1
|
Kummer K, Sheets PL. Targeting Prefrontal Cortex Dysfunction in Pain. J Pharmacol Exp Ther 2024; 389:268-276. [PMID: 38702195 PMCID: PMC11125798 DOI: 10.1124/jpet.123.002046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
The prefrontal cortex (PFC) has justifiably become a significant focus of chronic pain research. Collectively, decades of rodent and human research have provided strong rationale for studying the dysfunction of the PFC as a contributing factor in the development and persistence of chronic pain and as a key supraspinal mechanism for pain-induced comorbidities such as anxiety, depression, and cognitive decline. Chronic pain alters the structure, chemistry, and connectivity of PFC in both humans and rodents. In this review, we broadly summarize the complexities of reported changes within both rodent and human PFC caused by pain and offer insight into potential pharmacological and nonpharmacological approaches for targeting PFC to treat chronic pain and pain-associated comorbidities. SIGNIFICANCE STATEMENT: Chronic pain is a significant unresolved medical problem causing detrimental changes to physiological, psychological, and behavioral aspects of life. Drawbacks of currently approved pain therapeutics include incomplete efficacy and potential for abuse producing a critical need for novel approaches to treat pain and comorbid disorders. This review provides insight into how manipulation of prefrontal cortex circuits could address this unmet need of more efficacious and safer pain therapeutics.
Collapse
Affiliation(s)
- Kai Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria (K.K.); Department of Pharmacology and Toxicology (P.L.S.), Medical Neurosciences Graduate Program (P.L.S.), and Stark Neurosciences Research Institute (P.L.S.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Patrick L Sheets
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria (K.K.); Department of Pharmacology and Toxicology (P.L.S.), Medical Neurosciences Graduate Program (P.L.S.), and Stark Neurosciences Research Institute (P.L.S.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
2
|
Moura-Pacheco TL, Martins-Pereira RC, Medeiros P, Sbragia L, Ramos Andrade Leite-Panissi C, Machado HR, Coimbra NC, de Freitas RL. Effect of electrical and chemical (activation versus inactivation) stimulation of the infralimbic division of the medial prefrontal cortex in rats with chronic neuropathic pain. Exp Brain Res 2023; 241:2591-2604. [PMID: 37725136 DOI: 10.1007/s00221-023-06657-y] [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: 04/23/2023] [Accepted: 06/20/2023] [Indexed: 09/21/2023]
Abstract
Neuropathic pain (NP) represents a complex disorder with sensory, cognitive, and emotional symptoms. The medial prefrontal cortex (mPFC) takes critical regulatory roles and may change functionally and morphologically during chronic NP. There needs to be a complete understanding of the neurophysiological and psychopharmacological bases of the NP phenomenon. This study aimed to investigate the participation of the infralimbic division (IFL) of the mPFC in chronic NP, as well as the role of the N-methyl-D-aspartic acid receptor (NMDAr) in the elaboration of chronic NP. Male Wistar rats were submitted to the von Frey and acetone tests to assess mechanical and cold allodynia after 21 days of chronic constriction injury (CCI) of the sciatic nerve or Sham-procedure ("false operated"). Electrical neurostimulation of the IFL/mPFC was performed by low-frequency stimuli (20 μA, 100 Hz) applied for 15 s by deep brain stimulation (DBS) device 21 days after CCI. Either cobalt chloride (CoCl2 at 1.0 mM/200 nL), NMDAr agonist (at 0.25, 1.0, and 2.0 nmol/200 nL) or physiological saline (200 nL) was administered into the IFL/mPFC. CoCl2 administration in the IFL cortex did not alter either mechanical or cold allodynia. DBS stimulation of the IFL cortex decreased mechanical allodynia in CCI rats. Chemical stimulation of the IFL cortex by an NMDA agonist (at 2.0 nmol) decreased mechanical allodynia. NMDA at any dose (0.25, 1.0, and 2.0 nmol) reduced the flicking/licking duration in the cold test. These findings suggest that the IFL/mPFC and the NMDAr of the neocortex are involved in attenuating chronic NP in rats.
Collapse
Affiliation(s)
- Thais Lohanny Moura-Pacheco
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Laboratory of Neurosciences of Pain and Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Pediatric Surgery Laboratory, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Renata Cristina Martins-Pereira
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Laboratory of Neurosciences of Pain and Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Protection Laboratory in Childhood, Division of Neurosurgery, Department of Surgery and Anatomy, FMRP-USP, Avenida Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Priscila Medeiros
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Laboratory of Neurosciences of Pain and Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Department of General and Specialized Nursing, Ribeirão Preto Nursing School of the University of São Paulo (EERP-USP), Avenida Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Lourenço Sbragia
- Pediatric Surgery Laboratory, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Christie Ramos Andrade Leite-Panissi
- Department of Psychology,, Faculty of Philosophy, Science and Letters of Ribeirão Preto of the University of São Paulo (FFCLRP-USP), Ribeirão Preto, SP, 14040-901, Brazil
| | - Hélio Rubens Machado
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Department of Psychology,, Faculty of Philosophy, Science and Letters of Ribeirão Preto of the University of São Paulo (FFCLRP-USP), Ribeirão Preto, SP, 14040-901, Brazil
| | - Norberto Cysne Coimbra
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
- Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil
| | - Renato Leonardo de Freitas
- Multi-User Center of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil.
- Laboratory of Neurosciences of Pain and Emotions, Department of Surgery and Anatomy, FMRP-USP, Av. Bandeirantes, 3900, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
3
|
Gao X, Lin J, Sun L, Hu J, Gao W, Yu J. Activation of the N-methyl-D-aspartate receptor and calcium/calmodulin-dependent protein kinase IIα signal in the rostral anterior cingulate cortex is involved in pain-related aversion in rats with peripheral nerve injury. Behav Brain Res 2023; 452:114560. [PMID: 37394125 DOI: 10.1016/j.bbr.2023.114560] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/28/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
The rostral anterior cingulate cortex (rACC) of rat brain is associated with pain-related emotions. However, the underlying molecular mechanism remains unclear. Here, we investigated the effects of the N-methyl-D-aspartate (NMDA) receptor and Ca2+/Calmodulin-dependent protein kinase type II (CaMKII)α signal on pain-related aversion in the rACC of a rat model of neuropathic pain (NP). Mechanical and thermal hyperalgesia were examined using von Frey and hot plate tests in a rat model of NP induced by spared nerve injury (SNI) of the unilateral sciatic nerve. Bilateral rACC pretreatment with the CaMKII inhibitor tat-CN21 (derived from the cell-penetrating tat sequence and CaM-KIIN amino acids 43-63) or tat-Ctrl (the tat sequence and the scrambled sequence of CN21) was performed on postoperative days 29-35 in Sham rats or rats with SNI. Spatial memory performance was tested using an eight-arm radial maze on postoperative days 34-35. Pain-related negative emotions (aversions) were evaluated using the place escape/avoidance paradigm on postoperative day 35 following the spatial memory performance test. The percentage of time spent in the light area was used to assess pain-related negative emotions (i.e., aversion). The expression levels of the NMDA receptor GluN2B subunit, CaMKIIα, and CaMKII-Threonine at position 286 (Thr286) phosphorylation in contralateral rACC specimens were detected by Western blot or real time PCR following the aversion test. Our data showed that pretreatment of the rACC with tat-CN21 increased determinate behavior but did not alter hyperalgesia or spatial memory performance in rats with SNI. In addition, tat-CN21 reversed the enhanced CaMKII-Thr286 phosphorylation and had no effect on the upregulated expression of GluN2B, CaMKIIα protein, and mRNA. Our data suggested that activation of the NMDA receptor-CaMKIIα signal in rACC is associated with pain-related aversion in rats with NP. These data may provide a new approach for the development of drugs that modulate cognitive and emotional pain aspects.
Collapse
Affiliation(s)
- Xueqi Gao
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Jinhai Lin
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Lin Sun
- School of Psychology, Weifang Medical University, Weifang 261053, China
| | - Jun Hu
- Department of Anesthesiology, Weifang People's Hospital, Weifang 261044, China
| | - Wenjie Gao
- Department of Anesthesiology, Weifang People's Hospital, Weifang 261044, China
| | - Jianfeng Yu
- Experimental Center for Medical Research, School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
4
|
A randomized pharmacological fMRI trial investigating D-cycloserine and brain plasticity mechanisms in learned pain responses. Sci Rep 2022; 12:19080. [PMID: 36351953 PMCID: PMC9646732 DOI: 10.1038/s41598-022-23769-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Learning and negative outcome expectations can increase pain sensitivity, a phenomenon known as nocebo hyperalgesia. Here, we examined how a targeted pharmacological manipulation of learning would impact nocebo responses and their brain correlates. Participants received either a placebo (n = 27) or a single 80 mg dose of D-cycloserine (a partial NMDA receptor agonist; n = 23) and underwent fMRI. Behavioral conditioning and negative suggestions were used to induce nocebo responses. Participants underwent pre-conditioning outside the scanner. During scanning, we first delivered baseline pain stimulations, followed by nocebo acquisition and extinction phases. During acquisition, high intensity thermal pain was paired with supposed activation of sham electrical stimuli (nocebo trials), whereas moderate pain was administered with inactive electrical stimulation (control trials). Nocebo hyperalgesia was induced in both groups (p < 0.001). Nocebo magnitudes and brain activations did not show significant differences between D-cycloserine and placebo. In acquisition and extinction, there were significantly increased activations bilaterally in the amygdala, ACC, and insula, during nocebo compared to control trials. Nocebo acquisition trials also showed increased vlPFC activation. Increased opercular activation differentiated nocebo-augmented pain aggravation from baseline pain. These results support the involvement of integrative cognitive-emotional processes in nocebo hyperalgesia.
Collapse
|
5
|
Kanno K, Suzuki-Narita M, Kawarai Y, Hagiwara S, Yoh S, Nakamura J, Orita S, Inage K, Suzuki T, Ohtori S. Analgesic effects and arthritic changes following tramadol administration in a rat hip osteoarthritis model. J Orthop Res 2022; 40:1770-1777. [PMID: 34783063 DOI: 10.1002/jor.25208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/16/2021] [Accepted: 11/01/2021] [Indexed: 02/04/2023]
Abstract
We investigated the analgesic effects of tramadol and the arthritic changes following tramadol administration in the rat hip osteoarthritis (OA) model using mono-iodoacetate (MIA). The right hip joints of male Sprague-Dawley rats (n = 5 rats/group) in the Sham group were injected with 25 μl of sterile saline and 1% of fluorogold (FG) retrograde neurotracer. In the MIA + Vehicle and MIA + Tramadol groups, FG and 25 μl of sterile saline with 0.5 mg of MIA were injected into the right hip joint. The MIA + Vehicle and MIA + Tramadol groups were administered daily for 4 weeks, either sterile saline (10 mg/kg, intraperitoneal [i.p.]) or tramadol (10 mg/kg, i.p.). We assessed hyperalgesia every week after MIA administration. Histopathological changes and immunoreactive neurons for calcitonin gene-related peptide (CGRP) in dorsal root ganglia (DRG) were evaluated after 4 weeks of treatment. MIA injection into the hip joint led to mechanical hyperalgesia (p < 0.01), which was significantly reduced by tramadol administration (p < 0.01). Furthermore, daily i.p injection of tramadol significantly suppressed CGRP expression in DRG (p < 0.0001). MIA + Vehicle and MIA + Tramadol groups showed significant cartilage reduction and degeneration compared to the Sham group (p < 0.0001). Interestingly, OA changes significantly progressed in the MIA + Tramadol group compared to the MIA + Vehicle group (p < 0.0001).
