a marker of activated neuronal nuclei). IR for serotonin (5HT) in the DRN and NRM, tyrosine hydroxylase (TH) in the LC, and substance P (SP) and enkephalin (ENK) in the DHSC was also evaluated. MCS increased the nociceptive threshold of the animals; this increase was accompanied by activation of the NRM, while DRN activation was unchanged. However, cortical stimulation induced an increase in 5HT-IR in both serotonergic nuclei. MCS did not change the activation pattern or TH-IR in the LC, and it inhibited neuronal activation in the DHSC without altering SP or ENK-IR. Taken together, our results suggest that MCS induces the activation of serotonergic nuclei as well as the inhibition of spinal neurons, and such effects may contribute to the elevation of the nociceptive threshold in healthy rats. These results allow a better understanding of the circuitry involved in the antinociceptive top-down effect induced by MCS under basal conditions, reinforcing the role of primary motor cortex in pain control.
Collapse
Affiliation(s)
- Patrícia Sanae Souza Lopes
- Laboratory of Neuroscience, Hospital Sírio Libanês, São Paulo, SP, 01308-060, Brazil.,Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | | | - Erich Talamoni Fonoff
- Laboratory of Neuroscience, Hospital Sírio Libanês, São Paulo, SP, 01308-060, Brazil.,Department of Neurology, School of Medicine, University of São Paulo, São Paulo, SP, 01060-970, Brazil
| | - Luiz Roberto Giorgetti Britto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Rosana Lima Pagano
- Laboratory of Neuroscience, Hospital Sírio Libanês, São Paulo, SP, 01308-060, Brazil.
| |
Collapse
|
11
|
Manglik A, Lin H, Aryal DK, McCorvy JD, Dengler D, Corder G, Levit A, Kling RC, Bernat V, Hübner H, Huang XP, Sassano MF, Giguère PM, Löber S, Da Duan, Scherrer G, Kobilka BK, Gmeiner P, Roth BL, Shoichet BK. Structure-based discovery of opioid analgesics with reduced side effects. Nature 2016; 537:185-190. [PMID: 27533032 PMCID: PMC5161585 DOI: 10.1038/nature19112] [Citation(s) in RCA: 686] [Impact Index Per Article: 76.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 07/14/2016] [Indexed: 12/12/2022]
Abstract
Morphine is an alkaloid from the opium poppy used to treat pain. The potentially lethal side effects of morphine and related opioids-which include fatal respiratory depression-are thought to be mediated by μ-opioid-receptor (μOR) signalling through the β-arrestin pathway or by actions at other receptors. Conversely, G-protein μOR signalling is thought to confer analgesia. Here we computationally dock over 3 million molecules against the μOR structure and identify new scaffolds unrelated to known opioids. Structure-based optimization yields PZM21-a potent Gi activator with exceptional selectivity for μOR and minimal β-arrestin-2 recruitment. Unlike morphine, PZM21 is more efficacious for the affective component of analgesia versus the reflexive component and is devoid of both respiratory depression and morphine-like reinforcing activity in mice at equi-analgesic doses. PZM21 thus serves as both a probe to disentangle μOR signalling and a therapeutic lead that is devoid of many of the side effects of current opioids.
Collapse
MESH Headings
- Analgesia/methods
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/pharmacology
- Animals
- Drug Discovery
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- HEK293 Cells
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Docking Simulation
- Pain/drug therapy
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Spiro Compounds/pharmacology
- Structure-Activity Relationship
- Thiophenes/adverse effects
- Thiophenes/chemistry
- Thiophenes/pharmacology
- Urea/adverse effects
- Urea/analogs & derivatives
- Urea/chemistry
- Urea/pharmacology
Collapse
Affiliation(s)
- Aashish Manglik
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Henry Lin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, USA
| | - Dipendra K Aryal
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - John D McCorvy
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - Daniela Dengler
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Gregory Corder
- Department of Anesthesiology, Perioperative and Pain Medicine, Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Anat Levit
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, USA
| | - Ralf C Kling
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
- Institut für Physiologie und Pathophysiologie, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Viachaslau Bernat
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Xi-Ping Huang
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - Maria F Sassano
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - Patrick M Giguère
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - Stefan Löber
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Da Duan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, USA
| | - Grégory Scherrer
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Anesthesiology, Perioperative and Pain Medicine, Neurosurgery, Stanford Neurosciences Institute, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schuhstraße 19, 91052 Erlangen, Germany
| | - Bryan L Roth
- Department of Pharmacology, UNC Chapel Hill Medical School, Chapel Hill, North Carolina 27514, USA
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158, USA
| |
Collapse
|
12
|
Ang ST, Ariffin MZ, Khanna S. The forebrain medial septal region and nociception. Neurobiol Learn Mem 2016; 138:238-251. [PMID: 27444843 DOI: 10.1016/j.nlm.2016.07.017] [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: 05/12/2016] [Revised: 07/08/2016] [Accepted: 07/17/2016] [Indexed: 10/21/2022]
Abstract
The forebrain medial septum, which is an integral part of the septo-hippocampal network, is implicated in sensorimotor integration, fear and anxiety, and spatial learning and memory. A body of evidence also suggests that the septal region affects experimental pain. Indeed, some explorations in humans have raised the possibility that the region may modulate clinical pain as well. This review explores the evidence that implicates the medial septum in nociception and suggests that non-overlapping circuits in the region facilitate acute nociceptive behaviors and defensive behaviors that reflect affect and cognitive appraisal, especially in relation to persistent nociception. In line with a role in nociception, the region modulates nociceptive responses in the neuraxis, including the hippocampus and the anterior cingulate cortex. The aforementioned forebrain regions have also been implicated in persistent/long-lasting nociception. The review also weighs the effects of the medial septum on nociception vis-à-vis the known roles of the region and emphasizes the fact that the region is a part of network of forebrain structures which have been long associated with reward, cognition and affect-motivation and are now implicated in persistent/long-lasting nociception.
Collapse
Affiliation(s)
- Seok Ting Ang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mohammed Zacky Ariffin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sanjay Khanna
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Program, Life Sciences Institute, National University of Singapore, Singapore.
| |
Collapse
|
13
|
Dimov LF, Franciosi AC, Campos ACP, Brunoni AR, Pagano RL. Top-Down Effect of Direct Current Stimulation on the Nociceptive Response of Rats. PLoS One 2016; 11:e0153506. [PMID: 27071073 PMCID: PMC4829148 DOI: 10.1371/journal.pone.0153506] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/30/2016] [Indexed: 01/06/2023] Open
Abstract
Transcranial direct current stimulation (tDCS) is an emerging, noninvasive technique of neurostimulation for treating pain. However, the mechanisms and pathways involved in its analgesic effects are poorly understood. Therefore, we investigated the effects of direct current stimulation (DCS) on thermal and mechanical nociceptive thresholds and on the activation of the midbrain periaqueductal gray (PAG) and the dorsal horn of the spinal cord (DHSC) in rats; these central nervous system areas are associated with pain processing. Male Wistar rats underwent cathodal DCS of the motor cortex and, while still under stimulation, were evaluated using tail-flick and paw pressure nociceptive tests. Sham stimulation and naive rats were used as controls. We used a randomized design; the assays were not blinded to the experimenter. Immunoreactivity of the early growth response gene 1 (Egr-1), which is a marker of neuronal activation, was evaluated in the PAG and DHSC, and enkephalin immunoreactivity was evaluated in the DHSC. DCS did not change the thermal nociceptive threshold; however, it increased the mechanical nociceptive threshold of both hind paws compared with that of controls, characterizing a topographical effect. DCS decreased the Egr-1 labeling in the PAG and DHSC as well as the immunoreactivity of spinal enkephalin. Altogether, the data suggest that DCS disinhibits the midbrain descending analgesic pathway, consequently inhibiting spinal nociceptive neurons and causing an increase in the nociceptive threshold. This study reinforces the idea that the motor cortex participates in the neurocircuitry that is involved in analgesia and further clarifies the mechanisms of action of tDCS in pain treatment.
