1
|
Upadhyay A, Gradwell MA, Vajtay TJ, Conner J, Sanyal AA, Azadegan C, Patel KR, Thackray JK, Bohic M, Imai F, Ogundare SO, Yoshida Y, Abdus-Saboor I, Azim E, Abraira VE. The dorsal column nuclei scale mechanical sensitivity in naive and neuropathic pain states. Cell Rep 2025; 44:115556. [PMID: 40202848 DOI: 10.1016/j.celrep.2025.115556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
During pathological conditions, tactile stimuli can aberrantly engage nociceptive pathways leading to the perception of touch as pain, known as mechanical allodynia. The brain stem dorsal column nuclei integrate tactile inputs, yet their role in mediating tactile sensitivity and allodynia remains understudied. We found that gracile nucleus (Gr) inhibitory interneurons and thalamus-projecting neurons are differentially innervated by primary afferents and spinal inputs. Functional manipulations of these distinct Gr neuronal populations bidirectionally shifted tactile sensitivity but did not affect noxious mechanical or thermal sensitivity. During neuropathic pain, Gr neurons exhibited increased sensory-evoked activity and asynchronous excitatory drive from primary afferents. Silencing Gr projection neurons or activating Gr inhibitory neurons in neuropathic mice reduced tactile hypersensitivity, and enhancing inhibition ameliorated paw-withdrawal signatures of neuropathic pain and induced conditioned place preference. These results suggest that Gr activity contributes to tactile sensitivity and affective, pain-associated phenotypes of mechanical allodynia.
Collapse
Affiliation(s)
- Aman Upadhyay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA; Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA
| | - Mark A Gradwell
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Thomas J Vajtay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - James Conner
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Arnab A Sanyal
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Chloe Azadegan
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Komal R Patel
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joshua K Thackray
- Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA
| | - Manon Bohic
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Simon O Ogundare
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, NY, USA; Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Victoria E Abraira
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Cell Biology and Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
2
|
Ginsberg AG, Lempka SF, Duan B, Booth V, Crodelle J. Mechanisms for dysregulation of excitatory-inhibitory balance underlying allodynia in dorsal horn neural subcircuits. PLoS Comput Biol 2025; 21:e1012234. [PMID: 39808669 PMCID: PMC11771949 DOI: 10.1371/journal.pcbi.1012234] [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: 06/07/2024] [Revised: 01/27/2025] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Chronic pain is a wide-spread condition that is debilitating and expensive to manage, costing the United States alone around $600 billion in 2010. In a common symptom of chronic pain called allodynia, non-painful stimuli produce painful responses with highly variable presentations across individuals. While the specific mechanisms remain unclear, allodynia is hypothesized to be caused by the dysregulation of excitatory-inhibitory (E-I) balance in pain-processing neural circuitry in the dorsal horn of the spinal cord. In this work, we analyze biophysically-motivated subcircuit structures that represent common motifs in neural circuits in laminae I-II of the dorsal horn. These circuits are hypothesized to be part of the neural pathways that mediate two different types of allodynia: static and dynamic. We use neural firing rate models to describe the activity of populations of excitatory and inhibitory interneurons within each subcircuit. By accounting for experimentally-observed responses under healthy conditions, we specify model parameters defining populations of subcircuits that yield typical behavior under normal conditions. Then, we implement a sensitivity analysis approach to identify the mechanisms most likely to cause allodynia-producing dysregulation of the subcircuit's E-I signaling. We find that disruption of E-I balance generally occurs either due to downregulation of inhibitory signaling so that excitatory neurons are "released" from inhibitory control, or due to upregulation of excitatory neuron responses so that excitatory neurons "escape" their inhibitory control. Which of these mechanisms is most likely to occur, the subcircuit components involved in the mechanism, and the proportion of subcircuits exhibiting the mechanism can vary depending on the subcircuit structure. These results suggest specific hypotheses about diverse mechanisms that may be most likely responsible for allodynia, thus offering predictions for the high interindividual variability observed in allodynia and identifying targets for further experimental studies on the underlying mechanisms of this chronic pain symptom.