Collapse
Affiliation(s)
- Keijiro Kanno
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Miyako Suzuki-Narita
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Yuya Kawarai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Shigeo Hagiwara
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Satoshi Yoh
- Department of Orthopaedic Surgery, Eastern Chiba Medical Center, Togane, Japan
| | - Junichi Nakamura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Sumihisa Orita
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Kazuhide Inage
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Takane Suzuki
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba City, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, Chiba City, Japan
| |
Collapse
|
6
|
Burgdorf JS, Zhang XL, Stanton PK, Moskal JR, Donello JE. Zelquistinel Is an Orally Bioavailable Novel NMDA Receptor Allosteric Modulator That Exhibits Rapid and Sustained Antidepressant-Like Effects. Int J Neuropsychopharmacol 2022; 25:979-991. [PMID: 35882204 PMCID: PMC9743962 DOI: 10.1093/ijnp/pyac043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/22/2022] [Accepted: 07/25/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The role of glutamatergic receptors in major depressive disorder continues to be of great interest for therapeutic development. Recent studies suggest that both negative and positive modulation of N-methyl-D-aspartate receptors (NMDAR) can produce rapid antidepressant effects. Here we report that zelquistinel, a novel NMDAR allosteric modulator, exhibits high oral bioavailability and dose-proportional exposures in plasma and the central nervous system and produces rapid and sustained antidepressant-like effects in rodents by enhancing activity-dependent, long-term synaptic plasticity. METHODS NMDAR-mediated functional activity was measured in cultured rat brain cortical neurons (calcium imaging), hNR2A or B subtype-expressing HEK cells, and synaptic plasticity in rat hippocampal and medial prefrontal cortex slices in vitro. Pharmacokinetics were evaluated in rats following oral administration. Antidepressant-like effects were assessed in the rat forced swim test and the chronic social deficit mouse model. Target engagement and the safety/tolerability profile was assessed using phencyclidine-induced hyperlocomotion and rotarod rodent models. RESULTS Following a single oral dose, zelquistinel (0.1-100 µg/kg) produced rapid and sustained antidepressant-like effects in the rodent depression models. Brain/ cerebrospinal fluid concentrations associated with zelquistinel antidepressant-like activity also increased NMDAR function and rapidly and persistently enhanced activity-dependent synaptic plasticity (long-term potentiation), suggesting that zelquistinel produces antidepressant-like effects by enhancing NMDAR function and synaptic plasticity. Furthermore, Zelquistinel inhibited phencyclidine (an NMDAR antagonist)-induced hyperlocomotion and did not impact rotarod performance. CONCLUSIONS Zelquistinel produces rapid and sustained antidepressant effects by positively modulating the NMDARs, thereby enhancing long-term potentiation of synaptic transmission.
Collapse
Affiliation(s)
- Jeffrey S Burgdorf
- Correspondence: Jeffrey Burgdorf, PhD, 1801 Maple Ave, Suite 4300, Evanston, IL, 60201, USA ()
| | - Xiao-Lei Zhang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Patric K Stanton
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, USA
| | - Joseph R Moskal
- Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, McCormick School of Engineering and Applied Sciences, Northwestern University, Evanston, Illinois, USA
| | | |
Collapse
|
7
|
Dai W, Huang S, Luo Y, Cheng X, Xia P, Yang M, Zhao P, Zhang Y, Lin WJ, Ye X. Sex-Specific Transcriptomic Signatures in Brain Regions Critical for Neuropathic Pain-Induced Depression. Front Mol Neurosci 2022; 15:886916. [PMID: 35663269 PMCID: PMC9159910 DOI: 10.3389/fnmol.2022.886916] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
Neuropathic pain is a chronic debilitating condition with a high comorbidity with depression. Clinical reports and animal studies have suggested that both the medial prefrontal cortex (mPFC) and the anterior cingulate cortex (ACC) are critically implicated in regulating the affective symptoms of neuropathic pain. Neuropathic pain induces differential long-term structural, functional, and biochemical changes in both regions, which are thought to be regulated by multiple waves of gene transcription. However, the differences in the transcriptomic profiles changed by neuropathic pain between these regions are largely unknown. Furthermore, women are more susceptible to pain and depression than men. The molecular mechanisms underlying this sexual dimorphism remain to be explored. Here, we performed RNA sequencing and analyzed the transcriptomic profiles of the mPFC and ACC of female and male mice at 2 weeks after spared nerve injury (SNI), an early time point when the mice began to show mild depressive symptoms. Our results showed that the SNI-induced transcriptomic changes in female and male mice were largely distinct. Interestingly, the female mice exhibited more robust transcriptomic changes in the ACC than male, whereas the opposite pattern occurred in the mPFC. Cell type enrichment analyses revealed that the differentially expressed genes involved genes enriched in neurons, various types of glia and endothelial cells. We further performed gene set enrichment analysis (GSEA), which revealed significant de-enrichment of myelin sheath development in both female and male mPFC after SNI. In the female ACC, gene sets for synaptic organization were enriched, and gene sets for extracellular matrix were de-enriched after SNI, while such signatures were absent in male ACC. Collectively, these findings revealed region-specific and sexual dimorphism at the transcriptional levels induced by neuropathic pain, and provided novel therapeutic targets for chronic pain and its associated affective disorders.
Collapse
Affiliation(s)
- Weiping Dai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuying Huang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Luo
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Pei Xia
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengqian Yang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Panwu Zhao
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yingying Zhang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wei-Jye Lin
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaojing Ye,
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Wei-Jye Lin,
| |
Collapse
|
8
|
Barroso J, Branco P, Apkarian AV. Brain mechanisms of chronic pain: critical role of translational approach. Transl Res 2021; 238:76-89. [PMID: 34182187 PMCID: PMC8572168 DOI: 10.1016/j.trsl.2021.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 01/15/2023]
Abstract
Chronic pain is a leading cause of disability worldwide and its prevalence is likely to increase over the next decades. Treatment for chronic pain remains insufficient and therapeutical advances have not made comparable progress with that for many chronic disorders, thus amplifying the concern on the future burden of the disease. At the same time, and even after decades of intense research, the underlying pathophysiology of chronic pain remains minimally understood. We believe advancing our current understanding of chronic pain requires mechanistically explicit, hypothesis-driven, and clinically focused models. In this review we highlight some of the main findings over the last decades that have contributed to the present knowledge of brain mechanisms of chronic pain, and how such advances were possible due to a reverse translational research approach. We argue that this approach is essential in the chronic pain field, in order to generate new scientific hypotheses, probe physiological mechanisms, develop therapeutic strategies and translate findings back into promising human clinical trials.
Collapse
Affiliation(s)
- Joana Barroso
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Physical Medicine and Rehabilitation, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Center for Chronic Pain and Drug Abuse, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Paulo Branco
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Center for Chronic Pain and Drug Abuse, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Apkar Vania Apkarian
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Physical Medicine and Rehabilitation, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Center for Chronic Pain and Drug Abuse, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Anesthesiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
9
|
Phelps CE, Navratilova E, Porreca F. Chronic Pain Produces Reversible Memory Deficits That Depend on Task Difficulty in Rats. THE JOURNAL OF PAIN 2021; 22:1467-1476. [PMID: 34023503 PMCID: PMC8578143 DOI: 10.1016/j.jpain.2021.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/05/2021] [Accepted: 04/30/2021] [Indexed: 11/29/2022]
Abstract
Cognitive impairment associated with chronic pain remains relatively poorly understood. Use of analgesic drugs and often present co-morbidities in patients can preclude conclusions of causative relationships between chronic pain and cognitive deficits. Here, the impact of pain resulting from spinal nerve ligation (SNL) injury in rats on short and long-term memory was assessed in the novel object recognition task. To understand if chronic pain seizes the limited cognitive resources that are available at any given time, task difficulty was varied by using either very different (ie, easy task) or similar (ie, difficult task) pairs of objects. Nerve-injured, male rats exhibited no short or long-term memory deficits under easy task conditions. However, unlike sham-operated controls, injured rats showed deficits in both short and long-term memory by failing to differentiate similar objects in the difficult task version. In SNL rats, duloxetine produced anti-allodynic effects and ameliorated long-term memory deficits in the difficult task suggesting benefits of pain relief possibly complemented by noradrenergic mediated cognitive enhancement. Together these data suggest chronic pain reversibly takes up a significant amount of limited cognitive resources, leaving sufficient available for easy, but not difficult, tasks. PERSPECTIVE: Memory deficits in a rat model of chronic pain were only seen when the cognitive load was high, ie, in a difficult task. Acute treatment with duloxetine was sufficient to relieve memory deficits, suggesting chronic pain induces memory deficits by seizing limited cognitive resources to the detriment of task-related stimuli.
Collapse
Affiliation(s)
- Caroline E Phelps
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona
| | - Edita Navratilova
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona
| | - Frank Porreca
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, Arizona.
| |
Collapse
|
10
|
Jiang M, Chen X, Zhang L, Liu W, Yu X, Wang Z, Zheng M. Electroacupuncture suppresses glucose metabolism and GLUT-3 expression in medial prefrontal cortical in rats with neuropathic pain. Biol Res 2021; 54:24. [PMID: 34362470 PMCID: PMC8344173 DOI: 10.1186/s40659-021-00348-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 07/30/2021] [Indexed: 11/10/2022] Open
Abstract
Background Accumulating evidence has demonstrated that the electroacupuncture (EA) stimulation could effectively alleviate neuropathic pain. The medial prefrontal cortex (mPFC) is a vital part of the cortical representation of pain in the brain, and its glucose metabolism is mostly affected in the progression of pain. However, the central mechanism of EA analgesia remains unclear. Methods Fifty-four male SD rats were equally randomized into sham surgery (Sham) group, chronic constriction injury (CCI) group and EA stimulation (EA) group. The CCI model, involving ligature of the right sciatic nerve, was established in all animals except the Sham group. EA stimulation was applied on the right side acupoints of Huantiao (GB30) and Yanglingquan (GB34) in the EA group. Paw withdrawal threshold (PWT) and paw thermal withdrawal latency (PWL) were measured. The 18 F-fluorodeoxyglucose positron emission tomography (FDG-PET) was used to evaluate glucose metabolism changes in the mPFC. The expression of glucose transporter 3 (GLUT-3) in the mPFC was determined by immune histochemistry and ELISA. Results Comparing with CCI groups, EA treatment was obviously reversed CCI-induced mechanical allodynia (P < 0.01), thermal hyperalgesia (P < 0.01) and the increase of glucose metabolism in the left mPFC (P < 0.05). Furthermore, EA treatment significantly decreased the protein expression of GLUT-3 in the left mPFC (P < 0.01). Conclusions Our results indicate that EA analgesia effect may be related to suppressing the glucose metabolism and GLUT-3 expression in the mPFC. This study could provide a potential insight into the central mechanisms involved in the analgesic effect of EA.