Collapse
Affiliation(s)
- Luiz Fabio Dimov
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - Adriano Cardozo Franciosi
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - Ana Carolina Pinheiro Campos
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| | - André Russowsky Brunoni
- Service of Interdisciplinary Neuromodulation (SIN), Department and Institute of Psychiatry, Faculty of Medicine of University of São Paulo, Laboratory of Neuroscience (LIM27), Department and Institute of Psychiatry, University of São Paulo, Rua Doutor Ovidio Pires de Campos, 785, Sao Paulo, SP, 05403-000, Brazil.,Center for Clinical and Epidemiological Research & Interdisciplinary Center for Applied Neuromodulation (CINA), University Hospital, University of São Paulo, São Paulo, Avenida Professor Lineu Prestes 2565, ext. 3, Sao Paulo, SP, 05508-000, Brazil
| | - Rosana Lima Pagano
- Laboratory of Neuromodulation and Experimental Pain, Hospital Sírio Libanês, Rua Prof Daher Cutait, 69, Sao Paulo, SP, 01308-060, Brazil
| |
Collapse
|
14
|
Chang KH, Bai SJ, Lee H, Lee BH. Effects of acupuncture stimulation at different acupoints on formalin-induced pain in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:121-7. [PMID: 24757373 PMCID: PMC3994298 DOI: 10.4196/kjpp.2014.18.2.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/05/2014] [Accepted: 02/28/2014] [Indexed: 11/22/2022]
Abstract
Acupuncture is the process of stimulating skin regions called meridians or acupoints and has been used to treat pain-related symptoms. However, the pain-relieving effects of acupuncture may be different depending on acupoints. In the present study, the effects of acupuncture on behavioral responses and c-Fos expression were evaluated using a formalin test in male Sprague-Dawley rats in order to clarify the analgesic effects of three different acupoints. Each rat received manual acupuncture at the ST36 (Zusanli), SP9 (Yinlingquan) or BL60 (Kunlun) acupoint before formalin injection. Flinching and licking behaviors were counted by two blinded investigators. Fos-like immunoreactivity was examined by immunohistochemistry in the rat spinal cord. Manual acupuncture treatment at BL60 acupoint showed significant inhibition in flinching behavior but not in licking. Manual acupuncture at ST36 or SP9 tended to inhibit flinching and licking behaviors but the effects were not statistically significant. The acupuncture at ST36, SP9, or BL60 reduced c-Fos expression as compared with the control group. These results suggest that acupuncture especially at the BL60 acupoint is more effective in relieving inflammatory pain than other acupoints.
Collapse
Affiliation(s)
- Kyung Ha Chang
- Department of Physiology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Sun Joon Bai
- Department of Anesthesiology and Pain Medicine and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Hyejung Lee
- Acupuncture and Meridian Science Research Center, Kyung Hee University, Seoul 130-701, Korea
| | - Bae Hwan Lee
- Department of Physiology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| |
Collapse
|
15
|
Spinal distribution of c-Fos activated neurons expressing enkephalin in acute and chronic pain models. Brain Res 2014; 1543:83-92. [DOI: 10.1016/j.brainres.2013.10.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 01/08/2023]
|
16
|
Kwok CHT, Devonshire IM, Bennett AJ, Hathway GJ. Postnatal maturation of endogenous opioid systems within the periaqueductal grey and spinal dorsal horn of the rat. Pain 2013; 155:168-178. [PMID: 24076162 PMCID: PMC3894430 DOI: 10.1016/j.pain.2013.09.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/03/2013] [Accepted: 09/20/2013] [Indexed: 12/31/2022]
Abstract
Significant opioid-dependent changes occur during the fourth postnatal week in supraspinal sites (rostroventral medulla [RVM], periaqueductal grey [PAG]) that are involved in the descending control of spinal excitability via the dorsal horn (DH). Here we report developmentally regulated changes in the opioidergic signalling within the PAG and DH, which further increase our understanding of pain processing during early life. Microinjection of the μ-opioid receptor (MOR) agonist DAMGO (30 ng) into the PAG of Sprague-Dawley rats increased spinal excitability and lowered mechanical threshold to noxious stimuli in postnatal day (P)21 rats, but had inhibitory effects in adults and lacked efficacy in P10 pups. A tonic opioidergic tone within the PAG was revealed in adult rats by intra-PAG microinjection of CTOP (120 ng, MOR antagonist), which lowered mechanical thresholds and increased spinal reflex excitability. Spinal adminstration of DAMGO inhibited spinal excitability in all ages, yet the magnitude of this was greater in younger animals than in adults. The expression of MOR and related peptides were also investigated using TaqMan real-time polymerase chain reaction and immunohistochemistry. We found that pro-opiomelanocortin peaked at P21 in the ventral PAG, and MOR increased significantly in the DH as the animals aged. Enkephalin mRNA transcripts preceded the increase in enkephalin immunoreactive fibres in the superficial dorsal horn from P21 onwards. These results illustrate that profound differences in the endogenous opioidergic signalling system occur throughout postnatal development.
Collapse
Affiliation(s)
- Charlie H T Kwok
- Laboratory of Developmental Nociception, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK FRAME Laboratory, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
17
|
Dynamic genotype-selective "phenotypic switching" of CGRP expression contributes to differential neuropathic pain phenotype. Exp Neurol 2013; 250:194-204. [PMID: 24076003 DOI: 10.1016/j.expneurol.2013.09.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 08/18/2013] [Accepted: 09/16/2013] [Indexed: 11/23/2022]
Abstract
Using a genetic model we demonstrate the role played by "phenotypic switching" of calcitonin gene related peptide (CGRP) expression in axotomized large Aβ afferents in the development of neuropathic pain behavior in rats. After nerve injury both substance P and CGRP are upregulated in Aβ afferents in the corresponding DRGs. It has been proposed that intraspinal release of these neurotransmitters upon gentle stroking of skin drives ascending pain signaling pathways resulting in tactile allodynia. We reported previously that in rat lines genetically selected for high (HA) vs. low (LA) pain phenotype, SP is upregulated equally in both strains, but that CGRP is upregulated exclusively in the pain prone HA line (Nitzan-Luques et al., 2011). This implicates CGRP as the principal driver of tactile allodynia. Here we confirm this conclusion by showing: 1) that the time of emergence of CGRP-IR in DRG Aβ neurons and their central terminals in HA rats matches that of pain behavior, 2) that following spinal nerve lesion (SNL) selective activation of low threshold afferents indeed drives postsynaptic pain-signaling neurons and induces central sensitization in HA rats, as monitored using c-Fos as a marker. These changes are much less prominent in LA rats, 3) that intrathecal (i.t.) administration of CGRP induces tactile allodynia in naïve rats and 4) that i.t. administration of the CGRP-receptor antagonist BIBN4096BS (Olcegepant) attenuates SNL-evoked tactile allodynia, without blocking baseline nociception. Together, these observations support the hypothesis that genotype-selective phenotypic switching of CGRP expression in Aβ afferents following nerve injury is a fundamental mechanism of neuropathic tactile allodynia.
Collapse
|
18
|
Martins I, Cabral L, Pinto A, Wilson S, Lima D, Tavares I. Reversal of inflammatory pain by HSV-1-mediated overexpression of enkephalin in the caudal ventrolateral medulla. Eur J Pain 2012; 15:1008-14. [DOI: 10.1016/j.ejpain.2011.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 03/11/2011] [Accepted: 04/05/2011] [Indexed: 01/28/2023]
|
19
|
Tavares I, Almeida A, Albino-Teixeira A, Lima D. Lesions of the caudal ventrolateral medulla block the hypertension-induced inhibition of noxious-evoked c-fos expression in the rat spinal cord. Eur J Pain 2012; 1:149-60. [PMID: 15102416 DOI: 10.1016/s1090-3801(97)90073-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/1997] [Accepted: 07/11/1997] [Indexed: 11/20/2022]
Abstract
The effect of lesioning the lateral portion of the caudal ventrolateral medullary reticular formation (VLMIat) on the noxious-evoked expression of the c-fos proto-oncogene in spinal neurons, was studied in short-term hypertensive rats. Occlusion of the renal artery for 96 h in unlesioned animals induced a 52% increase in blood pressure (BP) and a 66% decrease in the number of Fos-immunoreactive (Fos-IR) spinal cells following noxious cutaneous stimulation, as compared to values in normotensive controls. Lesioning the VLMIat in hypertensive rats by unilateral quinolinic acid (QA) injection (0.3 microl of a 180 nmol/microl solution) 24 h before noxious stimulation, prevented the Fos-IR cell decrease. In normotensive rats, lesioning the VLMIat produced no changes in c-fos expression. To investigate the role played by the VLMIat in cardiovascular control, BP and heart rate (HR) were measured during local injections of QA or glutamate (0.5 microl of a 100 nmol/microl solution) to normotensive animals. Injections of QA produced an immediate rise in BP and HR which reached maximal values (18 and 14% increase, respectively) 5 min after the administration onset, then returning gradually to baseline levels. Glutamate injections resulted in an immediate decrease of the same values, which reached 29 and 39%, respectively, 4 min after the beginning of injection, after which they decreased to baseline levels. These results suggest that VLMIat neurons inhibit nociceptive spinal neurons in response to rises in blood pressure, while exerting negative control of cardiovascular parameters. It is suggested that the VLMIat is involved in the genesis of hypoalgesia during hypertension.