Collapse
Affiliation(s)
- Alexander G. Ginsberg
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Scott F. Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Bo Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Victoria Booth
- Department of Mathematics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jennifer Crodelle
- Department of Mathematics and Statistics, Middlebury College, Middlebury, Vermont, United States of America
| |
Collapse
|
3
|
Upadhyay A, Gradwell MA, Vajtay TJ, Conner J, Sanyal AA, Azadegan C, Patel KR, Thackray JK, Bohic M, Imai F, Ogundare SO, Yoshida Y, Abdus-Saboor I, Azim E, Abraira VE. The Dorsal Column Nuclei Scale Mechanical Sensitivity in Naive and Neuropathic Pain States. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.20.581208. [PMID: 38712022 PMCID: PMC11071288 DOI: 10.1101/2024.02.20.581208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tactile perception relies on reliable transmission and modulation of low-threshold information as it travels from the periphery to the brain. During pathological conditions, tactile stimuli can aberrantly engage nociceptive pathways leading to the perception of touch as pain, known as mechanical allodynia. Two main drivers of peripheral tactile information, low-threshold mechanoreceptors (LTMRs) and postsynaptic dorsal column neurons (PSDCs), terminate in the brainstem dorsal column nuclei (DCN). Activity within the DRG, spinal cord, and DCN have all been implicated in mediating allodynia, yet the DCN remains understudied at the cellular, circuit, and functional levels compared to the other two. Here, we show that the gracile nucleus (Gr) of the DCN mediates tactile sensitivity for low-threshold stimuli and contributes to mechanical allodynia during neuropathic pain in mice. We found that the Gr contains local inhibitory interneurons in addition to thalamus-projecting neurons, which are differentially innervated by primary afferents and spinal inputs. Functional manipulations of these distinct Gr neuronal populations resulted in bidirectional changes to tactile sensitivity, but did not affect noxious mechanical or thermal sensitivity. During neuropathic pain, silencing Gr projection neurons or activating Gr inhibitory neurons was able to reduce tactile hypersensitivity, and enhancing inhibition was able to ameliorate paw withdrawal signatures of neuropathic pain, like shaking. Collectively, these results suggest that the Gr plays a specific role in mediating hypersensitivity to low-threshold, innocuous mechanical stimuli during neuropathic pain, and that Gr activity contributes to affective, pain-associated phenotypes of mechanical allodynia. Therefore, these brainstem circuits work in tandem with traditional spinal circuits underlying allodynia, resulting in enhanced signaling of tactile stimuli in the brain during neuropathic pain.
Collapse
Affiliation(s)
- Aman Upadhyay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
- Neuroscience PhD program at Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, USA
| | - Mark A Gradwell
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Thomas J Vajtay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - James Conner
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Arnab A Sanyal
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Chloe Azadegan
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Komal R Patel
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Joshua K Thackray
- Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Manon Bohic
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, New York City, New York, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA
| | - Simon O Ogundare
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York City, New York, USA
| | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, New York City, New York, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York City, New York, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Department of Biological Sciences, Columbia University, New York City, New York, USA
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Victoria E Abraira
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA; Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, New Jersey, USA
- Brain Health Institute, Rutgers University, Piscataway, New Jersey, USA
- Lead contact
| |
Collapse
|
4
|
Yang Y, Qu JY, Guo H, Zhou HY, Ruan X, Peng YC, Shen XF, Xiong J, Wang YL. Electroacupuncture at Sensitized Acupoints Relieves Somatic Referred Pain in Colitis Rats by Inhibiting Sympathetic-Sensory Coupling to Interfere with 5-HT Signaling Pathway. Chin J Integr Med 2024; 30:152-162. [PMID: 38038835 DOI: 10.1007/s11655-023-3565-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 12/02/2023]
Abstract
OBJECTIVE To investigate whether electroacupuncture (EA) at sensitized acupoints could reduce sympathetic-sensory coupling (SSC) and neurogenic inflammatory response by interfering with 5-hydroxytryptamine (5-HT)ergic neural pathways to relieve colitis and somatic referred pain, and explore the underlying mechanisms. METHODS Rats were treated with 5% dextran sodium sulfate (DSS) solution for 7 days to establish a colitis model. Twelve rats were randomly divided into the control and model groups according to a random number table (n=6). According to the "Research on Rat Acupoint Atlas", sensitized acupoints and non-sensitized acupoints were determined. Rats were randomly divided into the control, model, Zusanli-EA (ST 36), Dachangshu-EA (BL 25), and Xinshu (BL 15) groups (n=6), as well as the control, model, EA, and EA + GR113808 (a 5-HT inhibitor) groups (n=6). The rats in the control group received no treatment. Acupuncture was administered on 2 days after modeling using the stimulation pavameters: 1 mA, 2 Hz, for 30 min, with sparse and dense waves, for 14 consecutive days. GR113808 was injected into the tail vein at 5 mg/kg before EA for 10 min for 7 consecutive days. Mechanical sensitivity was assessed with von Frey filaments. Body weight and disease activity index (DAI) scores of rats were determined. Hematoxylin and eosin staining was performed to observe colon histopathology. SSC was analyzed by immunofluorescence staining. Immunohistochemical staining was performed to detect 5-HT and substance P (SP) expressions. The calcitonin gene-related peptide (CGRP) in skin tissue and tyrosine hydroxylase (TH) protein levels in DRG were detected by Western blot. The levels of hyaluronic acid (HA), bradykinin (BK), prostaglandin I2 (PGI2) in skin tissue, 5-HT, tryptophan hydroxylase 1 (TPH1), serotonin transporters (SERT), 5-HT 3 receptor (5-HT3R), and 5-HT 4 receptor (5-HT4R) in colon tissue were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS BL 25 and ST 36 acupoints were determined as sensitized acupoints, and BL 15 acupoint was used as a non-sensitized acupoint. EA at sensitized acupoints improved the DAI score, increased mechanical withdrawal thresholds, and alleviated colonic pathological damage of rats. EA at sensitized acupoints reduced SSC structures and decreased TH and CGRP expression levels (P<0.05). Furthermore, EA at sensitized acupoints reduced BK, PGI2, 5-HT, 5-HT3R and TPH1 levels, and increased HA, 5-HT4R and SERT levels in colitis rats (P<0.05). GR113808 treatment diminished the protective effect of EA at sensitized acupoints in colitis rats (P<0.05). CONCLUSION EA at sensitized acupoints alleviated DSS-induced somatic referred pain in colitis rats by interfering with 5-HTergic neural pathway, and reducing SSC inflammatory response.