Collapse
Affiliation(s)
- Menghong Jiang
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Xiaomei Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Liangping Zhang
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Weiting Liu
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Xiangmei Yu
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Zhifu Wang
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China. .,Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation, Fujian University of Chinese Medicine Affiliated Rehabilitation Hospital, Fuzhou, 350122, Fujian, China.
| | - Meifeng Zheng
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
11
|
Sex-Specific Disruption of Distinct mPFC Inhibitory Neurons in Spared-Nerve Injury Model of Neuropathic Pain. Cell Rep 2021; 31:107729. [PMID: 32521254 DOI: 10.1016/j.celrep.2020.107729] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022] Open
Abstract
The medial prefrontal cortex (mPFC) modulates a range of behaviors, including responses to noxious stimuli. While various pain modalities alter mPFC function, our understanding of changes to specific cell types underlying pain-induced mPFC dysfunction remains incomplete. Proper activity of cortical GABAergic interneurons is essential for normal circuit function. We find that nerve injury increases excitability of layer 5 parvalbumin-expressing neurons in the prelimbic (PL) region of the mPFC from male, but not female, mice. Conversely, nerve injury dampens excitability in somatostatin-expressing neurons in layer 2/3 of the PL region; however, effects are differential between males and females. Nerve injury slightly increases the frequency of spontaneous excitatory post-synaptic currents (sEPSCs) in layer 5 parvalbumin-expressing neurons in males but reduces frequency of sEPSCs in layer 2/3 somatostatin-expressing neurons in females. Our findings provide key insight into how nerve injury drives maladaptive and sex-specific alterations to GABAergic circuits in cortical regions implicated in chronic pain.
Collapse
|
12
|
Phelps CE, Navratilova E, Porreca F. Cognition in the Chronic Pain Experience: Preclinical Insights. Trends Cogn Sci 2021; 25:365-376. [PMID: 33509733 PMCID: PMC8035230 DOI: 10.1016/j.tics.2021.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 12/24/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022]
Abstract
Acutely, pain is protective. It promotes escape from, and future avoidance of, noxious stimuli through strong and often lifetime associative memories. However, with persistent acute pain or when pain becomes chronic, these memories can promote negative emotions and poor decisions often associated with deleterious behaviors. In this review, we discuss how preclinical studies can provide insights into the relationship between cognition and chronic pain. We also discuss the concept of pain as a cognitive disorder and new strategies for treating chronic pain that emphasize inhibiting the formation of pain memories or promoting 'forgetting' of established pain memories.
Collapse
Affiliation(s)
- Caroline E Phelps
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA.
| | - Edita Navratilova
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA
| | - Frank Porreca
- Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
13
|
The transition from acute to chronic pain: dynamic epigenetic reprogramming of the mouse prefrontal cortex up to 1 year after nerve injury. Pain 2021; 161:2394-2409. [PMID: 32427748 PMCID: PMC7497614 DOI: 10.1097/j.pain.0000000000001917] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Supplemental Digital Content is Available in the Text. DNA methylation undergoes rapid and large changes in mouse prefrontal cortex at multiple time points postinjury, implicating hundreds of genes in a time-dependent manner. Chronic pain is associated with persistent structural and functional changes throughout the neuroaxis, including in the prefrontal cortex (PFC). The PFC is important in the integration of sensory, cognitive, and emotional information and in conditioned pain modulation. We previously reported widespread epigenetic reprogramming in the PFC many months after nerve injury in rodents. Epigenetic modifications, including DNA methylation, can drive changes in gene expression without modifying DNA sequences. To date, little is known about epigenetic dysregulation at the onset of acute pain or how it progresses as pain transitions from acute to chronic. We hypothesize that acute pain after injury results in rapid and persistent epigenetic remodelling in the PFC that evolves as pain becomes chronic. We further propose that understanding epigenetic remodelling will provide insights into the mechanisms driving pain-related changes in the brain. Epigenome-wide analysis was performed in the mouse PFC 1 day, 2 weeks, 6 months, and 1 year after peripheral injury using the spared nerve injury in mice. Spared nerve injury resulted in rapid and persistent changes in DNA methylation, with robust differential methylation observed between spared nerve injury and sham-operated control mice at all time points. Hundreds of differentially methylated genes were identified, including many with known function in pain. Pathway analysis revealed enrichment in genes related to stimulus response at early time points, immune function at later time points, and actin and cytoskeletal regulation throughout the time course. These results emphasize the importance of considering pain chronicity in both pain research and in treatment optimization.
Collapse
|
14
|
Gajda JM, Asiedu M, Morrison G, Dunning JA, Ghoreishi-Haack N, Barth AL. NYX-2925, A NOVEL, NON-OPIOID, SMALL-MOLECULE MODULATOR OF THE N-METHYL-d-ASPARTATE RECEPTOR (NMDAR), DEMONSTRATES POTENTIAL TO TREAT CHRONIC, SUPRASPINAL CENTRALIZED PAIN CONDITIONS. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2020.100067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
15
|
Cortical Modulation of Nociception. Neuroscience 2021; 458:256-270. [PMID: 33465410 DOI: 10.1016/j.neuroscience.2021.01.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [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
|
16
|
Kummer KK, Mitrić M, Kalpachidou T, Kress M. The Medial Prefrontal Cortex as a Central Hub for Mental Comorbidities Associated with Chronic Pain. Int J Mol Sci 2020; 21:E3440. [PMID: 32414089 PMCID: PMC7279227 DOI: 10.3390/ijms21103440] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic pain patients frequently develop and suffer from mental comorbidities such as depressive mood, impaired cognition, and other significant constraints of daily life, which can only insufficiently be overcome by medication. The emotional and cognitive components of pain are processed by the medial prefrontal cortex, which comprises the anterior cingulate cortex, the prelimbic, and the infralimbic cortex. All three subregions are significantly affected by chronic pain: magnetic resonance imaging has revealed gray matter loss in all these areas in chronic pain conditions. While the anterior cingulate cortex appears hyperactive, prelimbic, and infralimbic regions show reduced activity. The medial prefrontal cortex receives ascending, nociceptive input, but also exerts important top-down control of pain sensation: its projections are the main cortical input of the periaqueductal gray, which is part of the descending inhibitory pain control system at the spinal level. A multitude of neurotransmitter systems contributes to the fine-tuning of the local circuitry, of which cholinergic and GABAergic signaling are particularly emerging as relevant components of affective pain processing within the prefrontal cortex. Accordingly, factors such as distraction, positive mood, and anticipation of pain relief such as placebo can ameliorate pain by affecting mPFC function, making this cortical area a promising target region for medical as well as psychosocial interventions for pain therapy.
Collapse
Affiliation(s)
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (K.K.K.); (M.M.); (T.K.)
| |
Collapse
|
17
|
The geriatric pain experience in mice: intact cutaneous thresholds but altered responses to tonic and chronic pain. Neurobiol Aging 2020; 89:1-11. [DOI: 10.1016/j.neurobiolaging.2019.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 11/29/2019] [Accepted: 12/19/2019] [Indexed: 11/23/2022]
|
18
|
Kawarai Y, Orita S, Nakamura J, Miyamoto S, Suzuki M, Inage K, Hagiwara S, Suzuki T, Nakajima T, Akazawa T, Ohtori S. Analgesic Effect of Duloxetine on an Animal Model of Monosodium Iodoacetate-Induced Hip Osteoarthritis. J Orthop Res 2020; 38:422-430. [PMID: 31538672 DOI: 10.1002/jor.24480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 09/13/2019] [Indexed: 02/04/2023]
Abstract
We investigated the efficacy of duloxetine on hyperalgesia, histopathological and radiographic findings, pain-related sensory innervation of dorsal-root ganglia (DRG), and spinal changes in a rat model of induced hip osteoarthritis (OA). The right hip joints of male Sprague-Dawley rats (n = 6 rats/group) in the Sham group were injected with 25 μl of sterile saline and 25 μl of sterile saline with 2 mg of monosodium iodoacetate (MIA) were injected to the MIA + Vehicle and MIA + Duloxetine groups. We injected duloxetine 20 mg/kg intraperitoneally in the MIA + Duloxetine group 28 days after injection, whereas rats in the MIA + Vehicle group were injected with 0.5 ml of 20% dimethyl sulfoxide. We assessed hyperalgesia, histopathological changes, immunoreactive (-ir) neurons for calcitonin gene-related peptide and activating transcription factor 3 in DRG, and immunoreactive neurons for ionized-calcium-binding adaptor molecule 1 (Iba1) in the dorsal horn of the spinal cord. MIA administration into the hip joint let to mechanical hyperalgesia of the ipsilateral hind paw (p < 0.05). A single injection of duloxetine significantly attenuated it in induced hip OA (p < 0.05) and suppressed the number of Iba1-ir microglia of the ipsilateral dorsal horn (p < 0.05). These results suggest that a single injection of duloxetine suppressed mechanical hyperalgesia and may influence the expression of Iba1 in the microglia of the ipsilateral dorsal horn in the MIA-induced hip OA. This finding implies the inhibitory effects of duloxetine against neuropathic pain, which may lead to a change of microglial activities. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:422-430, 2020.