Collapse
Affiliation(s)
- I Tavares
- Institute of Histology and Embryology, Faculty of Medicine and IBMC of the University of Oporto, Porto, Portugal
| | | | | | | |
Collapse
|
20
|
Kovelowski CJ, Raffa RB, Porreca F. Tramadol and its enantiomers differentially suppress c-fos-like immunoreactivity in rat brain and spinal cord following acute noxious stimulus. Eur J Pain 2012; 2:211-9. [PMID: 15102381 DOI: 10.1016/s1090-3801(98)90017-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/1997] [Revised: 04/16/1998] [Accepted: 05/05/1998] [Indexed: 11/26/2022]
Abstract
Tramadol hydrochloride, (1RS,2RS)-2-[(dimethylamino)methyl]-1-(3-methoxyphenyl)-cyclohexanol hydrochloride, is an orally-active, centrally-acting analgesic with a putative dual mechanism of action, including an opioid and non-opioid component. The analgesic properties of tramadol and the possible co-existence of dual mechanisms has been postulated to be due to complementary and interactive pharmacologies of its enantiomers. We examined the ability of tramadol, its enantiomers, and morphine as reference to suppress c-fos-like immunoreactivity (c-fos-ir) in rat spinal cord and brain regions following a noxious stimulus (i.p. administration of 3.5% acetic acid). c-fos-ir was measured by immunocytochemistry and the stained cells in each region were counted 2 h after the acetic-acid injection (2:25 h after tramadol or morphine). Equi-analgesic doses of s.c. morphine (10 mg/kg) or tramadol (30 mg/kg) significantly suppressed c-fos-ir in all areas examined, except dorsal central gray of the spinal cord. The enantiomers of tramadol had distinctive patterns of suppression, neither one suppressed c-fos-ir in all of the regions, and hence neither one alone accounted for the suppression produced by the racemate. These findings support differential and complementary effects of tramadol enantiomers in sub-populations of spinal and supraspinal nociceptive neurons, consistent with the proposed antinociceptive interaction between the enantiomers.
Collapse
Affiliation(s)
- C J Kovelowski
- Department of Pharmacology, The University of Arizona Health Sciences Center, Tucson, AZ, USA
| | | | | |
Collapse
|
21
|
Pan Y, Yin Z, Yang J, Zhao Y, Yan X, Qiu P, Wang D. Oxytocin in rat nucleus raphe magnus influences pain modulation. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/wjns.2012.22017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Forebrain medial septum region facilitates nociception in a rat formalin model of inflammatory pain. Pain 2011; 152:2528-2542. [DOI: 10.1016/j.pain.2011.07.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 06/16/2011] [Accepted: 07/26/2011] [Indexed: 12/25/2022]
|
23
|
Pagano RL, Assis DV, Clara JA, Alves AS, Dale CS, Teixeira MJ, Fonoff ET, Britto LR. Transdural motor cortex stimulation reverses neuropathic pain in rats: a profile of neuronal activation. Eur J Pain 2011; 15:268.e1-14. [PMID: 20817578 DOI: 10.1016/j.ejpain.2010.08.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Revised: 07/19/2010] [Accepted: 08/06/2010] [Indexed: 10/19/2022]
Abstract
Motor cortex stimulation (MCS) has been used to treat patients with neuropathic pain resistant to other therapeutic approaches; however, the mechanisms of pain control by MCS are still not clearly understood. We have demonstrated that MCS increases the nociceptive threshold of naive conscious rats, with opioid participation. In the present study, the effect of transdural MCS on neuropathic pain in rats subjected to chronic constriction injury of the sciatic nerve was investigated. In addition, the pattern of neuronal activation, evaluated by Fos and Zif268 immunolabel, was performed in the spinal cord and brain sites associated with the modulation of persistent pain. MCS reversed the mechanical hyperalgesia and allodynia induced by peripheral neuropathy. After stimulation, Fos immunoreactivity (Fos-IR) decreased in the dorsal horn of the spinal cord and in the ventral posterior lateral and medial nuclei of the thalamus, when compared to animals with neuropathic pain. Furthermore, the MCS increased the Fos-IR in the periaqueductal gray, the anterior cingulate cortex and the central and basolateral amygdaloid nuclei. Zif268 results were similar to those obtained for Fos, although no changes were observed for Zif268 in the anterior cingulate cortex and the central amygdaloid nucleus after MCS. The present findings suggest that MCS reverts neuropathic pain phenomena in rats, mimicking the effect observed in humans, through activation of the limbic and descending pain inhibitory systems. Further investigation of the mechanisms involved in this effect may contribute to the improvement of the clinical treatment of persistent pain.
Collapse
Affiliation(s)
- Rosana L Pagano
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Hwang HJ, Kim P, Kim CJ, Lee HJ, Shim I, Yin CS, Yang Y, Hahm DH. Antinociceptive effect of amygdalin isolated from Prunus armeniaca on formalin-induced pain in rats. Biol Pharm Bull 2008; 31:1559-64. [PMID: 18670089 DOI: 10.1248/bpb.31.1559] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amygdalin is a plant glucoside isolated from the stones of rosaceous fruits, such as apricots, peaches, almond, cherries, and plums. To investigate the pain-relieving activity of amygdalin, we induced pain in rats through intraplantar injection of formalin, and evaluated the antinociceptive effect of amygdalin at doses of 0.1, 0.5, 1.0, and 10.0 mg/kg-body weight by observing nociceptive behavior such as licking, biting and shaking, the number of Fos-immunoreactive neurons in the spinal cord, and the mRNA expression of inflammatory cytokines in the plantar skin. The intramuscular injection of amygdalin significantly reduced the formalin-induced tonic pain in both early (the initial 10 min after formalin injection) and late phases (10-30 min following the initial formalin injection). During the late phase, amygdalin did reduce the formalin-induced pain in a dose-dependent manner in a dose range less than 1 mg/kg. Molecular analysis targeting c-Fos and inflammatory cytokines such as tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1beta) also showed a significant effect of amygdalin, which matched the results of the behavioral pain analysis. These results suggest that amygdalin is effective at alleviating inflammatory pain and that it can be used as an analgesic with anti-nociceptive and anti-inflammatory activities.
Collapse
Affiliation(s)
- Hye-Jeong Hwang
- Acupuncture & Meridian Science Research Center, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen T, Hu Z, Quirion R, Hong Y. Modulation of NMDA receptors by intrathecal administration of the sensory neuron-specific receptor agonist BAM8-22. Neuropharmacology 2007; 54:796-803. [PMID: 18249418 DOI: 10.1016/j.neuropharm.2007.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2007] [Revised: 12/07/2007] [Accepted: 12/07/2007] [Indexed: 11/16/2022]
Abstract
The sensory neuron-specific receptor (SNSR) is exclusively distributed in dorsal root ganglion (DRG) cells. We have demonstrated that intrathecal (i.t.) administration of SNSR agonists inhibits formalin-evoked responses and the development of morphine tolerance [Chen, T., Cai, Q., Hong, Y., 2006. Intrathecal sensory neuron-specific receptor agonists bovine adrenal medulla 8-22 and (tyr(6))-gamma2-msh-6-12 inhibit formalin-evoked nociception and neuronal fos-like immunoreactivity in the spinal cord of the rat. Neuroscience 141, 965-975]. The present study was undertaken to examine the possible impact of the activation of SNSR on NMDA receptors. I.t. administration of NMDA (6.8 nmol) induced nociceptive behaviors, including scratching, biting and lifting, followed by thermal hypoalgesia and hyperalgesia. These responses were associated with the expression of Fos-like immunoreactivity (FLI) throughout the spinal dorsal horn with highest effect seen in laminae I-II. I.t. NMDA also induced an increase in nitric oxide synthase (NOS) activity in superficial layers of the dorsal horn, but not around the central canal, as revealed by NADPH diaphorase histochemistry. Pretreatment with the SNSR agonist bovine adrenal medulla 8-22 (3, 10 and 30 nmol) dose-dependently diminished NMDA-evoked nocifensive behaviors and hyperalgesia. This agonist also reduced NMDA-evoked expression of FLI and NADPH reactivity in the spinal dorsal horn. Taken together, these data suggest that the activation of SNSR induces spinal analgesia by suppressing NMDA receptor-mediated activation of spinal dorsal horn neurons and an increase in NOS activity.