Collapse
Affiliation(s)
- Ying Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, China
| | - Jin-Yu Qu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Neurology, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Hua Guo
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, 610041, China
| | - Hai-Ying Zhou
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xia Ruan
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Ying-Chun Peng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Neurology, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Xue-Fang Shen
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Neurology, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Jin Xiong
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
- Department of Neurology, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China
| | - Yi-Li Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, 610500, China.
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, China.
| |
Collapse
|
5
|
Sauer AK, Vigouroux M, Dougherty PM, Cata JP, Ingelmo PM. Pain Experience and Sensory Changes in Astronauts During and After Short-Lasting Commercial Spaceflight: A Proof-of-Concept Study. J Pain Res 2023; 16:4253-4266. [PMID: 38107368 PMCID: PMC10723599 DOI: 10.2147/jpr.s440630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/02/2023] [Indexed: 12/19/2023] Open
Abstract
Space travel has been associated with musculoskeletal pain, yet little is known about the nociceptive changes and pain experience during spaceflight. This preliminary study aims to investigate the pain experience and sensory alterations in astronauts following a 17-day mission to the International Space Station (ISS) on Axiom Space's AX-1 commercial space flight. Two participants were enrolled, and data were collected pre-flight, in-flight, post-flight, and three-month post-flight. Validated pain questionnaires assessed anxiety, catastrophizing, impact on physical and mental health, disability, and overall pain experience. Qualitative interviews were conducted post-landing and conditioned pain modulation (CPM) and quantitative sensory testing (QST) were performed. Both astronauts reported musculoskeletal pain during and after the flight, which was managed with anti-inflammatories and stretching techniques. Pain levels returned to baseline after three months. Pain questionnaires revealed heightened pain experiences in-flight and immediately post-flight, although their adequacy in assessing pain in space is uncertain. Qualitative interviews allowed astronauts to describe their pain experiences during the flight. Sensory changes included increased mechanical touch detection thresholds, temporal pain summation, heat pain thresholds, and differences in conditioned pain modulation post-flight. This preliminary study suggested that spaceflight may affect various aspects of sensory perception and regulation in astronauts, albeit in a variable manner. More data are needed to gain insight of on gain and loss of sensory functions during space missions. Further investigation into the multifactorial stressors affecting the somatosensory system during space travel could contribute to advancements in space and pain medicine.
Collapse
Affiliation(s)
- Andrea K Sauer
- Department of Anesthesia, University of Bonn, Bonn, Germany
| | - Marie Vigouroux
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children’s Hospital, Montreal, QC, Canada
| | - Patrick M Dougherty
- Department of Pain Medicine, The University of Texas – MD Anderson Cancer Center, Houston, TX, USA
| | - Juan Pablo Cata
- Department of Anesthesia and Perioperative Medicine, The University of Texas – MD Anderson Cancer Center, Houston, TX, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Pablo M Ingelmo
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children’s Hospital, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Research Institute, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
6
|
Liu F, Zhang YH, Zhang YY, Lin J, Liu YJ, Li YL, Fang ZH, Liao HL, Wang H, Shen JF. Phosphorylation of the AMPARs regulated by protein kinase C (PKC) and protein interacting with C-kinase 1 (PICK1) contribute to orofacial neuropathic pain. Brain Res 2023; 1820:148578. [PMID: 37709161 DOI: 10.1016/j.brainres.2023.148578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
The α-amino-3-hydroxy-5-methylisoxazole-4-isoxazolepropionic acid receptor (AMPAR) has been recognized to play a vital role in the development of neuropathic pain. Recent studies have indicated that protein kinase C (PKC) and protein interacting with C-kinase 1 (PICK1) are involved in the phosphorylation of AMPARs. However, whether PKC and PICK1 were involved in the AMPAR phosphorylation in the trigeminal ganglion (TG) to participate in orofacial neuropathic pain remains enigmatic. A behavioral test was utilized to evaluate the head withdrawal threshold (HWT) after chronic constriction injury of the infraorbital nerve (CCI-ION). The distribution and expression of GluA1, GluA2, PKC, and PICK1 were examined in the trigeminal ganglion (TG) by immunofluorescence, real-time reverse transcription-quantitative polymerase chain reaction, immunoblotting, and co-immunoprecipitation. Intra-ganglionic injections of drugs were performed to investigate the regulation mechanism. The present study demonstrated that CCI-ION-induced mechanical allodynia was maintained over at least 21 days. GluA1 and GluA2 were mainly expressed in the neurons. Trigeminal nerve injury potentiated the phosphorylation of GluA1, GluA2, and PKC in the TG, which was prevented by inhibiting PKC with chelerythrine chloride. Additionally, PICK1 colocalized and interacted with GluA2 in the TG. Following blocking PICK1 with FSC-231, the phosphorylation of GluA2 decreased. Finally, inhibition of PKC and PICK1 both alleviated mechanical allodynia in the whisker pad of CCI-ION mice. In conclusion, activation of PKC and PICK1 contribute to orofacial allodynia by regulating AMPAR phosphorylation in the TG of male mice, which provides potential therapeutic targets for alleviating orofacial neuropathic pain.