Collapse
Affiliation(s)
- Yuya Kawarai
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| | - Sumihisa Orita
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| | - Junichi Nakamura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| | - Shuichi Miyamoto
- Department of Orthopaedic Surgery, Matsudo City General Hospital, 993-1 Sendabori, Matsudo, Chiba, 270-2296, Japan
| | - Miyako Suzuki
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| | - Kazuhide Inage
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| | - Shigeo Hagiwara
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| | - Takane Suzuki
- Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8677, Japan
| | - Takayuki Nakajima
- Department of Orthopaedic Surgery, Eastern Chiba Medical Center, 3-6-2 Okayamadai, Togane, Chiba, 283-8686, Japan
| | - Tsutomu Akazawa
- Department of Orthopaedic Surgery, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Chiba, 260-8677, Japan
| |
Collapse
|
19
|
Morrison G, Asiedu MN, Priebe JM, Dunning J, Ghoreishi-Haack N, Kroes RA, Bowers MS, Barth AL, Cearley CN, Moskal JR. The NMDAR modulator NYX-2925 alleviates neuropathic pain via a Src-dependent mechanism in the mPFC. NEUROBIOLOGY OF PAIN 2019; 7:100039. [PMID: 31909296 PMCID: PMC6938954 DOI: 10.1016/j.ynpai.2019.100039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 01/05/2023]
Abstract
Previous studies have shown that oral administration of the NMDAR modulator NYX-2925 alleviates pain in several animal models of neuropathic pain and this appears to be through mPFC, but not spinal, mediated mechanisms. While much is known about the impact of neuropathic pain on NMDAR-mediated signaling in the spinal cord, limited studies have focused on the brain. In the current study, we assess signaling changes associated with NMDAR-mediated plasticity in the mPFC and the impact of NYX-2925 administration on the normalization of these signaling changes. We found a decrease in activated Src levels in the mPFC of animals with chronic constriction injury (CCI) of the sciatic nerve. While Src mediated activation of NMDARs was also decreased in CCI animals, the main NMDAR phosphorylation site of CAMKII was not affected. This is in opposition to what has been found in the spinal cord, where both Src and CAMKII activation are increased. Oral administration of NYX-2925 restored levels of activated Src and Src phosphorylation sites on GluN2A and GluN2B in the mPFC, with no effect on activated CAMKII levels. The analgesic effect of NYX-2925 appears dependent on this restoration of Src activation in the mPFC, as co-administering Src activation inhibitors prevented the NYX-2925 analgesic effect. Overall, these data suggest that NMDAR-mediated signaling plays a key role in neuropathic pain, albeit in different directions in the spinal cord vs. the mPFC. Furthermore, the analgesic effect of NYX-2925 appears to involve a restoration of NMDAR-mediated signaling in the mPFC.
Collapse
Affiliation(s)
| | | | | | | | | | - Roger A Kroes
- Aptinyx Inc., Evanston, IL, United States.,Falk Ctr. for Mol. Therapeutics, McCormick School of Engineering, Northwestern University, Evanston, IL, United States
| | - M Scott Bowers
- Aptinyx Inc., Evanston, IL, United States.,Falk Ctr. for Mol. Therapeutics, McCormick School of Engineering, Northwestern University, Evanston, IL, United States
| | | | | | - Joseph R Moskal
- Aptinyx Inc., Evanston, IL, United States.,Falk Ctr. for Mol. Therapeutics, McCormick School of Engineering, Northwestern University, Evanston, IL, United States
| |
Collapse
|
20
|
Rat ultrasonic vocalizations as a measure of the emotional component of chronic pain. Neuroreport 2019; 30:863-866. [DOI: 10.1097/wnr.0000000000001282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
21
|
Medeiros P, de Freitas RL, Boccella S, Iannotta M, Belardo C, Mazzitelli M, Romano R, De Gregorio D, Coimbra NC, Palazzo E, Maione S. Characterization of the sensory, affective, cognitive, biochemical, and neuronal alterations in a modified chronic constriction injury model of neuropathic pain in mice. J Neurosci Res 2019; 98:338-352. [PMID: 31396990 DOI: 10.1002/jnr.24501] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/24/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
The chronic constriction injury (CCI) of the sciatic nerve is a nerve injury-based model of neuropathic pain (NP). Comorbidities of NP such as depression, anxiety, and cognitive deficits are associated with a functional reorganization of the medial prefrontal cortex (mPFC). Here, we have employed an adapted model of CCI by placing one single loose ligature around the sciatic nerve in mice for investigating the alterations in sensory, motor, affective, and cognitive behavior and in electrophysiological and biochemical properties in the prelimbic division (PrL) of the mPFC. Our adapted model of CCI induced mechanical allodynia, motor, and cognitive impairments and anxiety- and depression-like behavior. In the PrL division of mPFC was observed an increase in GABA and a decrease in d-aspartate levels. Moreover an increase in the activity of neurons responding to mechanical stimulation with an excitation, mPFC (+), and a decrease in those responding with an inhibition, mPFC (-), was found. Altogether these findings demonstrate that a single ligature around the sciatic nerve was able to induce sensory, affective, cognitive, biochemical, and functional alterations already observed in other neuropathic pain models and it may be an appropriate and easily reproducible model for studying neuropathic pain mechanisms and treatments.
Collapse
Affiliation(s)
- Priscila Medeiros
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.,Laboratory of Neurosciences of Pain & Emotions and Multi-User Centre of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Ribeirão Preto, Brazil.,Behavioural Neurosciences Institute (INeC), Ribeirão Preto, Brazil
| | - Renato Leonardo de Freitas
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy.,Laboratory of Neurosciences of Pain & Emotions and Multi-User Centre of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Ribeirão Preto, Brazil.,Behavioural Neurosciences Institute (INeC), Ribeirão Preto, Brazil.,Biomedical Sciences Institute, Federal University of Alfenas (UNIFAL), Alfenas (MG), Brazil
| | - Serena Boccella
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Carmela Belardo
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Mariacristina Mazzitelli
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Rosaria Romano
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Norberto Cysne Coimbra
- Laboratory of Neurosciences of Pain & Emotions and Multi-User Centre of Neuroelectrophysiology, Department of Surgery and Anatomy, Ribeirão Preto Medical School of the University of São Paulo (FMRP-USP), Ribeirão Preto (SP), Brazil.,Laboratory of Neuroanatomy and Neuropsychobiology, Department of Pharmacology, FMRP-USP, Ribeirão Preto, Brazil.,Behavioural Neurosciences Institute (INeC), Ribeirão Preto, Brazil
| | - Enza Palazzo
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Division of Pharmacology, Department of Experimental Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
22
|
N-methyl-d-aspartate Receptors in the Prelimbic Cortex are Critical for the Maintenance of Neuropathic Pain. Neurochem Res 2019; 44:2068-2080. [DOI: 10.1007/s11064-019-02843-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 06/27/2019] [Accepted: 07/04/2019] [Indexed: 12/13/2022]
|
23
|
Mussio CA, Harte SE, Borszcz GS. Regional Differences Within the Anterior Cingulate Cortex in the Generation Versus Suppression of Pain Affect in Rats. THE JOURNAL OF PAIN 2019; 21:121-134. [PMID: 31201992 DOI: 10.1016/j.jpain.2019.06.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/22/2019] [Accepted: 06/02/2019] [Indexed: 01/08/2023]
Abstract
The anterior cingulate cortex (ACC) modulates emotional responses to pain. Whereas, the caudal ACC (cACC) promotes expression of pain affect, the rostral ACC (rACC) contributes to its suppression. Both subdivisions receive glutamatergic innervation, and the present study evaluated the contribution of N-methyl-d-aspartic acid (NMDA) receptors within these subdivisions to rats' expression of pain affect. Vocalizations that follow a brief noxious tail shock (vocalization afterdischarges, VAD) are a validated rodent model of pain affect. The threshold current for eliciting VAD was increased in a dose-dependent manner by injecting NMDA into the rACC, but performance (latency, amplitude, and duration) at threshold was not altered. Alternately, the threshold current for eliciting VAD was not altered following injection of NMDA into the cACC, but its amplitude and duration at threshold were increased in a dose-dependent manner. These effects were limited to Cg1 of the rACC and cACC, and blocked by pretreatment of the ACC with the NMDA receptor antagonist d-2-amino-5-phosphonovalerate. These findings demonstrate that NMDA receptor agonism within the cACC and rACC either increases or decreases emotional responses to noxious stimulation, respectively. PERSPECTIVE: NMDA receptor activation of the rostral and caudal ACC respectively inhibited or enhanced rats' emotional response to pain. These findings mirror those obtained from human neuroimaging studies; thereby, supporting the use of this model system in evaluating the contribution of ACC to pain affect.
Collapse
Affiliation(s)
- Casey A Mussio
- Behavioral and Cognitive Neuroscience Program, Department of Psychology, Wayne State University, Detroit, Michigan
| | - Steven E Harte
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan
| | - George S Borszcz
- Behavioral and Cognitive Neuroscience Program, Department of Psychology, Wayne State University, Detroit, Michigan.
| |
Collapse
|
24
|
Alterations in brain neurocircuitry following treatment with the chemotherapeutic agent paclitaxel in rats. NEUROBIOLOGY OF PAIN 2019; 6:100034. [PMID: 31223138 PMCID: PMC6565758 DOI: 10.1016/j.ynpai.2019.100034] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/09/2019] [Accepted: 05/26/2019] [Indexed: 12/21/2022]
Abstract
Imaging the reorganization of pain neural circuitry within 8 days of chemotherapy. Using rat model of neuropathy with multimodal MRI. Showing loss of anticorrelation between prefrontal cortex and PAG. Identifying the interaction between periaqueductal gray and brainstem raphe.
Human and animal studies suggest that both traumatic nerve injury and toxic challenge with chemotherapeutic agents involves the reorganization of neural circuits in the brain. However, there have been no prospective studies, human or animal, using magnetic resonance imaging (MRI) to identify changes in brain neural circuitry that accompany the development of chemotherapy-induced neuropathic pain (i.e. within days following cessation of chemotherapy treatment and without the confound cancer). To this end, different MRI protocols were used to ascertain whether a reorganization of brain neural circuits is observed in otherwise normal rats exposed to the taxane chemotherapeutic agent paclitaxel. We conducted an imaging study to evaluate the impact of a well-established paclitaxel dosing regimen, validated to induce allodynia in control rats within eight days of treatment, on brain neural circuitry. Rats received either paclitaxel (2 mg/kg/day i.p; cumulative dose of 8 mg/kg) or its vehicle four times on alternate days (i.e. day 0, 2, 4, 6). Following the cessation of treatments (i.e. on day 8), all rats were tested for responsiveness to cold followed by diffusion weighted magnetic resonance imaging and assessment of resting state functional connectivity. Imaging data were analyzed using a 3D MRI rat with 173 segmented and annotated brain areas. Paclitaxel-treated rats were more sensitive to a cold stimulus compared to controls. Diffusion weighted imaging identified brain areas involved in the emotional and motivational response to chronic pain that were impacted by paclitaxel treatment. Affected brain regions included the prefrontal cortex, amygdala, hippocampus, hypothalamus and the striatum/nucleus accumbens. This putative reorganization of gray matter microarchitecture formed a continuum of brain areas stretching from the basal medial/lateral forebrain to the midbrain. Resting state functional connectivity showed reorganization between the periaqueductal gray, a key node in nociceptive neural circuitry, and connections to the brainstem. Our results, employing different imaging modalities to assess the central nervous system effects of chemotherapy, fit the theory that chronic pain is regulated by emotion and motivation and influences activity in the periaqueductal gray and brainstem to modulate pain perception.