Collapse
Affiliation(s)
- Tingjun Chen
- College of Life Sciences and Provincial Key Laboratory of Developmental Biology Neuroscience, Fujian Normal University Fuzhou, Fujian 350108, People's Republic of China
| | | | | | | |
Collapse
|
26
|
Koutsikou S, Parry DM, MacMillan FM, Lumb BM. Laminar organization of spinal dorsal horn neurones activated by C- vs. A-heat nociceptors and their descending control from the periaqueductal grey in the rat. Eur J Neurosci 2007; 26:943-52. [PMID: 17714188 PMCID: PMC2121136 DOI: 10.1111/j.1460-9568.2007.05716.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The periaqueductal grey can differentially control A- vs. C-nociceptor-evoked spinal reflexes and deep spinal dorsal horn neuronal responses. However, little is known about the control of A- vs. C-fibre inputs to lamina I and the lateral spinal nucleus, and how this correlates with the control of deeper laminae. To address this, the laminar distributions of neurones expressing Fos-like immunoreactivity were determined following preferential activation of A- or C-heat nociceptors, using fast or slow rates of skin heating, respectively, in the absence or presence of descending control evoked from the periaqueductal grey. In lamina I, numbers of Fos-positive neurones following both fast and slow rates of skin heating were reduced significantly following activation in the ventrolateral and dorsolateral/lateral periaqueductal grey. In contrast, in the deep dorsal horn (laminae III–VI), activation in both the ventrolateral and dorsolateral/lateral periaqueductal grey significantly reduced the numbers of Fos-positive neurones evoked by C- but not A-nociceptor stimulation. C- but not A-heat nociceptor activation evoked Fos bilaterally in the lateral spinal nucleus. Stimulation in the ventrolateral but not the dorsolateral/lateral periaqueductal grey significantly increased the numbers of Fos-positive neurones evoked by A- and C-nociceptor stimulation bilaterally in the lateral spinal nucleus. These data have demonstrated differences in the descending control of the superficial vs. the deep dorsal horn and lateral spinal nucleus with respect to the processing of A- and C-fibre-evoked events. The data are discussed in relation to the roles of A- and C-nociceptors in acute and chronic pain.
Collapse
Affiliation(s)
- Stella Koutsikou
- Department of Physiology, School of Medical Sciences, University Walk, University of Bristol, UK.
| | | | | | | |
Collapse
|
27
|
Castellanos DA, Daniels LA, Morales MP, Hama AT, Sagen J. Expansion of formalin-evoked Fos-immunoreactivity in rats with a spinal cord injury. Neurosci Res 2007; 58:386-93. [PMID: 17531342 PMCID: PMC2211738 DOI: 10.1016/j.neures.2007.04.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 04/24/2007] [Accepted: 04/26/2007] [Indexed: 11/23/2022]
Abstract
Peripheral tissue injury as well as spinal cord injury (SCI) may lead to sensitization of dorsal horn neurons and alterations in nociceptive processing. Thus, peripheral injuries experienced by SCI patients, even if not initially perceived, could result in a persistent and widespread activation of dorsal horn neurons and emerge as chronic pain with interventive repair or modest recovery from SCI. To visualize the spinal neuron response to peripheral tissue injury following complete SCI in rats, the neural transcription factor Fos was quantitated in the spinal cord. Two weeks following either a complete transection of the spinal cord at the level of T8 or a sham surgery (laminectomy), rats were injected with formalin into the left hind paw. Sham-operated rats demonstrated biphasic hind paw pain-related behavior following formalin injection, but transected rats displayed fewer behaviors in the second (tonic) phase. Stereological analysis of the sham group revealed that the extent of formalin-induced Fos expression was within the lumbar dorsal horn, with numerous Fos-like immunoreactive profiles in the ipsilateral dorsal horn and some contralateral immunoreactive profiles. In contrast, the level of Fos-like immunoreactivity in the transected group was significantly elevated and expanded in range compared to the sham group, with increases observed in the normal laminar distribution regions, as well as multi-segmentally through sacral levels and increases in the contralateral dorsal horn segments. The data demonstrate that widespread activation of spinal, especially dorsal horn, neurons following peripheral insult can occur in the injured spinal cord, despite reduced pain responsiveness, and suggests that exaggerated pain may emerge as spinal recovery or repair progresses.
Collapse
Affiliation(s)
- Daniel A Castellanos
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, R-48, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
28
|
Wiley RG, Kline RH, Vierck CJ. Anti-nociceptive effects of selectively destroying substance P receptor-expressing dorsal horn neurons using [Sar9,Met(O2)11]-substance P-saporin: behavioral and anatomical analyses. Neuroscience 2007; 146:1333-45. [PMID: 17418497 DOI: 10.1016/j.neuroscience.2007.01.066] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 01/29/2007] [Accepted: 01/31/2007] [Indexed: 10/23/2022]
Abstract
Lumbar intrathecal injections of substance P-saporin (SP-sap) destroy dorsal horn neurons that express the neurokinin-1 receptor (NK-1R) resulting in decreased responses to a range of noxious stimuli and decreased hyperalgesia and allodynia. Forebrain injections of SP-sap produce considerable non-specific damage raising some concern about use of this toxin in vivo. The more stable and selective substance P congener, [Sar9,Met(O2)11]substance P coupled to saporin (SSP-sap) produces much more selective forebrain lesions at significantly lower doses. The present study sought to determine the anatomic and nocifensive behavioral effects of lumbar intrathecal injections of the more precisely targeted SSP-sap. On the basis of loss of lamina I NK-1R staining, lumbar intrathecal SSP-sap was seven times more potent than SP-sap and produced no loss of NK-1R expressing neurons in deeper laminae (III-VI or X). Transient decreases in hotplate responding occurred at 44 degrees C and 47 degrees C but not 52 degrees C during the first 3 weeks after SSP-sap injection with return to baseline by 4 weeks. Operant escape responses were reduced at 0.3 degrees C, 44 degrees C and 47 degrees C for at least 4 months. In the formalin test, SSP-sap also was about seven times more potent than SP-sap in reducing phase two behavior in both female Long Evans and male Sprague-Dawley rats. Both SSP-sap and SP-sap reduced formalin-induced FOS expression in deep and superficial laminae of the L4 dorsal horn in parallel with the reduction in phase 2 behavior. In summary, SSP-sap is highly effective in destroying lamina I NK-1R expressing neurons, without loss of deep NK-1R neurons. The behavioral effects of SSP-sap are similar to SP-sap suggesting that the antinociceptive effects of both toxins are indeed due to selective loss of NK-1R neurons in lamina I. SSP-sap is an attractive agent for possible treatment of chronic pain.
Collapse
MESH Headings
- Analgesics
- Animals
- Behavior, Animal/drug effects
- Conditioning, Operant/drug effects
- Data Interpretation, Statistical
- Escape Reaction/drug effects
- Female
- Formaldehyde
- Genes, fos/drug effects
- Hot Temperature
- Immunohistochemistry
- Injections, Spinal
- Male
- Pain Measurement/drug effects
- Posterior Horn Cells/drug effects
- Posterior Horn Cells/metabolism
- Posterior Horn Cells/ultrastructure
- Rats
- Rats, Long-Evans
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Receptors, Neurokinin-1/biosynthesis
- Receptors, Neurokinin-1/metabolism
- Recombinant Fusion Proteins/pharmacology
- Ribosome Inactivating Proteins, Type 1
- Saporins
Collapse
Affiliation(s)
- R G Wiley
- Laboratory of Experimental Neurology, VA TVHS, Nashville, TN 37212-2637, USA.
| | | | | |
Collapse
|
29
|
Kim HY, Hahm DH, Chae Y, An K, Pyun KH, Lee H, Shim I. Acupuncture at GV01 Relieves Somatic Pain Referred by Colitis in Rats. J Physiol Sci 2007; 57:253-8. [PMID: 17666159 DOI: 10.2170/physiolsci.sc001607] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Accepted: 07/29/2007] [Indexed: 11/05/2022]
Abstract
The present study aimed to expand our previous findings regarding the therapeutic effects and underlying mechanisms of acupuncture at GV01 in colitis. Our results showed that acupuncture at GV01 has antinociceptive effects on referred somatic pain induced by experimental colitis, and that endogenous opioid pathways may mediate these effects.
Collapse
Affiliation(s)
- Hee-Young Kim
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555-1069, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Yang J, Chen JM, Liu WY, Song CY, Wang CH, Lin BC. Effect of arginine vasopressin in the nucleus raphe magnus on antinociception in the rat. Peptides 2006; 27:2224-9. [PMID: 16621154 DOI: 10.1016/j.peptides.2006.03.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2005] [Revised: 03/05/2006] [Accepted: 03/14/2006] [Indexed: 10/24/2022]
Abstract
Previous work has shown that arginine vasopressin (AVP) regulates antinociception through brain nuclei rather than the spinal cord and peripheral organs. The present study investigated the nociceptive effect of AVP in the nucleus raphe magnus (NRM) of the rat. Microinjection of AVP into the NRM increased pain threshold in a dose-dependent manner, while local administration of AVP-receptor antagonist-d(CH2)5Tyr(Et)DAVP decreased the pain threshold. Pain stimulation elevated AVP concentration in the NRM perfuse liquid. NRM pretreatment with AVP-receptor antagonist completely reversed AVP's effect on pain threshold in the NRM. The data suggest that AVP in the NRM is involved in antinociception.