Collapse
Affiliation(s)
- Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu-Han Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhong-Han Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Lin Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Center for Stomatology, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Xu G, Gong W, Dong S, Hu G, Tang W, Yu H. Analysis of the Risk Factors for Mechanical Allodynia in Herpetic Neuralgia: A Retrospective Cross-Sectional Study. J Pain Res 2023; 16:3309-3318. [PMID: 37808462 PMCID: PMC10557995 DOI: 10.2147/jpr.s417454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Mechanical allodynia is reportedly common during herpetic neuralgia. The purpose of this study was to establish a risk prediction model to predict the individual risk of allodynia in herpetic neuralgia. Methods Three hundred and eighty-six patients with trunk herpetic neuralgia were divided into two regions, T2-5 and T6-11. The causality between allodynia and other factors was analyzed by a binary logistic regression model. Results 42.2% of subjects had allodynia, 137 suffered from dynamic allodynia, and 110 with dynamic allodynia experienced local sweating. The following 5 items as predictors determined this model: local sweating (Odd Ratio = 27.57, P<0.001), lesion location (Odd Ratio=2.46, P =0.017), pain intensity (Odd Ratio=1.38, P =0.020), pain duration (Odd Ratio=0.94, P =0.006), and local scars (Odd Ratio=0.07, P<0.001). The presence and development of allodynia are associated with local sweating. The positive proportion of the Iodine-starch test between the T2-5 (50.0%) with the T6-11 (23.7%) had a statistically significant difference (χ2=5.36, P=0.021). 29.5% of patients at the T2-6 had obvious sweating, which was different from only sticky feelings at the T6-11 (70.5%, χ2=10.88, P=0.001). 19.2% of patients with residual scars and allodynia was significantly lower than 48.5% of patients without allodynia (χ2=15.28, P<0.001). Conclusion This analysis suggests that local sweating is a concomitant symptom in dynamic allodynia, which imply the sympathetic nerves innervating the sweat glands of the skin were also involved during herpetic neuralgia. This may assist in the evaluation of dynamic allodynia and prove the role of sympathetic nerve intervention for herpetic neuralgia.