Collapse
|
25
|
Davis MP, Behm B, Mehta Z, Fernandez C. The Potential Benefits of Palmitoylethanolamide in Palliation: A Qualitative Systematic Review. Am J Hosp Palliat Care 2019; 36:1134-1154. [PMID: 31113223 DOI: 10.1177/1049909119850807] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Palmitoylethanolamide (PEA) is a nutraceutical endocannabinoid that was retrospectively discovered in egg yolks. Feeding poor children with known streptococcal infections prevented rheumatic fever. Subsequently, it was found to alter the course of influenza. Unfortunately, there is little known about its pharmacokinetics. Palmitoylethanolamide targets nonclassical cannabinoid receptors rather than CB1 and CB2 receptors. Palmitoylethanolamide will only indirectly activate classical cannabinoid receptors by an entourage effect. There are a significant number of prospective and randomized trials demonstrating the pain-relieving effects of PEA. There is lesser evidence of benefit in patients with nonpain symptoms related to depression, Parkinson disease, strokes, and autism. There are no reported drug-drug interactions and very few reported adverse effects from PEA. Further research is needed to define the palliative benefits to PEA.
Collapse
|
26
|
Forebrain medial septum sustains experimental neuropathic pain. Sci Rep 2018; 8:11892. [PMID: 30089875 PMCID: PMC6082830 DOI: 10.1038/s41598-018-30177-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
The present study explored the role of the medial septal region (MS) in experimental neuropathic pain. For the first time, we found that the MS sustains nociceptive behaviors in rodent models of neuropathic pain, especially in the chronic constriction injury (CCI) model and the paclitaxel model of chemotherapy-induced neuropathic pain. For example, inactivation of the MS with intraseptal muscimol (2 μg/μl, 0.5 μl), a GABA mimetic, reversed peripheral hypersensitivity (PH) in the CCI model and induced place preference in a conditioned place preference task, a surrogate measure of spontaneous nociception. The effect of intraseptal muscimol on PH was comparable to that seen with microinjection of the local anesthetic, lidocaine, into rostral ventromedial medulla which is implicated in facilitating experimental chronic nociception. Cellular analysis in the CCI model showed that the MS region sustains nociceptive gain with CCI by facilitating basal nociceptive processing and the amplification of stimulus-evoked neural processing. Indeed, consistent with the idea that excitatory transmission through MS facilitates chronic experimental pain, intraseptal microinjection of antagonists acting at AMPA and NMDA glutamate receptors attenuated CCI-induced PH. We propose that the MS is a central monitor of bodily nociception which sustains molecular plasticity triggered by persistent noxious insult.
Collapse
|
27
|
Ghoreishi-Haack N, Priebe JM, Aguado JD, Colechio EM, Burgdorf JS, Bowers MS, Cearley CN, Khan MA, Moskal JR. NYX-2925 Is a Novel N-Methyl-d-Aspartate Receptor Modulator that Induces Rapid and Long-Lasting Analgesia in Rat Models of Neuropathic Pain. J Pharmacol Exp Ther 2018; 366:485-497. [DOI: 10.1124/jpet.118.249409] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023] Open
|
28
|
Ong WY, Stohler CS, Herr DR. Role of the Prefrontal Cortex in Pain Processing. Mol Neurobiol 2018; 56:1137-1166. [PMID: 29876878 PMCID: PMC6400876 DOI: 10.1007/s12035-018-1130-9] [Citation(s) in RCA: 380] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022]
Abstract
The prefrontal cortex (PFC) is not only important in executive functions, but also pain processing. The latter is dependent on its connections to other areas of the cerebral neocortex, hippocampus, periaqueductal gray (PAG), thalamus, amygdala, and basal nuclei. Changes in neurotransmitters, gene expression, glial cells, and neuroinflammation occur in the PFC during acute and chronic pain, that result in alterations to its structure, activity, and connectivity. The medial PFC (mPFC) could serve dual, opposing roles in pain: (1) it mediates antinociceptive effects, due to its connections with other cortical areas, and as the main source of cortical afferents to the PAG for modulation of pain. This is a ‘loop’ where, on one side, a sensory stimulus is transformed into a perceptual signal through high brain processing activity, and perceptual activity is then utilized to control the flow of afferent sensory stimuli at their entrance (dorsal horn) to the CNS. (2) It could induce pain chronification via its corticostriatal projection, possibly depending on the level of dopamine receptor activation (or lack of) in the ventral tegmental area-nucleus accumbens reward pathway. The PFC is involved in biopsychosocial pain management. This includes repetitive transcranial magnetic stimulation, transcranial direct current stimulation, antidepressants, acupuncture, cognitive behavioral therapy, mindfulness, music, exercise, partner support, empathy, meditation, and prayer. Studies demonstrate the role of the PFC during placebo analgesia, and in establishing links between pain and depression, anxiety, and loss of cognition. In particular, losses in PFC grey matter are often reversible after successful treatment of chronic pain.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore.
- Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 119260, Singapore.
| | | | - Deron R Herr
- Department of Pharmacology, National University of Singapore, Singapore, 119260, Singapore.
| |
Collapse
|
29
|
Altered Excitability and Local Connectivity of mPFC-PAG Neurons in a Mouse Model of Neuropathic Pain. J Neurosci 2018; 38:4829-4839. [PMID: 29695413 DOI: 10.1523/jneurosci.2731-17.2018] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/09/2018] [Accepted: 03/20/2018] [Indexed: 12/29/2022] Open
Abstract
The medial prefrontal cortex (mPFC) plays a major role in both sensory and affective aspects of pain. There is extensive evidence that chronic pain produces functional changes within the mPFC. However, our understanding of local circuit changes to defined subpopulations of mPFC neurons in chronic pain models remains unclear. A major subpopulation of mPFC neurons project to the periaqueductal gray (PAG), which is a key midbrain structure involved in endogenous pain suppression and facilitation. Here, we used laser scanning photostimulation of caged glutamate to map cortical circuits of retrogradely labeled cortico-PAG (CP) neurons in layer 5 (L5) of mPFC in brain slices prepared from male mice having undergone chronic constriction injury (CCI) of the sciatic nerve. Whole-cell recordings revealed a significant reduction in excitability for L5 CP neurons contralateral to CCI in the prelimbic (PL), but not infralimbic (IL), region of mPFC. Circuit mapping showed that excitatory inputs to L5 CP neurons in both PL and IL arose primarily from layer 2/3 (L2/3) and were significantly reduced in CCI mice. Glutamate stimulation of L2/3 and L5 elicited inhibitory inputs to CP neurons in both PL and IL, but only L2/3 input was significantly reduced in CP neurons of CCI mice. We also observed significant reduction in excitability and L2/3 inhibitory input to CP neurons ipsilateral to CCI. These results demonstrating region and laminar specific changes to mPFC-PAG neurons suggest that a unilateral CCI bilaterally alters cortical circuits upstream of the endogenous analgesic network, which may contribute to persistence of chronic pain.SIGNIFICANCE STATEMENT Chronic pain is a significant unresolved medical problem that is refractory to traditional analgesics and can negatively affect emotional health. The role of central circuits in mediating the persistent nature of chronic pain remains unclear. Local circuits within the medial prefrontal cortex (mPFC) process ascending pain inputs and can modulate endogenous analgesia via direct projections to the periaqueductal gray (PAG). However, the mechanisms by which chronic pain alters intracortical circuitry of mPFC-PAG neurons are unknown. Here, we report specific changes to local circuits of mPFC-PAG neurons in mice displaying chronic pain behavior after nerve injury. These findings provide evidence for a neural mechanism by which chronic pain disrupts the descending analgesic system via functional changes to cortical circuits.
Collapse
|
30
|
Lyons DN, Zhang L, Pandya JD, Danaher RJ, Ma F, Miller CS, Sullivan PG, Sirbu C, Westlund KN. Combination Drug Therapy of Pioglitazone and D-cycloserine Attenuates Chronic Orofacial Neuropathic Pain and Anxiety by Improving Mitochondrial Function Following Trigeminal Nerve Injury. Clin J Pain 2018; 34:168-177. [PMID: 28542026 PMCID: PMC5701889 DOI: 10.1097/ajp.0000000000000515] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The study aim was to determine how peripheral trigeminal nerve injury affects mitochondrial respiration and to test efficacy of combined treatment with 2 Federal Drug Administration approved drugs with potential for improving mitochondrial bioenergetics, pain and anxiety-related behaviors in a chronic orofacial neuropathic pain mouse model. METHODS Efficacy of (R)-(+)-4-amino-3-isoxazolidinone (D-cycloserine, DCS), an N-Methyl-D-aspartate antagonist/agonist, and Pioglitazone (PIO), a selective agonist of nuclear receptor peroxisome proliferator-activated receptor gamma was investigate in the trigeminal inflammatory compression (TIC) neuropathic nerve injury mouse model. Combined low doses of these drugs (80 mg/kg DCS and 100 mg/kg PIO) were given as a single bolus or daily for 7 days post-TIC to test ability to attenuate neuropathic nociceptive and associated cognitive dependent anxiety behaviors. In addition, beneficial effects of the DCS/PIO drug combination were explored ex vivo in isolated cortex/brainstem mitochondria at 28 weeks post-TIC. RESULTS The DCS/PIO combination not only attenuated orofacial neuropathic pain and anxiety-related behaviors associated with trigeminal nerve injury, but it also improved mitochondrial bioenergetics. DISCUSSION The DCS/PIO combination uncoupled mitochondrial respiration in the TIC model to improve cortical mitochondrial dysfunction, as well as reduced nociceptive and anxiety behaviors present in mice with centralized chronic neuropathic nerve injury. Combining these drugs could be a beneficial treatment for patients with depression, anxiety, or other psychological conditions due to their chronic pain status.
Collapse
Affiliation(s)
| | - Liping Zhang
- Department of Physiology, University of Kentucky
| | - Jignesh D. Pandya
- Spinal Cord and Brain Injury Research Center, University of Kentucky
| | | | - Fei Ma
- Department of Physiology, University of Kentucky
| | | | | | - Cristian Sirbu
- Department of Behavioral Medicine & Psychiatry, West Virginia University
| | | |
Collapse
|
31
|
Novel analgesic effects of melanin-concentrating hormone on persistent neuropathic and inflammatory pain in mice. Sci Rep 2018; 8:707. [PMID: 29335480 PMCID: PMC5768747 DOI: 10.1038/s41598-018-19145-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 12/22/2017] [Indexed: 12/17/2022] Open
Abstract
The melanin-concentrating hormone (MCH) is a peptidergic neuromodulator synthesized by neurons in the lateral hypothalamus and zona incerta. MCHergic neurons project throughout the central nervous system, indicating the involvements of many physiological functions, but the role in pain has yet to be determined. In this study, we found that pMCH-/- mice showed lower baseline pain thresholds to mechanical and thermal stimuli than did pMCH+/+ mice, and the time to reach the maximum hyperalgesic response was also significantly earlier in both inflammatory and neuropathic pain. To examine its pharmacological properties, MCH was administered intranasally into mice, and results indicated that MCH treatment significantly increased mechanical and thermal pain thresholds in both pain models. Antagonist challenges with naltrexone (opioid receptor antagonist) and AM251 (cannabinoid 1 receptor antagonist) reversed the analgesic effects of MCH in both pain models, suggesting the involvement of opioid and cannabinoid systems. MCH treatment also increased the expression and activation of CB1R in the medial prefrontal cortex and dorsolateral- and ventrolateral periaqueductal grey. The MCH1R antagonist abolished the effects induced by MCH. This is the first study to suggest novel analgesic actions of MCH, which holds great promise for the application of MCH in the therapy of pain-related diseases.