Collapse
Affiliation(s)
- Jun Yang
- Institute for Pharmaceutical and Medicinal Science, Guangdong Bangmin Pharmaceutical Co. Ltd., Jianghai Distract, Jiangmen, Guangdong 529000, China.
| | | | | | | | | | | |
Collapse
|
31
|
Kim JH, Kim HK, Park YI, Sohn IC, Choi DO, Kim MS, Park BR. Moxibustion at ST36 alleviates pain in complete Freund's adjuvant-induced arthritic rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2006; 34:57-67. [PMID: 16437739 DOI: 10.1142/s0192415x06003631] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study was to investigate the antinociceptive effects of moxibustion in a complete Freund's adjuvant (CFA)-induced arthritic rat model, and the effects of moxibustion on immunohistochemical changes at the spinal cord level. Moxibustion was applied to the ipsilateral (right) Zusanli (ST36) acupoint to the lesion side for 9 days to CFA-induced arthritic rats. The stepping force was measured as a behavioral test, c-Fos immunohistochemistry, NO production and nNOS Western blots were examined to evaluate antinociceptive effects. Moxibustion at ST36 significantly improved the stepping force in the affected hind limb in CFA-induced arthritis. Moreover, moxibustion at ST36 suppressed the production of NO and the protein expression of c-Fos and nNOS induced by arthritis. These results suggest that moxibustion at ST36 has a potent antinociceptive effect in an arthritic rat model, and modulates neuronal excitability and endogenous NO production by suppressing c-Fos and nNOS protein expression.
Collapse
Affiliation(s)
- Jae-Hyo Kim
- Department of Meridian and Acupoint, College of Oriental Medicine, Wonkwang University, Iksan 570-749, Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
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: 5.9] [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
|
33
|
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]
|
34
|
Morato M, Pinho D, Sousa T, Tavares I, Albino-Teixeira A. Inhibition of nociceptive responses of spinal cord neurones during hypertension involves the spinal GABAergic system and a pain modulatory center located at the caudal ventrolateral medulla. J Neurosci Res 2006; 83:647-55. [PMID: 16453312 DOI: 10.1002/jnr.20770] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanisms of hypertension-induced hypoalgesia were studied in a model of hypertension induced by adenosine receptors blockade with the non-selective antagonist 1,3-dipropyl-8-sulfophenylxanthine (DPSPX) during 7 days. Based on the positive correlation between pain thresholds and noxious-evoked expression of the c-fos protooncogene in spinal cord neurones, we used this marker of nociceptive activation of spinal neurones to evaluate the involvement of the spinal GABAergic system and the caudal ventrolateral medulla (VLM), an important inhibitory component of the supraspinal endogenous pain modulatory system. In DPSPX-treated animals, a 20% increase in blood pressure was achieved along with a decrease in Fos expression in the superficial (laminae I-II) and deep (laminae III-VII) dorsal horn. In these animals, lower percentages of neurones labeled for GABAB receptors that expressed Fos were obtained in the superficial dorsal horn. Lesioning the VLMlat with quinolinic acid prevented the decrease in Fos expression at the spinal cord of DPSPX-hypertensive rats whereas in normotensive animals, no changes in Fos expression were detected. The present results support previous findings that hypertension is associated with a decrease of nociceptive activation of spinal cord neurones, through descending inhibition exerted by the VLMlat. This study further shows that during hypertension a decrease in the expression of GABAB receptors in nociceptive spinal neurones occurs, probably due to changes in the local GABAergic inhibitory system.
Collapse
Affiliation(s)
- Manuela Morato
- Institute of Pharmacology and Therapeutics, Faculty of Medicine of Porto and IBMC, University of Porto, Porto, Portugal
| | | | | | | | | |
Collapse
|
35
|
Sol JC, Li RY, Sallerin B, Jozan S, Zhou H, Lauwers-Cances V, Tortosa F, Chaynes P, Mascott C, Bastide R, Lazorthes Y. Intrathecal grafting of porcine chromaffin cells reduces formalin-evoked c-Fos expression in the rat spinal cord. Cell Transplant 2005; 14:353-65. [PMID: 16180654 DOI: 10.3727/000000005783982963] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Chromaffin cells from the adrenal gland secrete a combination of neuroactive compounds including catecholamines, opioid peptides, and growth factors that have strong analgesic effects, especially when administered intrathecally. Preclinical studies of intrathecal implantation with xenogeneic bovine chromaffin cells in rats have provided conflicting data with regard to analgesic effects, and recent concern over risk of prion transmission has precluded their use in human clinical trials. We previously developed a new, safer source of adult adrenal chromaffin cells of porcine origin and demonstrated an in vivo antinociceptive effect in the formalin test, a rodent model of tonic pain. The goal of the present study was to confirm porcine chromaffin cell analgesic effects at the molecular level by evaluating neural activity as reflected by spinal cord c-Fos protein expression. To this end, the expression of c-Fos in response to intraplantar formalin injection was evaluated in animals following intrathecal grafting of 10(6) porcine or bovine chromaffin cells. For the two species, adrenal chromaffin cells significantly reduced the tonic phases of the formalin response. Similarly, c-Fos-like immunoreactive neurons were markedly reduced in the dorsal horns of animals that had received injections of xenogeneic chromaffin cells. This reduction was observed in both the superficial (I-II) and deep (V-VI) lamina of the dorsal horn. The present study demonstrates that both xenogeneic porcine and bovine chromaffin cells transplanted into the spinal subarachnoid space of the rat can suppress formalin-evoked c-Fos expression equally, in parallel with suppression of nociceptive behaviors in the tonic phase of the test. These findings confirm previous reports that adrenal chromaffin cells may produce antinociception by inhibiting activation of nociceptive neurons in the spinal dorsal horn. Taken together these results support the concept that porcine chromaffin cells may offer an alternative xenogeneic cell source for transplants delivering pain-reducing neuroactive substances.
Collapse
Affiliation(s)
- J C Sol
- Laboratory of Pain and Cell Therapy, Rangueil Medical School, University Paul Sabatier, 133 route de Narbonne, 31062 Toulouse, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bhalla S, Matwyshyn G, Gulati A. Morphine tolerance does not develop in mice treated with endothelin-A receptor antagonists. Brain Res 2005; 1064:126-35. [PMID: 16289404 DOI: 10.1016/j.brainres.2005.09.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Revised: 09/05/2005] [Accepted: 09/24/2005] [Indexed: 10/25/2022]
Abstract
Long-term use of morphine leads to development of antinociceptive tolerance. We provide evidence that central endothelin (ET) mechanisms are involved in development of morphine tolerance. In the present study, we investigated the effect of ET(A) receptor antagonists, BQ123 and BMS182874, on morphine antinociception and tolerance in mice. Mechanism of interaction of ET(A) receptor antagonists with morphine was investigated. BQ123 (3 microg, i.c.v.) and BMS182874 (50 microg, i.c.v.) significantly enhanced antinociceptive effect of morphine (P < 0.05), through an opioid-mediated effect. Treatment with a single dose of BQ123 (3 microg, i.c.v.) reversed tolerance to morphine antinociception in morphine-tolerant mice. BQ123 or BMS182874 did not affect naloxone binding in the brain. Therefore, ET(A) receptor antagonists did not bind directly to opioid receptors. [35S]GTPgammaS binding was stimulated by morphine and ET-1 in non-tolerant mice. Morphine- and ET-1-induced GTP stimulation was significantly lower (P < 0.05) in morphine-tolerant group (33% and 42%, respectively) compared to control group. BQ123 and BMS182874 did not activate binding in non-tolerant mice. BQ123 and BMS182874 significantly increased G protein activation in morphine-tolerant mice (96% and 86%, respectively; P < 0.05). These results provide evidence that uncoupling of G protein occurs in morphine-tolerant mice, and ET(A) antagonists promote coupling of G protein to its receptors, thereby restoring antinociceptive effect. These findings indicate that ET(A) receptor antagonists potentiate morphine antinociception and reverse antinociceptive tolerance in mice, through their ability to couple G proteins to opioid receptors.