Collapse
Affiliation(s)
- Gang Xu
- Department of Rehabilitation Medicine, Affiliated Tenth People’s Hospital of Tongji University, Shanghai Tenth People’s Hospital, Shanghai, 200072, People’s Republic of China
- Department of Rehabilitation Medicine, Tongji University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Weiwei Gong
- Department of Rehabilitation Medicine, Tongji University School of Medicine, Shanghai, 200092, People’s Republic of China
- Department of Rehabilitation Medicine, Shanghai First Rehabilitation Hospital, Shanghai, 200090, People’s Republic of China
| | - Shihong Dong
- Department of Rehabilitation Medicine, Tongji University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Guojiong Hu
- Department of Rehabilitation Medicine, Tongji University School of Medicine, Shanghai, 200092, People’s Republic of China
- Department of Rehabilitation Medicine, Affiliated Yangzhi Rehabilitation Hospital of Tongji University, Shanghai, 201619, People’s Republic of China
| | - Weizhen Tang
- Department of Rehabilitation Medicine, Affiliated Tenth People’s Hospital of Tongji University, Shanghai Tenth People’s Hospital, Shanghai, 200072, People’s Republic of China
| | - Hecheng Yu
- Department of Rehabilitation Medicine, Affiliated Tenth People’s Hospital of Tongji University, Shanghai Tenth People’s Hospital, Shanghai, 200072, People’s Republic of China
| |
Collapse
|
8
|
Ehlers VL, Sadler KE, Stucky CL. Peripheral transient receptor potential vanilloid type 4 hypersensitivity contributes to chronic sickle cell disease pain. Pain 2023; 164:1874-1886. [PMID: 36897169 PMCID: PMC10363186 DOI: 10.1097/j.pain.0000000000002889] [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: 09/27/2022] [Accepted: 12/08/2022] [Indexed: 03/11/2023]
Abstract
ABSTRACT Debilitating pain affects the lives of patients with sickle cell disease (SCD). Current pain treatment for patients with SCD fail to completely resolve acute or chronic SCD pain. Previous research indicates that the cation channel transient receptor potential vanilloid type 4 (TRPV4) mediates peripheral hypersensitivity in various inflammatory and neuropathic pain conditions that may share similar pathophysiology with SCD, but this channel's role in chronic SCD pain remains unknown. Thus, the current experiments examined whether TRPV4 regulates hyperalgesia in transgenic mouse models of SCD. Acute blockade of TRPV4 alleviated evoked behavioral hypersensitivity to punctate, but not dynamic, mechanical stimuli in mice with SCD. TRPV4 blockade also reduced the mechanical sensitivity of small, but not large, dorsal root ganglia neurons from mice with SCD. Furthermore, keratinocytes from mice with SCD showed sensitized TRPV4-dependent calcium responses. These results shed new light on the role of TRPV4 in SCD chronic pain and are the first to suggest a role for epidermal keratinocytes in the heightened sensitivity observed in SCD.
Collapse
Affiliation(s)
- Vanessa L Ehlers
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | | |
Collapse
|
9
|
Presto P, Ji G, Ponomareva O, Ponomarev I, Neugebauer V. Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain. Int J Mol Sci 2023; 24:11944. [PMID: 37569320 PMCID: PMC10418916 DOI: 10.3390/ijms241511944] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic pain presents a therapeutic challenge due to the highly complex interplay of sensory, emotional-affective and cognitive factors. The mechanisms of the transition from acute to chronic pain are not well understood. We hypothesized that neuroimmune mechanisms in the amygdala, a brain region involved in the emotional-affective component of pain and pain modulation, play an important role through high motility group box 1 (Hmgb1), a pro-inflammatory molecule that has been linked to neuroimmune signaling in spinal nociception. Transcriptomic analysis revealed an upregulation of Hmgb1 mRNA in the right but not left central nucleus of the amygdala (CeA) at the chronic stage of a spinal nerve ligation (SNL) rat model of neuropathic pain. Hmgb1 silencing with a stereotaxic injection of siRNA for Hmgb1 into the right CeA of adult male and female rats 1 week after (post-treatment), but not 2 weeks before (pre-treatment) SNL induction decreased mechanical hypersensitivity and emotional-affective responses, but not anxiety-like behaviors, measured 4 weeks after SNL. Immunohistochemical data suggest that neurons are a major source of Hmgb1 in the CeA. Therefore, Hmgb1 in the amygdala may contribute to the transition from acute to chronic neuropathic pain, and the inhibition of Hmgb1 at a subacute time point can mitigate neuropathic pain.
Collapse
Affiliation(s)
- Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
10
|
Wang L, Su X, Yan J, Wu Q, Xu X, Wang X, Liu X, Song X, Zhang Z, Hu W, Liu X, Zhang Y. Involvement of Mrgprd-expressing nociceptors-recruited spinal mechanisms in nerve injury-induced mechanical allodynia. iScience 2023; 26:106764. [PMID: 37250305 PMCID: PMC10214713 DOI: 10.1016/j.isci.2023.106764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/17/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Mechanical allodynia and hyperalgesia are intractable symptoms lacking effective clinical treatments in patients with neuropathic pain. However, whether and how mechanically responsive non-peptidergic nociceptors are involved remains elusive. Here, we showed that von Frey-evoked static allodynia and aversion, along with mechanical hyperalgesia after spared nerve injury (SNI) were reduced by ablation of MrgprdCreERT2-marked neurons. Electrophysiological recordings revealed that SNI-opened Aβ-fiber inputs to laminae I-IIo and vIIi, as well as C-fiber inputs to vIIi, were all attenuated in Mrgprd-ablated mice. In addition, priming chemogenetic or optogenetic activation of Mrgprd+ neurons drove mechanical allodynia and aversion to low-threshold mechanical stimuli, along with mechanical hyperalgesia. Mechanistically, gated Aβ and C inputs to vIIi were opened, potentially via central sensitization by dampening potassium currents. Altogether, we uncovered the involvement of Mrgprd+ nociceptors in nerve injury-induced mechanical pain and dissected the underlying spinal mechanisms, thus providing insights into potential therapeutic targets for pain management.