Collapse
|
32
|
Khan MA, Houck DR, Gross AL, Zhang XL, Cearley C, Madsen TM, Kroes RA, Stanton PK, Burgdorf J, Moskal JR. NYX-2925 Is a Novel NMDA Receptor-Specific Spirocyclic-β-Lactam That Modulates Synaptic Plasticity Processes Associated with Learning and Memory. Int J Neuropsychopharmacol 2017; 21:242-254. [PMID: 29099938 PMCID: PMC5838819 DOI: 10.1093/ijnp/pyx096] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/17/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND N-methyl-D-aspartate receptors are one member of a family of ionotropic glutamate receptors that play a pivotal role in synaptic plasticity processes associated with learning and have become attractive therapeutic targets for diseases such as depression, anxiety, schizophrenia, and neuropathic pain. NYX-2925 ((2S, 3R)-3-hydroxy-2-((R)-5-isobutyryl-1-oxo-2,5-diazaspiro[3.4]octan-2-yl)butanamide) is one member of a spiro-β-lactam-based chemical platform that mimics some of the dipyrrolidine structural features of rapastinel (formerly GLYX-13: threonine-proline-proline-threonine) and is distinct from known N-methyl-D-aspartate receptor agonists or antagonists such as D-cycloserine, ketamine, MK-801, kynurenic acid, or ifenprodil. METHODS The in vitro and in vivo pharmacological properties of NYX-2925 were examined. RESULTS NYX-2925 has a low potential for "off-target" activity, as it did not exhibit any significant affinity for a large panel of neuroactive receptors, including hERG receptors. NYX-2925 increased MK-801 binding to human N-methyl-D-aspartate receptor NR2A-D subtypes expressed in HEK cells and enhanced N-methyl-D-aspartate receptor current and long-term potentiation (LTP) in rat hippocampal slices (100-500 nM). Single dose ex vivo studies showed increased metaplasticity in a hippocampal LTP paradigm and structural plasticity 24 hours after administration (1 mg/kg p.o.). Significant learning enhancement in both novel object recognition and positive emotional learning paradigms were observed (0.01-1 mg/kg p.o.), and these effects were blocked by the N-methyl-D-aspartate receptor antagonist CPP. NYX-2925 does not show any addictive or sedative/ataxic side effects and has a therapeutic index of >1000. NYX-2925 (1 mg/kg p.o.) has a cerebrospinal fluid half-life of 1.2 hours with a Cmax of 44 nM at 1 hour. CONCLUSIONS NYX-2925, like rapastinel, activates an NMDA receptor-mediated synaptic plasticity process and may have therapeutic potential for a variety of NMDA receptor-mediated central nervous system disorders.
Collapse
Affiliation(s)
| | | | | | - Xiao-lei Zhang
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York
| | | | | | - Roger A Kroes
- Aptinyx Inc., Evanston, Ilinois,Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Ilinois
| | - Patric K Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, New York
| | - Jeffrey Burgdorf
- Aptinyx Inc., Evanston, Ilinois,Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Ilinois
| | - Joseph R Moskal
- Aptinyx Inc., Evanston, Ilinois,Falk Center for Molecular Therapeutics, Department of Biomedical Engineering, Northwestern University, Evanston, Ilinois,Correspondence: Joseph Moskal, PhD, Falk Center for Molecular Therapeutics, Northwestern University Department of Biomedical Engineering, 1801 Maple Ave, Suite 4300, Evanston, IL, 60201 ()
| |
Collapse
|
33
|
D'Aniello A, Luongo L, Romano R, Iannotta M, Marabese I, Boccella S, Belardo C, de Novellis V, Arra C, Barbieri A, D'Aniello B, Scandurra A, Magliozzi L, Fisher G, Guida F, Maione S. d-Aspartic acid ameliorates painful and neuropsychiatric changes and reduces β-amyloid Aβ 1-42 peptide in a long lasting model of neuropathic pain. Neurosci Lett 2017; 651:151-158. [PMID: 28487079 DOI: 10.1016/j.neulet.2017.04.041] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/11/2017] [Accepted: 04/21/2017] [Indexed: 11/28/2022]
Abstract
Depressive symptoms and other neuropsychiatric dysfunctions are common in neurodegenerative disorders, including chronic pain and dementia. A correlation between the β-amyloid protein accumulation and the development of depression has been suggested, however the underlying mechanisms are unknown. d-Aspartate (d-Asp) is a free d-amino acid found in the mammalian brain and involved in neurological and psychiatric processes, such as cognition and affective disorders. In this study we have investigated the effects of a repeated treatment with d-Asp in a long-lasting (12 months) model of neuropathic pain, the spared nerve injury (SNI), in mice. Specifically, we evaluated i) the pain sensitivity and related emotional/cognitive dysfunctions induced by SNI, ii) possible changes in the β-amyloid protein accumulation in specific brain regions involved in pain mechanisms ii) possible changes in steroids level in neuropathic animals with or without d-Asp in the same brain areas. SNI mice showed an increase of the insoluble form of Aβ1-42 at hippocampal level and displayed cognitive impairments, stereotypical and depressive-like behaviours. d-Asp treatment reduced abnormal behaviours and normalized the β-amyloid protein expression. Moreover, d-Asp dramatically increased steroids level measured in the prefrontal cortex and in the hippocampus. Our findings provide new insights into pain mechanisms and suggest a possible role of β-amyloid protein in neuropsychiatric dysfunctions associated with chronic pain.
Collapse
Affiliation(s)
- Antimo D'Aniello
- Department of Neurobiology and Comparative Physiology, Zoological Station "A. Dohrn", Napoli, Italy; Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy.
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Rosaria Romano
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Claudio Arra
- Animal Facility Unit Traslational Research Department, Istituto Nazionale Tumori -IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Antonio Barbieri
- Animal Facility Unit Traslational Research Department, Istituto Nazionale Tumori -IRCCS-Fondazione G. Pascale, Napoli, Italia
| | - Biagio D'Aniello
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Anna Scandurra
- Department of Biology, University of Naples "Federico II", Naples, Italy
| | - Laura Magliozzi
- Department of Environmental and Biological Sciences and Technologies, University of Salento, CoNISMa, Lecce, Italy
| | - George Fisher
- Department of Physical Sciences, Barry University, Miami Schores , USA
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, Università della Campania, Naples, Italy.
| |
Collapse
|
34
|
Therapeutic benefits of the methyl donor S-adenosylmethionine on nerve injury–induced mechanical hypersensitivity and cognitive impairment in mice. Pain 2016; 158:802-810. [DOI: 10.1097/j.pain.0000000000000811] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Schnitzer TJ, Torbey S, Herrmann K, Kaushal G, Yeasted R, Vania Apkarian A. A randomized placebo-controlled pilot study of the efficacy and safety of D-cycloserine in people with chronic back pain. Mol Pain 2016; 12:1744806916678627. [PMID: 27852965 PMCID: PMC5117251 DOI: 10.1177/1744806916678627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/04/2016] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Few effective pharmacological treatment options exist for chronic back pain, the leading cause of disability in the US, and all are associated with significant adverse effects. OBJECTIVE To determine the efficacy and safety of D-cycloserine, a partial agonist to the N-methyl-D-aspartate receptor, in the treatment of chronic low back pain. METHODS A total of 41 participants with chronic back pain who met all inclusion and exclusion criteria were enrolled in a double-blind, placebo-controlled randomized pilot trial of D-cycloserine. Treatment was administered orally for six weeks at escalating daily doses of 100 mg, 200 mg, and 400 mg, each for two weeks. The primary outcome measure was back pain intensity using the Numeric Rating Scale (0-10). Secondary measures were back pain-related questionnaires: McGill Pain Questionnaire short form, painDETECT, PANAS, and BDI. The pre-specified analysis was a two-way repeated measures analysis of variance. RESULTS A treatment difference was observed between groups treated with D-cycloserine and placebo at six weeks of 1.05 ± 3.1 units on the Numeric Rating Scale, with an effect size of 0.4 and p = 0.14. This trend of better chronic back pain relief with D-cycloserine was also observed in the secondary measures. No safety issues were seen. CONCLUSION The difference in mean pain between the D-cycloserine and placebo groups did not reach statistical significance. However, a clinically meaningful effect size in the magnitude of pain relief was observed with a consistent pattern across multiple outcome measures with good safety, supporting further research into the effectiveness of D-cycloserine for chronic back pain.
Collapse
Affiliation(s)
- Thomas J Schnitzer
- Department of Physical Medicine and Rehabilitation and Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Souraya Torbey
- Department of Psychiatry, University of Virginia School of Medicine, VA, USA
| | - Kristi Herrmann
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gagan Kaushal
- Jefferson School of Pharmacy, Thomas Jefferson University, PA, USA
| | - Renita Yeasted
- Department of Physical Medicine and Rehabilitation and Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - A Vania Apkarian
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
36
|
Future Targets for Female Sexual Dysfunction. J Sex Med 2016; 13:1147-65. [DOI: 10.1016/j.jsxm.2016.05.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 05/22/2016] [Accepted: 05/27/2016] [Indexed: 12/18/2022]
|
37
|
d-Aspartate drinking solution alleviates pain and cognitive impairment in neuropathic mice. Amino Acids 2016; 48:1553-67. [DOI: 10.1007/s00726-016-2205-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/23/2016] [Indexed: 12/15/2022]
|
38
|
Specific Targeting of the Basolateral Amygdala to Projectionally Defined Pyramidal Neurons in Prelimbic and Infralimbic Cortex. eNeuro 2016; 3:eN-NWR-0002-16. [PMID: 27022632 PMCID: PMC4804386 DOI: 10.1523/eneuro.0002-16.2016] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 11/21/2022] Open
Abstract
Adjacent prelimbic (PL) and infralimbic (IL) regions in the medial prefrontal cortex have distinct roles in emotional learning. A complete mechanistic understanding underlying this dichotomy remains unclear. Here we explored targeting of specific PL and IL neurons by the basolateral amygdala (BLA), a limbic structure pivotal in pain and fear processing. In mice, we used retrograde labeling, brain-slice recordings, and adenoviral optogenetics to dissect connectivity of ascending BLA input onto PL and IL neurons projecting to the periaqueductal gray (PAG) or the amygdala. We found differential targeting of BLA projections to PL and IL cortex. Activating BLA projections evoked excitatory and inhibitory responses in cortico-PAG (CP) neurons in layer 5 (L5) of both PL and IL cortex. However, all inhibitory responses were polysynaptic and monosynaptic BLA input was stronger to CP neurons in IL cortex. Conversely, the BLA preferentially targeted corticoamygdalar (CA) neurons in layer 2 (L2) of PL over IL cortex. We also reveal that BLA input is projection specific by showing preferential targeting of L5 CP neurons over neighboring L3/5 CA neurons in IL cortex. We conclude by showing that BLA input is laminar-specific by producing stronger excitatory responses CA neurons in L3/5 compared with L2 in IL cortex. Collectively, this study reveals differential targeting of the BLA to PL and IL cortex, which depends both on laminar location and projection target of cortical neurons. Overall, our findings should have important implications for understanding the processing of pain and fear input by the PL and IL cortex.