Collapse
Affiliation(s)
- Shaifali Bhalla
- Department of Biopharmaceutical Sciences (M/C 865), University of Illinois at Chicago, 833 South Wood Street, Chicago, IL 60612, USA
| | | | | |
Collapse
|
37
|
Kina VAV, Villarreal CF, Prado WA. The effects of intraspinal L-NOARG or SIN-1 on the control by descending pathways of incisional pain in rats. Life Sci 2005; 76:1939-51. [PMID: 15707877 DOI: 10.1016/j.lfs.2004.08.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Accepted: 08/10/2004] [Indexed: 11/30/2022]
Abstract
The modulation by spinal nitric oxide (NO) of descending pathways travelling through the dorsal lateral funiculus (DLF) is a mechanism proposed for the antinociceptive effects of drugs that changes the NO metabolism. In this study we confirm that a surgical incision in the mid-plantar hind paw of rats reduces the threshold to mechanical stimulation with von Frey filaments. The incisional pain was further increased in rats with ipsilateral DLF lesion. Intrathecal L-NOARG (50-300 microg), or SIN-1 (0.1-5.0 microg) reduced, while SIN-1 (10 and 20 microg) intensified the incisional pain in rats with sham or effective lesion of the DLF. Stimulation of the dorsal raphe (DRN) or anterior pretectal (APtN) nuclei with stepwise increased electrical currents (7, 14, 21, 28 and 35 microA r.m.s.) produced a current-related reduction of the incisional pain. These nuclei activate pain inhibitory pathways that descend to the spinal cord mainly through the DLF. Intrathecal SIN-1 (5 microg) reduced, SIN-1 (20 microg) decreased and L-NOARG (150 microg) did not change the EC50 for the DRN or APtN stimulation-induced reduction of incisional pain. We conclude that the antinociceptive effects of L-NOARG or low doses of SIN-1 are independent on the activity of descending pain control pathways travelling via the DLF, but the antinociceptive effect of stimulating electrically the DRN or APtN can be summated to the effect of low dose of SIN-1 or overcome by the high dose of SIN-1.
Collapse
Affiliation(s)
- Vania Aparecida V Kina
- Department of Pharmacology, Faculty of Medicine of Ribeirão Preto-USP, Ribeirão Preto, SP, 14049-900, Brazil
| | | | | |
Collapse
|
38
|
Soygüder Z. Multiple neurotransmitter receptors contribute to the spinal Fos expression. Brain Res 2005; 1033:202-9. [PMID: 15694925 DOI: 10.1016/j.brainres.2004.11.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2004] [Indexed: 10/25/2022]
Abstract
The aim of this study is to identify the receptors which could potentially mediate the activation of c-Fos. Therefore, the effects of neurotransmitter receptor agonists in the activation of c-Fos in spinal neurons were studied by intrathecal injection of excitatory amino acid (EAA) receptor agonists: N-Methyl-D-Aspartate (NMDA), (S)-alpha-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic acid (AMPA), 2-Carboxyl-3-carboxymethyl-4-isopropenylpyMidine (Kainic acid, KA), (1S-3R)-1-Aminocyclopentane-1, 3-dicarboxylacid (ACPD), and substance-P receptor (neurokinin-1) agonist, [Sar9, Met (O2)11] SP (SarMet-SP). All drugs tested activated the production of c-Fos in spinal dorsal horn neurons. AMPA was found as the most potent agonist tested producing market production of c-Fos particularly in neurons of lamina II at doses of 10 pM per 10-microl injection. At this dose, other agonists were relatively ineffective. At higher doses, AMPA significantly increased the activated cells. NMDA significantly increased c-Fos production to a marked extent only at doses above 10 nM per 10-microl injection. KA and ACPD were least potent of the excitatory amino acid agonists. Injection of SarMet-SP at doses of 1 nM activated Fos selectively in neurons of lamina I. A dose-dependent increase in number of c-Fos-positive cells was observed for AMPA, KA, ACPD, and SarMet-SP, whereas NMDA gave a very strong expression after a high dose with no dose dependency. These finding suggest that multiple neurotransmitter receptors lead to c-Fos production in spinal neurons.
Collapse
Affiliation(s)
- Zafer Soygüder
- University of Yüzüncü Yil, Veterinary Faculty, Department of Anatomy, Van, Turkey.
| |
Collapse
|
39
|
Song SO, Seok JH, Lee DH, Park DP, Kim SY, Lim JS, Song SK, Lee NH. Effects of Naloxone on Morphine Analgesia and Spinal c-fos Expression in Rat Formalin Test. Korean J Pain 2005. [DOI: 10.3344/kjp.2005.18.2.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Sun Ok Song
- Department of Anesthesiology and Pain Medicine, College of Medicine, Yeungnam University, Daegu, Korea
| | - Je Hong Seok
- Department of Anesthesiology and Pain Medicine, College of Medicine, Yeungnam University, Daegu, Korea
| | - Deok Hee Lee
- Department of Anesthesiology and Pain Medicine, College of Medicine, Yeungnam University, Daegu, Korea
| | - Dae Pal Park
- Department of Anesthesiology and Pain Medicine, College of Medicine, Yeungnam University, Daegu, Korea
| | - Seong Yong Kim
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, Daegu, Korea
| | - Jeong Sook Lim
- Department of Biochemistry & Molecular Biology, College of Medicine, Yeungnam University, Daegu, Korea
| | - Sun Kyo Song
- Department of Surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Nam Hyuk Lee
- Department of Surgery, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|
40
|
Wei H, Chen Y, Hong Y. The contribution of peripheral 5-hydroxytryptamine2A receptor to carrageenan-evoked hyperalgesia, inflammation and spinal Fos protein expression in the rat. Neuroscience 2005; 132:1073-82. [PMID: 15857711 DOI: 10.1016/j.neuroscience.2004.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Revised: 11/29/2004] [Accepted: 12/05/2004] [Indexed: 11/17/2022]
Abstract
The present study was conducted to test the hypothesis that the peripheral 5-hydroxytryptamine (5-HT)2A receptor is involved in inflammatory hyperalgesia and production of noxious stimulus-induced neuronal activity at the level of the spinal cord dorsal horn. Intraplantar (i.pl.) injection of carrageenan dramatically reduced paw withdrawal latency to noxious heat (47 degrees C) and caused paw swelling. Pretreatment with ketanserin, a selective antagonist of 5-HT2A receptor, in the hindpaw produced dose-dependent inhibition of the hyperalgesia (0.5, 3 and 5 mug; i.pl.) with full relief at 5 mug. The drug also moderately reduced carrageenan-induced paw swelling in a dose-dependent manner. Carrageenan induced conspicuous expression of c-fos-like immunoreactivity (FLI) in the spinal dorsal horn of segments L4-5. Ketanserin (5 mug) markedly reduced carrageenan-induced FLI in all laminae of the dorsal horn. However, blockade of peripheral 5-HT1A receptors by (N-2-[4-(2-methoxyphenyl-1-piperazinyl] ethyl]-N-2-pyridinylcyclohexanecarboxamide at maximally effective doses (30 and 100 mug; i.pl.) did not alter carrageenan-induced hyperalgesia, edema or expression of FLI. The present study provided evidence at cellular level that the peripheral 5-HT2A receptor is preferentially involved in the development of thermal hyperalgesia in the carrageenan model of inflammation.
Collapse
Affiliation(s)
- H Wei
- Department of Anatomy and Physiology, Bioengineering School, Fujian Normal University, Fuzhou, People's Republic of China
| | | | | |
Collapse
|
41
|
Zeng X, Huang H, Hong Y. Effects of intrathecal BAM22 on noxious stimulus-evoked c-fos expression in the rat spinal dorsal horn. Brain Res 2004; 1028:170-9. [PMID: 15527742 DOI: 10.1016/j.brainres.2004.09.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2004] [Indexed: 01/28/2023]
Abstract
The effects of bovine adrenal medulla 22 (BAM22), a cleaved product of proenkephalin A, were investigated on the noxious stimulus-evoked expressions of spinal c-fos-like immunoreactivity (FLI). Heat (51 degrees C) applied to the tail evoked FLI predominantly in laminae I-II of the sacral spinal cord. Intrathecal (i.t.) BAM22 at a dose of 7 nmol decreased the expressions of the heat-evoked FLI by 68%, 64% and 56% in laminae I-II, III-IV and V-VI, respectively, and the decrease pattern was comparable to that induced by i.t. morphine (10 mug). Naloxone (1 mg/kg, i.p.) significantly enhanced the heat-evoked FLI in laminae III-VI, prevented the morphine-induced inhibition, and decreased the potencies of BAM22 in laminae I-II and V-VI by 23-40%. Higher dose of naloxone (10 mg/kg, i.p.) also partially reduced the BAM22-induced suppression. Following intraplantar injection of formalin (2.5%), FLI neurons were preferentially distributed not only in laminae I-II but also in laminae III-IV and V-VI of segments L4-L5. Pretreatment with BAM22 (7 nmol, i.t.) reduced the formalin-evoked FLI neurons by 72%, 61% and 58%, in laminae I-II, III-IV and V-VI, respectively. Naloxone (1 mg/kg. i.p.) enhanced the formalin-evoked expressions of FLI in laminae III-VI and decreased the potencies of BAM22 by 22-38% in laminae I-II and V-VI. The present study provided evidence at a cellular level showing that opioid and non-opioid effects of BAM22 on nociceptive processing in acute and persistent pain models were associated with modulation of noxious stimulus-evoked activity of the spinal dorsal horn neurons.