Collapse
Affiliation(s)
- Liangbiao Wang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaojing Su
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jinjin Yan
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Qiaofeng Wu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiang Xu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xinyue Wang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xiaoqing Liu
- School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiaoyuan Song
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhi Zhang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Wei Hu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Xinfeng Liu
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Yan Zhang
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| |
Collapse
|
11
|
Qureshi S, Ali G, Muhammad T, Idrees M, Ullah S, Ali Khan S, Ullah R, Khan R, Ul-Haq Z, Haseeb Mohsin A, Kong IK. Thiadiazine-thione derivatives ameliorate STZ-induced diabetic neuropathy by regulating insulin and neuroinflammatory signaling. Int Immunopharmacol 2022; 113:109421. [PMID: 36403520 DOI: 10.1016/j.intimp.2022.109421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/18/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022]
Abstract
Diabetes Mellitus is accompanied by chronic hyperglycemia, inflammation, and related molecular processes, which leads to diabetic neuropathy. In this work, we tested Thiadiazine-thione (TDT) synthetic derivatives TDT1 and TDT2 against streptozotocin (STZ)-induced diabetic neuropathy. Sprague Dawley's rats, SH-SY5Y neuronal and BV2 microglial cells were employed in this work, followed by behavioral, biochemical, and morphological studies utilizing RT-qPCR, ELISA, Immunoblotting, immunohistochemistry, Immunofluorescence, and in silico analyses. TDT1 and TDT2 abolished STZ-induced allodynia and hyperalgesia. Next, we examined IRS1/PI3K/AKT signaling to assess TDT1 and TDT2's impact on diabetic neuropathy. STZ downregulated IRS1, PI3K, AKT mRNA and protein expression in rat spinal cord and SH-SY5Y neuronal cells. TDT1 and TDT2 improved IRS1, PI3k, and AKT mRNA and protein expression. STZ elevated GSK3β mRNA and protein expression in vivo and in vitro, whereas TDT1 and TDT2 mitigated it. STZ increased the expression of inflammatory mediators such as p-NF-κB, TNF-α, and COX-2 in rat spinal cord lysates. TDT1 and TDT2 co-treatment with STZ decreased inflammatory cytokine expression by ameliorating astrocytosis (revealed by increased GFAP) and microgliosis (indicated by increased Iba1). TDT1 and TDT2 reduced STZ-induced JNK, Iba1, and COX-2 upregulation in BV2 microglial cells validating our in vivo findings. In silico molecular docking and MD simulations analyses suggested that TDT1 and TDT2 have IRS binding affinity, however, both compounds had an identical binding affinity, but distinct interaction pattern with IRS protein residues. Overall, these findings demonstrate that TDT derivatives mitigated STZ-induced neuropathy through modulating the insulin and inflammatory signaling pathways.
Collapse
Affiliation(s)
- Sonia Qureshi
- Department of Pharmacy, University of Peshawar, Peshawar, 25120, Pakistan; Krembil Research Institute, University Health Network, M5G 1L7, Toronto, Ontario, Canada
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar, 25120, Pakistan.
| | - Tahir Muhammad
- Molecular Neuropsychiatry and Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Muhammad Idrees
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Gyeongnam Province, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Gyeongnam Province, Republic of Korea
| | - Sultan Ullah
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, 33458, USA
| | - Salman Ali Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rahim Ullah
- Department of Pharmacy, University of Peshawar, Peshawar, 25120, Pakistan
| | - Rasool Khan
- Institute of chemical sciences, University of Peshawar, Peshawar, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan; Institute of chemical sciences, University of Peshawar, Peshawar, Pakistan
| | | | - Il-Keun Kong
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju 52828, Gyeongnam Province, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Gyeongnam Province, Republic of Korea; The Kingkong Co. Ltd., Gyeongsang National University, Jinju 52828, Gyeongnam Province, Republic of Korea
| |
Collapse
|
12
|
Bruneau A, Carrié S, Moscaritolo L, Ingelmo P. Mechanism-Based Pharmacological Treatment for Chronic Non-cancer Pain in Adolescents: Current Approaches and Future Directions. Paediatr Drugs 2022; 24:573-583. [PMID: 36053398 DOI: 10.1007/s40272-022-00534-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 11/26/2022]
Abstract
Chronic non-cancer pain in pediatrics is a widespread phenomenon that affects about 20% of adolescents (10-19 years old). Although interdisciplinary pain treatment programs, which often include pharmacological treatment, have emerged as the standard of care in management of this patient population, evidence regarding an optimal treatment is lacking. The efficacy and safety profiles of pharmacological treatments used to help adolescents suffering from chronic non-cancer pain remain understudied. This lack of evidence may increase polypharmacy and the risk of drug interactions and adverse events. This review examines evidence for the use of pharmacological treatments prescribed to treat chronic pain in adolescents (10-19 years old), with a focus on mechanism-based pharmacology. The objectives of this review are to: (a) review the evidence for mechanism-based pharmacological treatments for chronic non-cancer pain in adolescents and (b) describe the pharmacological agents that are commonly prescribed to manage chronic pain in adolescents, including dosage information, mechanism, and potential adverse effects. Pharmacological treatments should be used carefully with adolescents, ideally within an interdisciplinary treatment program that will incorporate physical rehabilitation, integrative medicine/active mind-body techniques, psychology, and global efforts to normalize daily activities.