Collapse
|
39
|
Vachon-Presseau E, Centeno MV, Ren W, Berger SE, Tétreault P, Ghantous M, Baria A, Farmer M, Baliki MN, Schnitzer TJ, Apkarian AV. The Emotional Brain as a Predictor and Amplifier of Chronic Pain. J Dent Res 2016; 95:605-12. [PMID: 26965423 DOI: 10.1177/0022034516638027] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human neuroimaging studies and complementary animal experiments now identify the gross elements of the brain involved in the chronification of pain. We briefly review these advances in relation to somatic and orofacial persistent pain conditions. First, we emphasize the importance of reverse translational research for understanding chronic pain-that is, the power of deriving hypotheses directly from human brain imaging of clinical conditions that can be invasively and mechanistically studied in animal models. We then review recent findings demonstrating the importance of the emotional brain (i.e., the corticolimbic system) in the modulation of acute pain and in the prediction and amplification of chronic pain, contrasting this evidence with recent findings regarding the role of central sensitization in pain chronification, especially for orofacial pain. We next elaborate on the corticolimbic circuitry and underlying mechanisms that determine the transition to chronic pain. Given this knowledge, we advance a new mechanistic definition of chronic pain and discuss the clinical implications of this new definition as well as novel therapeutic potentials suggested by these advances.
Collapse
Affiliation(s)
- E Vachon-Presseau
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M V Centeno
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - W Ren
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - S E Berger
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - P Tétreault
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M Ghantous
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - A Baria
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M Farmer
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - M N Baliki
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - T J Schnitzer
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - A V Apkarian
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
40
|
Heinricher MM. Pain Modulation and the Transition from Acute to Chronic Pain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 904:105-15. [PMID: 26900066 DOI: 10.1007/978-94-017-7537-3_8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is now increasing evidence that pathological pain states are at least in part driven by changes in the brain itself. Descending modulatory pathways are known to mediate top-down regulation of nociceptive processing, transmitting cortical and limbic influences to the dorsal horn. However, these modulatory pathways are also intimately intertwined with ascending transmission pathways through positive and negative feedback loops. Models of persistent pain that fail to include descending modulatory pathways are thus incomplete. Although teasing out individual links in a recurrent network is never straightforward, it is imperative that understanding of pain modulation be fully integrated into how we think about pain.
Collapse
Affiliation(s)
- Mary M Heinricher
- Dept. Neurological Surgery, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
41
|
David-Pereira A, Puga S, Gonçalves S, Amorim D, Silva C, Pertovaara A, Almeida A, Pinto-Ribeiro F. Metabotropic glutamate 5 receptor in the infralimbic cortex contributes to descending pain facilitation in healthy and arthritic animals. Neuroscience 2015; 312:108-19. [PMID: 26548413 DOI: 10.1016/j.neuroscience.2015.10.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 10/29/2015] [Accepted: 10/30/2015] [Indexed: 12/30/2022]
Abstract
The involvement of the prefrontal cortex in pain processing has been recently addressed. We studied the role of the infralimbic cortex (IL) and group I metabotropic glutamate receptors (mGluRs) in descending modulation of nociception in control and monoarthritic (ARTH) conditions. Nociception was assessed using heat-induced paw withdrawal while drugs were microinjected in the IL of rats. Local anesthesia of the IL or the adjacent prelimbic cortex (PL) facilitated nociception, indicating that IL and PL are tonically promoting spinal antinociception. Phasic activation with glutamate (GLU) revealed opposing roles of the PL and IL; GLU in the PL had a fast antinociceptive action, while in the IL it had a slow onset pronociceptive action. IL administration of a local anesthetic or GLU produced identical results in ARTH and control animals. An mGluR5 agonist in the IL induced a pronociceptive effect in both groups, while mGluR5 antagonists had no effect in controls but induced antinociception in ARTH rats. Activation of the IL mGluR1 (through co-administration of mGluR1/5 agonist and mGluR5 antagonist) did not alter nociception in controls but induced antinociception in ARTH animals. IL administration of an mGluR1 antagonist failed to alter nociception in either experimental group. Finally, mGluR5 but not mGluR1 antagonists blocked the pronociceptive action of GLU in both groups. The results indicate that IL contributes to descending modulation of nociception. mGluR5 in the IL enhance nociception in healthy control and monoarthritic animals, an effect that is tonic in ARTH. Moreover, activation of IL mGluR1s attenuates nociception following the development of monoarthritis.
Collapse
Affiliation(s)
- A David-Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S Puga
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - S Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - D Amorim
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - C Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A Pertovaara
- Biomedicum Helsinki, Institute of Biomedicine/Physiology, University of Helsinki, Helsinki, Finland
| | - A Almeida
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - F Pinto-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences (ECS), Campus of Gualtar, University of Minho, 4750-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
42
|
Bradman MJ, Ferrini F, Salio C, Merighi A. Practical mechanical threshold estimation in rodents using von Frey hairs/Semmes–Weinstein monofilaments: Towards a rational method. J Neurosci Methods 2015; 255:92-103. [DOI: 10.1016/j.jneumeth.2015.08.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 12/27/2022]
|
43
|
Abstract
Recent neuroimaging studies suggest that the brain adapts with pain, as well as imparts risk for developing chronic pain. Within this context, we revisit the concepts for nociception, acute and chronic pain, and negative moods relative to behavior selection. We redefine nociception as the mechanism protecting the organism from injury, while acute pain as failure of avoidant behavior, and a mesolimbic threshold process that gates the transformation of nociceptive activity to conscious pain. Adaptations in this threshold process are envisioned to be critical for development of chronic pain. We deconstruct chronic pain into four distinct phases, each with specific mechanisms, and outline current state of knowledge regarding these mechanisms: the limbic brain imparting risk, and the mesolimbic learning processes reorganizing the neocortex into a chronic pain state. Moreover, pain and negative moods are envisioned as a continuum of aversive behavioral learning, which enhance survival by protecting against threats.
Collapse
Affiliation(s)
- Marwan N Baliki
- Department of Physiology, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60610, USA.
| | - A Vania Apkarian
- Department of Physiology, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60610, USA; Department of Anesthesia, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60610, USA; Department of Physical Medicine and Rehabilitation, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60610, USA.
| |
Collapse
|
44
|
Guida F, Luongo L, Marmo F, Romano R, Iannotta M, Napolitano F, Belardo C, Marabese I, D'Aniello A, De Gregorio D, Rossi F, Piscitelli F, Lattanzi R, de Bartolomeis A, Usiello A, Di Marzo V, de Novellis V, Maione S. Palmitoylethanolamide reduces pain-related behaviors and restores glutamatergic synapses homeostasis in the medial prefrontal cortex of neuropathic mice. Mol Brain 2015; 8:47. [PMID: 26260027 PMCID: PMC4532244 DOI: 10.1186/s13041-015-0139-5] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/02/2015] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Enhanced supraspinal glutamate levels following nerve injury are associated with pathophysiological mechanisms responsible for neuropathic pain. Chronic pain can interfere with specific brain areas involved in glutamate-dependent neuropsychological processes, such as cognition, memory, and decision-making. The medial prefrontal cortex (mPFC) is thought to play a critical role in pain-related depression and anxiety, which are frequent co-morbidities of chronic pain. Using an animal model of spared nerve injury (SNI) of the sciatic nerve, we assess bio-molecular modifications in glutamatergic synapses in the mPFC that underlie neuropathic pain-induced plastic changes at 30 days post-surgery. Moreover, we examine the effects of palmitoylethanolamide (PEA) administration on pain-related behaviours, as well as the cortical biochemical and morphological changes that occur in SNI animals. RESULTS At 1 month, SNI was associated with mechanical and thermal hypersensitivity, as well as depression-like behaviour, cognitive impairments, and obsessive-compulsive activities. Moreover, we observed an overall glutamate synapse modification in the mPFC, characterized by changes in synaptic density proteins and amino acid levels. Finally, with regard to the resolution of pain and depressive-like syndrome in SNI mice, PEA restored the glutamatergic synapse proteins and changes in amino acid release. CONCLUSIONS Given the potential role of the mPFC in pain mechanisms, our findings may provide novel insights into neuropathic pain forebrain processes and indicate PEA as a new pharmacological tool to treat neuropathic pain and the related negative affective states. Graphical Abstract Palmitoylethanolamide: a new pharmacological tool to treat neuropathic pain and the related negative affective states.