Collapse
Affiliation(s)
- Xueai Zeng
- Department of Anatomy and Physiology, Bioengineering School, Fujian Normal University, Fuzhou, People's Republic of China
| | | | | |
Collapse
|
42
|
Zhao CS, Tao YX, Tall JM, Donovan DM, Meyer RA, Raja SN. Role of micro-opioid receptors in formalin-induced pain behavior in mice. Exp Neurol 2004; 184:839-45. [PMID: 14769376 DOI: 10.1016/s0014-4886(03)00346-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2003] [Revised: 07/03/2003] [Accepted: 07/09/2003] [Indexed: 10/26/2022]
Abstract
Intraplantar formalin injection is widely used as an experimental model of tonic pain. We investigated the role of endogenous micro-opioid receptor mechanisms in formalin-induced nocifensive behavior in mice. The flinching response induced by formalin (2%, 20 microl) was studied in mice with normal (wild type, n = 8) and absent (homozygous micro-opioid receptor knockout, n = 8) micro-opioid receptor levels. The flinch responses were counted every 5 min for 60 min post-formalin injection. Lumbar spinal cord (L4, 5) was harvested 2 h post-formalin injection to examine c-Fos expression using immunohistochemistry. The effects of naloxone (5 mg/kg, sc) administered 30 min before the intraplantar formalin injection on the flinching response of wild-type mice (n = 7) were also recorded. The second-phase formalin response (10-60 min after formalin) was higher in homozygous micro-opioid receptor knockout mice compared to the wild-type mice (P < 0.01). Naloxone administration in wild-type mice before formalin injection resulted in pain behavior similar to that observed in homozygous micro-opioid receptor knockout mice (P > 0.05). The c-Fos expression induced by formalin injection in the knockout mice was not different from that observed in wild-type mice. Our results suggest that the endogenous micro-opioid system is activated by intraplantar formalin injection and exerts a tonic inhibitory effect on the pain behavior. These results suggest an important modulatory role of endogenous micro-opioid receptor mechanisms in tonic pain states.
Collapse
Affiliation(s)
- Cheng-Shui Zhao
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
43
|
Kim EH, Hoge SG, Lightner AM, Grady EF, Coelho AM, Kirkwood KS. Activation of nociceptive neurons in T9 and T10 in cerulein pancreatitis. J Surg Res 2004; 117:195-201. [PMID: 15047123 DOI: 10.1016/s0022-4804(03)00123-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2002] [Indexed: 10/26/2022]
Abstract
Mechanisms of pain transduction in acute pancreatitis are poorly understood. Increased Fos expression in the spinal cord is a marker of activation of nociceptive neurons. We hypothesized that cerulein pancreatitis leads to increased Fos expression at T9 and T10, which receive sensory input from the pancreas. Rats were injected with cerulein (100 microg/kg, s.c.) or saline carrier (NS). Endpoints at 4, 6, and 10 h were serum amylase, myeloperoxidase activity (MPO), and spinal cord Fos expression (number of immunoreactive nuclei/section dorsal gray matter). Fos-like immunoreactivity (FLI) at T9-T10 was compared to internal controls (T6, T12). An average of 20 spinal cord histologic sections were evaluated per rat. Some animals were injected with the mu-opioid receptor agonist, buprenorphine (90 microg/kg, s.c.), 3 h after cerulein, and their endpoints were measured at 6 h. Analysis of variance and t tests were used for statistical analysis. Results are means +/- SEM. As expected, cerulein induced edematous pancreatitis, with a 4-fold increase in serum amylase at 6 h [cer (n = 8): 14,000 +/- 1,300 U/ml versus NS (n = 10): 3,700 +/- 300, P < 0.005)] and a 2-fold increase in MPO activity (0.25 +/- 0.05) activity units/dry wt versus 0.13 +/- 0.02, P < 0.05). Cerulein induced nearly a 2-fold increase in FLI at T9 and T10 [n = 10 (cer) and n = 13 (NS): T9, 14 +/- 1.5 versus 7.8 +/- 0.88; T10, 15 +/- 1.7 versus 8.3 +/- 0.70; P < 0.05]. Peak effects of cerulein on FLI occurred at 6 h and were greatest at T9/T10 with relative sparing of T6/T12. T6/T12 expression was similar in experimental and control groups. Buprenorphine significantly reduced both serum amylase and FLI and T9/T10. Cerulein-induced acute pancreatitis in rat increases visceral nociceptive signaling at spinal cord levels T9 and T10, with a peak at 6 h. Blockade of this effect by the mu-opioid receptor agonist buprenorphine could occur either by direct activation of central opioid receptors and/or an anti-inflammatory mechanism. FLI is a useful tool for studying the pathophysiology of pain in experimental acute pancreatitis.
Collapse
Affiliation(s)
- Edward H Kim
- Department of Surgery, University of California-San Francisco, 533 Parnassus, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
44
|
Bon K, Wilson SG, Mogil JS, Roberts WJ. Genetic evidence for the correlation of deep dorsal horn Fos protein immunoreactivity with tonic formalin pain behavior. THE JOURNAL OF PAIN 2003; 3:181-9. [PMID: 14622771 DOI: 10.1054/jpai.2002.123710] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The formalin test is commonly used as a model of persistent pain. Besides producing pain behavior, hind paw formalin injection induces the expression of the immediate-early gene, c-fos. A current controversy is whether noxious stimulus-induced Fos protein immunoreactivity can be considered a proxy (biomarker) of nociception in the spinal cord. We investigated this issue by exploiting our recent demonstration of genotype-dependent behavioral differences in response to formalin injection among inbred mouse strains. Accordingly, 6 inbred and 2 outbred strains were administered formalin (5% in 25 microL) into the ventral hind paw, monitored for licking behavior, and then sacrificed at 90 minutes after injection for Fos protein immunocytochemistry. Significant strain differences were observed in both licking behavior and Fos counts in superficial and deep laminae. We observed a significant correlation among strains between licking behavior in the late phase (10 to 60 minutes) of the formalin test and Fos expression in laminae V-VI (but not laminae I-II) of the dorsal horn (r = 0.94). These findings reinforce the use of the Fos technique to study the neuronal processing underlying pain but suggest that Fos labeling reliably reflects tonic pain behavior only in neurons located in the neck of the dorsal horn in mice.
Collapse
Affiliation(s)
- Karine Bon
- Department of Psychology and Neuroscience Program, University of Illinois at Urbana-Champaign, USA
| | | | | | | |
Collapse
|
45
|
Tavares I, Lima D. The caudal ventrolateral medulla as an important inhibitory modulator of pain transmission in the spinal cord. THE JOURNAL OF PAIN 2003; 3:337-46. [PMID: 14622734 DOI: 10.1054/jpai.2002.127775] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The caudal ventrolateral medulla (VLM) has emerged during the last decade as one of the main components of the endogenous pain control system. Profound and long-lasting analgesia is produced by mild stimulation of the VLM. The VLMlat, the reticular formation located between the spinal trigeminal nucleus and the lateral reticular nucleus (LRt), appears to play a major role in that antinociceptive action. The projections to spinal cord laminae involved in nociceptive transmission originate exclusively in the VLMlat. The VLMlat participates in a disynaptic pathway involving spinally projecting pontine A5 noradrenergic neurons, which appears to convey alpha(2)-adrenoreceptor-mediated analgesia produced from the VLM. Neurons in the VLMlat and in lamina I are reciprocally connected by a closed loop that is likely to mediate feedback control of supraspinal nociceptive transmission. On the other hand, the LRt, which is targeted by ventral (lamina VII) and deep dorsal (laminae IV to V) horn inputs, projects to the premotor lamina VII. Nociceptive input ascending from the cord and increases in blood pressure are discussed as possible physiologic triggers of the analgesia produced by the VLM. The overall role of the VLM as a center for integration of nociceptive, cardiovascular, and motor functions is discussed. The putative therapeutic benefits of manipulating the VLM for the control of chronic pain are envisaged.