Collapse
Affiliation(s)
- Alice Bruneau
- Faculty of Medicine, McGill University, Montreal, QC, H3A 0C7, Canada.
| | - Sabrina Carrié
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children's Hospital, Montreal, QC, Canada
- Department of Anaesthesia, McGill University, Montreal, QC, Canada
| | - Lorenzo Moscaritolo
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children's Hospital, Montreal, QC, Canada
- Department of Anaesthesia, McGill University, Montreal, QC, Canada
| | - Pablo Ingelmo
- Edwards Family Interdisciplinary Center for Complex Pain, Montreal Children's Hospital, Montreal, QC, Canada
- Department of Anaesthesia, McGill University, Montreal, QC, Canada
- Alan Edward Centre for Research on Pain, Montreal, QC, Canada
- Research Institute, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
13
|
Presto P, Neugebauer V. Sex Differences in CGRP Regulation and Function in the Amygdala in a Rat Model of Neuropathic Pain. Front Mol Neurosci 2022; 15:928587. [PMID: 35726298 PMCID: PMC9206543 DOI: 10.3389/fnmol.2022.928587] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/12/2022] [Indexed: 12/02/2022] Open
Abstract
The amygdala has emerged as a key player in the emotional response to pain and pain modulation. The lateral and capsular regions of the central nucleus of the amygdala (CeA) represent the “nociceptive amygdala” due to their high content of neurons that process pain-related information. These CeA divisions are the targets of the spino-parabrachio-amygdaloid pain pathway, which is the predominant source of calcitonin gene-related peptide (CGRP) within the amygdala. Changes in lateral and capsular CeA neurons have previously been observed in pain models, and synaptic plasticity in these areas has been linked to pain-related behavior. CGRP has been demonstrated to play an important role in peripheral and spinal mechanisms, and in pain-related amygdala plasticity in male rats in an acute arthritis pain model. However, the role of CGRP in chronic neuropathic pain-related amygdala function and behaviors remains to be determined for both male and female rats. Here we tested the hypothesis that the CGRP1 receptor is involved in neuropathic pain-related amygdala activity, and that blockade of this receptor can inhibit neuropathic pain behaviors in both sexes. CGRP mRNA expression levels in the CeA of male rats were upregulated at the acute stage of the spinal nerve ligation (SNL) model of neuropathic pain, whereas female rats had significantly higher CGRP and CGRP receptor component expression at the chronic stage. A CGRP1 receptor antagonist (CGRP 8-37) administered into the CeA in chronic neuropathic rats reduced mechanical hypersensitivity (von Frey and paw compression tests) in both sexes but showed female-predominant effects on emotional-affective responses (ultrasonic vocalizations) and anxiety-like behaviors (open field test). CGRP 8-37 inhibited the activity of CeA output neurons assessed with calcium imaging in brain slices from chronic neuropathic pain rats. Together, these findings may suggest that CGRP1 receptors in the CeA are involved in neuropathic pain-related amygdala activity and contribute to sensory aspects in both sexes but to emotional-affective pain responses predominantly in females. The sexually dimorphic function of CGRP in the amygdala would make CGRP1 receptors a potential therapeutic target for neuropathic pain relief, particularly in females in chronic pain conditions.
Collapse
Affiliation(s)
- Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- *Correspondence: Volker Neugebauer
| |
Collapse
|
14
|
Sadler KE, Mogil JS, Stucky CL. Innovations and advances in modelling and measuring pain in animals. Nat Rev Neurosci 2022; 23:70-85. [PMID: 34837072 PMCID: PMC9098196 DOI: 10.1038/s41583-021-00536-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
Best practices in preclinical algesiometry (pain behaviour testing) have shifted over the past decade as a result of technological advancements, the continued dearth of translational progress and the emphasis that funding institutions and journals have placed on rigour and reproducibility. Here we describe the changing trends in research methods by analysing the methods reported in preclinical pain publications from the past 40 years, with a focus on the last 5 years. We also discuss how the status quo may be hampering translational success. This discussion is centred on four fundamental decisions that apply to every pain behaviour experiment: choice of subject (model organism), choice of assay (pain-inducing injury), laboratory environment and choice of outcome measures. Finally, we discuss how human tissues, which are increasingly accessible, can be used to validate the translatability of targets and mechanisms identified in animal pain models.