Collapse
Affiliation(s)
- F Guida
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - L Luongo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - F Marmo
- Department of Neuroscience, Laboratory of Molecular and Translational Psychiatry, University School of Medicine "Federico II", Naples, Italy
| | - R Romano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - M Iannotta
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - F Napolitano
- Behavioural Neuroscience Laboratory, CEINGE - Biotecnologie Avanzate, Via Comunale Margherita 482, 80145, Naples, Italy
| | - C Belardo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - I Marabese
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - A D'Aniello
- Stazione Zoologica "Anton Dohrn", Naples, Italy
| | - D De Gregorio
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - F Rossi
- Department of Women, Child and General and Specialistic Surgery, Second University of Naples, Naples, Italy
| | - F Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - R Lattanzi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185, Rome, Italy
| | - A de Bartolomeis
- Department of Neuroscience, Laboratory of Molecular and Translational Psychiatry, University School of Medicine "Federico II", Naples, Italy
| | - A Usiello
- Behavioural Neuroscience Laboratory, CEINGE - Biotecnologie Avanzate, Via Comunale Margherita 482, 80145, Naples, Italy
| | - V Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078, Pozzuoli, NA, Italy
| | - V de Novellis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy
| | - S Maione
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, 80138, Naples, Italy.
| |
Collapse
|
45
|
Lu HC, Chang WJ, Kuan YH, Huang ACW, Shyu BC. A [14C]iodoantipyrine study of inter-regional correlations of neural substrates following central post-stroke pain in rats. Mol Pain 2015; 11:9. [PMID: 25889278 PMCID: PMC4358859 DOI: 10.1186/s12990-015-0006-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 02/17/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Central pain syndrome is characterized by a combination of abnormal pain sensations, and pain medications often provide little or no relief. Accumulating animal and clinical studies have shown that impairments of the spinothalamic tract (STT) and thalamocingulate pathway causes somatosensory dysfunction in central post-stroke pain (CPSP), but the involvement of other neuronal circuitries in CPSP has not yet been systematically examined. The aim of the present study was to evaluate changes in brain activity and neuronal circuitry using [(14)C]iodoantipyrine (IAP) in an animal model of CPSP. RESULTS Rats were subjected to lateral thalamic hemorrhage to investigate the characteristics of CPSP. Thermal and mechanical hyperalgesia developed in rats that were subjected to thalamic hemorrhagic lesion. The medial prefrontal cortex (mPFC), anterior cingulate cortex (ACC), striatum, thalamus, hypothalamus, and amygdala were more active in the CPSP group compared with rats that were not subjected to lateral thalamic hemorrhage. The inter-regional correlation analysis showed that regional cerebral blood flow in the mPFC was highly correlated with the amygdala in the right brain, and the right brain showed complex connections among subregions of the ACC. Rats with CPSP exhibited strong activation of the thalamocingulate and mPFC-amygdala pathways. CONCLUSIONS These results corroborate previous findings that the STT and thalamocingulate pathway are involved in the pathophysiological mechanisms of CPSP symptoms. The mPFC, amygdala, and periaqueductal gray emerged as having important correlations in pain processing in CPSP. The present data provide a basis for a neural correlation hypothesis of CPSP, with implications for CPSP treatment.
Collapse
Affiliation(s)
- Hsiang-Chin Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| | - Wei-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| | - Yung-Hui Kuan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| | | | - Bai Chuang Shyu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
46
|
Lin HC, Huang YH, Chao THH, Lin WY, Sun WZ, Yen CT. Gabapentin reverses central hypersensitivity and suppresses medial prefrontal cortical glucose metabolism in rats with neuropathic pain. Mol Pain 2014; 10:63. [PMID: 25253440 PMCID: PMC4182821 DOI: 10.1186/1744-8069-10-63] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/10/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Gabapentin (GBP) is known to suppress neuropathic hypersensitivity of primary afferents and the spinal cord dorsal horn. However, its supra-spinal action sites are unclear. We identify the brain regions where GBP changes the brain glucose metabolic rate at the effective dose that alleviates mechanical allodynia using 18 F-fluorodeoxyglucose-positron emission tomography (FDG-PET) scanning. RESULTS Comparing the PET imaging data before and after the GBP treatment, the spared nerve injury-induced increases of glucose metabolism in the thalamus and cerebellar vermis were reversed, and a significant decrease occurred in glucose metabolism in the medial prefrontal cortex (mPFC), including the anterior cingulate cortex. GBP treatment also reversed post-SNI connectivity increases between limbic cortices and thalamus. CONCLUSIONS Our results indicate that GBP analgesic effect may be mediated by reversing central hypersensitivity, and suppressing mPFC, a crucial part of the cortical representation of pain, in the brain.
Collapse
Affiliation(s)
- Hsiao-Chun Lin
- />Department of Life Science, National Taiwan University, No 1, Section 4, Roosevelt Road, Taipei, 10617 Taiwan
| | - Yu-Hsin Huang
- />Department of Anesthesiology, National Taiwan University Hospital, Taipei, 10002 Taiwan
| | - Tzu-Hao Harry Chao
- />Department of Life Science, National Taiwan University, No 1, Section 4, Roosevelt Road, Taipei, 10617 Taiwan
| | - Wen-Ying Lin
- />Department of Life Science, National Taiwan University, No 1, Section 4, Roosevelt Road, Taipei, 10617 Taiwan
- />Department of Anesthesiology, National Taiwan University Hospital, Taipei, 10002 Taiwan
| | - Wei-Zen Sun
- />Department of Anesthesiology, National Taiwan University Hospital, Taipei, 10002 Taiwan
| | - Chen-Tung Yen
- />Department of Life Science, National Taiwan University, No 1, Section 4, Roosevelt Road, Taipei, 10617 Taiwan
| |
Collapse
|
47
|
Mansour AR, Farmer MA, Baliki MN, Apkarian AV. Chronic pain: the role of learning and brain plasticity. Restor Neurol Neurosci 2014; 32:129-39. [PMID: 23603439 DOI: 10.3233/rnn-139003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Based on theoretical considerations and recent observations, we argue that continued suffering of chronic pain is critically dependent on the state of motivational and emotional mesolimbic-prefrontal circuitry of the brain. The plastic changes that occur within this circuitry in relation to nociceptive inputs dictate the transition to chronic pain, rendering the pain less somatic and more affective in nature. This theoretical construct is a strong departure from the traditional scientific view of pain, which has focused on encoding and representation of nociceptive signals. We argue that the definition of chronic pain can be recast, within the associative learning and valuation concept, as an inability to extinguish the associated memory trace, implying that supraspinal/cortical manipulations may be a more fruitful venue for adequately modulating suffering and related behavior for chronic pain. We briefly review the evidence generated to date for the proposed model and emphasize that the details of underlying mechanisms remain to be expounded.
Collapse
Affiliation(s)
- A R Mansour
- Department of Physiology, Surgery, and Rehabilitation Institute of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - M A Farmer
- Department of Physiology, Surgery, and Rehabilitation Institute of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - M N Baliki
- Department of Physiology, Surgery, and Rehabilitation Institute of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - A Vania Apkarian
- Department of Physiology, Surgery, and Rehabilitation Institute of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois Department of Anesthesia, Surgery, and Rehabilitation Institute of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
48
|
Jiang ZC, Pan Q, Zheng C, Deng XF, Wang JY, Luo F. Inactivation of the prelimbic rather than infralimbic cortex impairs acquisition and expression of formalin-induced conditioned place avoidance. Neurosci Lett 2014; 569:89-93. [PMID: 24726402 PMCID: PMC4382360 DOI: 10.1016/j.neulet.2014.03.074] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/19/2014] [Accepted: 03/31/2014] [Indexed: 11/20/2022]
Abstract
Conditioned place avoidance (CPA) paradigm has been used to investigate the affective component of pain. Although the anterior cingulate cortex (ACC) has been demonstrated to play an important role in the affective aspect of pain, whether the other prefrontal subdivisions are involved in pain-related aversion is unknown. The present study investigated the role of the prelimbic cortex (PL) and infralimbic cortex (IL) in the acquisition and expression of formalin-induced CPA (F-CPA) in rats. GABAA receptor agonist muscimol was bilaterally microinjected into PL/IL before or after the formalin-paired training, to explore the effect of temporary inactivation of PL/IL on the acquisition and expression of F-CPA, respectively. The results showed that inactivation of PL rather than IL impaired the acquisition and expression of F-CPA. Moreover, the PL inactivation did not block the acquisition of LiCl-induced CPA, suggesting that PL may be specifically implicated in the pain-emotion related encoding. These results indicate that PL but not IL is involved in the aversive dimension of pain.
Collapse
Affiliation(s)
- Zhao-Cai Jiang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Qi Pan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Chun Zheng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100039, China; Department of Psychology, Southwest University for Nationalities, Chengdu 610041, China
| | - Xiao-Fei Deng
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Jin-Yan Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China.
| | - Fei Luo
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, 16 Lincui Road, Beijing 100101, China
| |
Collapse
|
49
|
Hong W, Chen L, Xie J. Molecular basis underlying Mycobacterium tuberculosis D-cycloserine resistance. Is there a role for ubiquinone and menaquinone metabolic pathways? Expert Opin Ther Targets 2014; 18:691-701. [PMID: 24773568 DOI: 10.1517/14728222.2014.902937] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Tuberculosis remains a formidable threat to global public health. Multidrug-resistant tuberculosis presents increasing burden on the control strategy. D-Cycloserine (DCS) is an effective second-line drug against Mycobacterium tuberculosis (M. tuberculosis), the causative agent of tuberculosis. Though less potent than isoniazid (INH) and streptomycin, DCS is crucial for antibiotic-resistant tuberculosis. One advantage of DCS is that less drug-resistant M. tuberculosis is reported in comparison with first-line antituberculosis drugs such as INH and rifampin. AREAS COVERED In this review, we summarise our current knowledge of DCS, and review the drug target and low-level resistance of DCS in M. tuberculosis. We summarise the metabolism of D-alanine (D-Ala) and peptidoglycan biosynthesis in bacteria. We first compared the amino acid similarity of Mycobacterium alanine racemase and D-Ala:D-alanine ligase and quite unexpectedly found that the two enzymes are highly conserved among Mycobacterium. EXPERT OPINION We summarise the drug targets of DCS and possible mechanisms underlying its low-level resistance for the first time. One significant finding is that ubiquinone and menaquinone metabolism-related genes are novel genes underlying DCS resistance in Escherichia coli and with homologues in M. tuberculosis. Further understanding of DCS targets and basis for its low-level resistance might inspire us to improve the use of DCS or find better drug targets.
Collapse
Affiliation(s)
- Weiling Hong
- Southwest University, Institute of Modern Biopharmaceuticals, School of Life Sciences, State Key Laboratory Breeding Base of Eco-Enviroment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education , Beibei, Chongqing 400715 , China +86 23 68367 108 ; +86 23 68367 108 ;
| | | | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Most individuals who develop pain following an inciting event will return to a healthy state as the injury heals. However, a small percentage continue to suffer, that is, transition to chronic pain. Chronic pain may persist for years and is accompanied by cognitive abnormalities, as well as diminished quality of life. In animals, persistent pain is characterized by peripheral and spinal cord reorganization, and recent evidence in humans also indicates cortical reorganization. Yet, despite more than 30 years of research, there is little agreement on the neural mechanisms that mediate the transition from acute to chronic pain. RECENT FINDINGS In a longitudinal brain-imaging study, individuals who developed an intense back pain episode were followed over a 1-year period, during which pain and brain parameters were collected repeatedly. A smaller number of healthy individuals and chronic back pain patients were also studied concomitantly, as positive and negative controls. At the time of entry into the study, strength of synchrony between the medial prefrontal cortex and nucleus accumbens (i.e. functional connectivity) was predictive (>80% accuracy) of individuals who subsequently transition to chronicity 1 year later. SUMMARY Properties of the brain's emotional learning circuitry predict the transition to chronic pain. The involvement of this circuitry in pain remains mostly unexplored. Future human and animal model studies are necessary to unravel underlying mechanisms driving pain chronicity, with the potential of advancing novel therapeutics for preventing pain chronification.
Collapse
|