Collapse
Affiliation(s)
- Isaura Tavares
- Institute of Histology and Embryology, Faculty of Medicine and IBMC, University of Porto, Portugal.
| | | |
Collapse
|
46
|
Hartwig AC, Mathias SI, Law AS, Gebhart GF. Characterization and opioid modulation of inflammatory temporomandibular joint pain in the rat. J Oral Maxillofac Surg 2003; 61:1302-9. [PMID: 14613087 DOI: 10.1016/s0278-2391(03)00732-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PURPOSE Experimental inflammation of the rat temporomandibular joint (TMJ) is commonly used to study trigeminal nociceptive processing. This study describes spontaneous pain-related behaviors following TMJ inflammation in the rat. The ability of preemptive systemic morphine to attenuate behaviors as well as immediate-early gene expression in the trigeminal nucleus is described. MATERIALS AND METHODS Adult male Sprague-Dawley rats received an intra-articular injection of mustard oil (0% to 20%, 50 microL) and were observed for behavioral changes. Morphine sulfate (0 to 10 mg/kg SC) was given 30 minutes before mustard oil; this was reversed in one group with naltrexone hydrochloride (5 mg/kg SC). Two hours after injection rats were killed and perfused. Immunohistochemistry for the protein product of the immediate-early gene c-fos was performed, and brain stem sections including the trigeminal subnucleus caudalis were examined for positive nuclei. RESULTS Mustard oil inflammation of the rat TMJ induces dose-dependent, morphine-sensitive behaviors. Behaviors observed included excessive grooming of the region, a chewing-like behavior, and head shaking. Fos expression in the trigeminal subnucleus caudalis parallels changes in behaviors. Morphine dose dependently attenuates the number of behaviors, as well as Fos expression; this effect is reversed by the micro-opioid receptor antagonist naltrexone. CONCLUSIONS Mustard oil inflammation of the rat TMJ causes reliable behavioral changes, which may be quantified and, together with Fos expression, used to assess various experimental TMJ treatment modalities.
Collapse
Affiliation(s)
- Andrew C Hartwig
- Department of Pharmacology, University of Iowa College of Medicine, Iowa City, IA 52242-1109, USA
| | | | | | | |
Collapse
|
47
|
Engström L, Engblom D, Blomqvist A. Systemic immune challenge induces preproenkephalin gene transcription in distinct autonomic structures of the rat brain. J Comp Neurol 2003; 462:450-61. [PMID: 12811812 DOI: 10.1002/cne.10770] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The involvement of enkephalins in the immune response was investigated in rats injected intravenously with interleukin-1beta (2 microg/kg). In situ hybridization with a riboprobe complementary to intron A of the preproenkephalin (ppENK) gene showed distinct transcriptional activation within several brain regions known to be activated by immune stimuli, including the nucleus of the solitary tract, the area postrema, the paraventricular hypothalamic nucleus, and the oval nucleus of the bed nucleus of the stria terminalis, and dual labeling confirmed that a large proportion of the intron expressing neurons co-expressed c-fos mRNA. Rats injected with saline (controls) showed little or no heteronuclear transcript in these structures. The induced signal was strongest after 1 hour but was present in some structures 30 minutes after interleukin-1beta injection. At 3 hours, transcriptional activity returned to basal levels. High basal expression of the heteronuclear transcript that appeared unchanged by the immune stimulus was seen in regions not primarily involved in the immune response, such as the striatum, the olfactory tubercle, and the islands of Calleja and in the immune activated central nucleus of the amygdala. The heteronuclear transcript colocalized with ppENK mRNA, demonstrating that it occurred in enkephalinergic neurons and was not the result of alternative transcription from the ppENK gene in other cells. These results demonstrated that enkephalin transcription is induced in central autonomic neurons during immune challenge, suggesting that enkephalins are involved in the centrally orchestrated response to such stimuli.
Collapse
Affiliation(s)
- Linda Engström
- Department of Biomedicine and Surgery, Division of Cell Biology, Faculty of Health Sciences, University of Linköping, SE-581 85 Linköping, Sweden
| | | | | |
Collapse
|
48
|
Kim HW, Kwon YB, Ham TW, Roh DH, Yoon SY, Lee HJ, Han HJ, Yang IS, Beitz AJ, Lee JH. Acupoint stimulation using bee venom attenuates formalin-induced pain behavior and spinal cord fos expression in rats. J Vet Med Sci 2003; 65:349-55. [PMID: 12679565 DOI: 10.1292/jvms.65.349] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In two previous reports, we have demonstrated that injection of bee venom (BV) into an acupoint produces a significant antinociceptive and anti-inflammatory effect in both a mouse model of visceral nociception and a rat model of chronic arthritis. The present study was designed to evaluate the potential antinociceptive effect of BV pretreatment on formalin-induced pain behavior and it associated spinal cord Fos expression in rats. Adult Sprague-Dawley rats were injected with BV directly into the Zusanli (ST36) acupoint or into an arbitrary non-acupoint located on the back. BV pretreatment into the Zusanli acupoint significantly decreased paw-licking time in the late phase of the formalin test. In contrast, BV injected into a non-acupoint in the back region did not suppress the paw-licking time. In addition, BV pretreatment into the Zusanli acupoint markedly inhibited spinal cord Fos expression induced by formalin injection. These findings indicate that BV pretreatment into the Zusanli acupoint has an antinociceptive effect on formalin-induced pain behavior.
Collapse
Affiliation(s)
- Hyun-Woo Kim
- Department of Veterinary Physiology, College of Veterinary Medicine and School of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gilbert AK, Franklin KBJ. The role of descending fibers from the rostral ventromedial medulla in opioid analgesia in rats. Eur J Pharmacol 2002; 449:75-84. [PMID: 12163109 DOI: 10.1016/s0014-2999(02)01974-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
There has been controversy as to whether the contribution of descending fibers from the rostral ventromedial medulla to opioid analgesia depends on the nature of the noxious stimulus eliciting pain. In the present study, inactivation of descending fibers by microinjection of muscimol (50 ng) in the rostral ventromedial medulla abolished morphine analgesia in the tail immersion and hot plate tests but decreased morphine analgesia by 60% in the formalin test. Analysis of the dose-response relation for morphine after inactivation of descending fibers revealed that, except for the tail immersion test, high doses of morphine could not overcome the block induced by muscimol. Also, morphine analgesia elicited supraspinally was not detectable when descending fibers were inactivated, suggesting that the analgesic effect of morphine in the brain requires a relay via the rostral ventromedial medulla. The analgesic effect of buprenorphine also depends on the integrity of descending fibers from the rostral ventromedial medulla. The results indicate that descending fibers from the rostral ventromedial medulla are critically important to the analgesic effect of opioids, regardless of the type of noxious stimulation eliciting pain. Residual analgesic effects of opioids after inactivation of descending fibers may be due to peripheral effects in the presence of inflammation.
Collapse
Affiliation(s)
- Annie-Kim Gilbert
- Department of Psychology, McGill University, 1205 Docteur Penfield Avenue, Montreal, Quebec, Canada H3A 1B1.
| | | |
Collapse
|
50
|
Rahman OIF, Terayama R, Ikeda T, Koganemaru M, Nakamura T, Shiba R, Nishimori T. Differential effects of NMDA and AMPA/KA receptor antagonists on c-Fos or Zif/268 expression in the rat spinal dorsal horn induced by noxious thermal or mechanical stimulation, or formalin injection. Neurosci Res 2002; 43:389-99. [PMID: 12135782 DOI: 10.1016/s0168-0102(02)00067-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The involvement of N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA)/kainate (KA) receptors in the induction of c-Fos and Zif/268 expression in spinal dorsal horn neurons following noxious thermal or mechanical stimulation, or formalin injection into the rat hind paw was examined by intrathecal administration of a competitive NMDA receptor antagonist, 2-amino-5-phosphonopentanoic acid (APV) or an AMPA/KA receptor antagonist, 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), or both, 30 min prior to noxious stimulation. APV caused a significant reduction in the level of c-Fos expression in the superficial layer induced by each of these three noxious stimuli. The effects of APV on Zif/268 expression or of CNQX on c-Fos or Zif/268 expression in the superficial layer induced by these three noxious stimuli were dependent on the type of stimulus applied to the rat hind paw. The noxious thermal stimulus-evoked c-Fos expression level was reduced by APV and/or CNQX, while Zif/268 expression was hardly changed. Both c-Fos and Zif/268 expressions following formalin injection were reduced by APV alone and APV+CNQX, but not by CNQX alone. Zif/268 expression following noxious mechanical stimulation was significantly reduced only by APV+CNQX although APV or CNQX alone did not affect the expression, while c-Fos expression was reduced by APV and APV+CNQX but not by CNQX alone. These findings suggest that NMDA and AMPA/KA receptors are differentially involved in c-Fos and Zif/268 expression in the spinal dorsal horn following noxious thermal, formalin and mechanical stimulation.
Collapse
Affiliation(s)
- Omar I F Rahman
- Department of Oral and Maxillofacial Surgery, Miyazaki Medical College, Kiyotake, Miyazaki 889-1692, Japan
| | | | | | | | | | | | | |
Collapse
|