Collapse
Affiliation(s)
- Katelyn E Sadler
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, QC, Canada
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Cheryl L Stucky
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
15
|
Qureshi S, Ali G, Idrees M, Muhammad T, Kong IK, Abbas M, Shah MIA, Ahmad S, Sewell RDE, Ullah S. Selected Thiadiazine-Thione Derivatives Attenuate Neuroinflammation in Chronic Constriction Injury Induced Neuropathy. Front Mol Neurosci 2021; 14:728128. [PMID: 34975395 PMCID: PMC8716630 DOI: 10.3389/fnmol.2021.728128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Neuropathic pain refers to a lesion or disease of peripheral and/or central somatosensory neurons and is an important body response to actual or potential nerve damage. We investigated the therapeutic potential of two thiadiazine-thione [TDT] derivatives, 2-(5-propyl-6-thioxo-1, 3, 5-thiadiazinan-3-yl) acetic acid [TDT1] and 2-(5-propyl-2-thioxo-1, 3, 5-thiadiazinan-3-yl) acetic acid [TDT2] against CCI (chronic constriction injury)-induced neuroinflammation and neuropathic pain. Mice were used for assessment of acute toxicity of TDT derivatives and no major toxic/bizarre responses were observed. Anti-inflammatory activity was assessed using the carrageenan test, and both TDT1 and TDT2 significantly reduced carrageenan-induced inflammation. We also used rats for the induction of CCI and performed allodynia and hyperalgesia-related behavioral tests followed by biochemical and morphological analysis using RT-qPCR, immunoblotting, immunohistochemistry and immunofluorescence. Our findings revealed that CCI induced clear-cut allodynia and hyperalgesia which was reversed by TDT1 and TDT2. To determine the function of TDT1 and TDT2 in glia-mediated neuroinflammation, Iba1 mRNA and protein levels were measured in spinal cord tissue sections from various experimental groups. Interestingly, TDT1 and TDT2 substantially reduced the mRNA expression and protein level of Iba1, implying that TDT1 and TDT2 may mitigate CCI-induced astrogliosis. In silico molecular docking studies predicted that both compounds had an effective binding affinity for TNF-α and COX-2. The compounds interactions with the proteins were dominated by both hydrogen bonding and van der Waals interactions. Overall, these results suggest that TDT1 and TDT2 exert their neuroprotective and analgesic potentials by ameliorating CCI-induced allodynia, hyperalgesia, neuroinflammation and neuronal degeneration in a dose-dependent manner.
Collapse
Affiliation(s)
- Sonia Qureshi
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
- Laboratory of Neurogenomics and Novel Therapies, The Ken and Ruth Davee Department of Neurology, Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Muhammad Idrees
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju, South Korea
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
| | - Tahir Muhammad
- Molecular Neuropsychiatry and Development (MiND) Lab, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Il-Keun Kong
- Division of Applied Life Science (BK21 Four), Gyeongsang National University, Jinju, South Korea
- Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, South Korea
- The Kingkong Co., Ltd., Gyeongsang National University, Jinju, South Korea
| | - Muzaffar Abbas
- Faculty of Pharmacy, Capital University of Science & Technology, Islamabad, Pakistan
| | | | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Robert D. E. Sewell
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - Sami Ullah
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| |
Collapse
|
16
|
Uta D, Tsuboshima K, Nishijo H, Mizumura K, Taguchi T. Neuronal Sensitization and Synaptic Facilitation in the Superficial Dorsal Horn of a Rat Reserpine-induced Pain Model. Neuroscience 2021; 479:125-139. [PMID: 34673142 DOI: 10.1016/j.neuroscience.2021.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022]
Abstract
Chronic widespread pain is one of the important issues to be solved in medical practice. Impaired spinal descending pain inhibitory system due to decreased monoamine neurotransmitters is assumed to cause nociceptive hypersensitivities in chronic painful conditions like that described in patients with fibromyalgia (FM). However, response behaviors and synaptic transmission of the spinal dorsal horn neurons in response to reserpine remain to be clarified. Here we examined the activities of superficial dorsal horn (SDH) neurons, as well as excitatory and inhibitory postsynaptic inputs to SDH neurons, using a putative rat model of FM that was established by injecting reserpine. Extracellular recordings in vivo revealed that SDH neurons were sensitized to mechanical stimulation applied to the neurons' receptive fields, and the mechanically sensitized neurons were spontaneously more active. The sensitizing effect was evident 1 day and 3 days after the reserpine treatment, but subsided 5 days after the treatment or later. Using patch-clamp recordings in vivo, spontaneous excitatory postsynaptic currents (sEPSCs) to SDH neurons were found to increase in the pain model, while spontaneous inhibitory postsynaptic currents (sIPSCs) to SDH neurons decreased. These results demonstrate that the SDH neurons were strongly sensitized in response to the reserpine treatment, and that increased excitatory and decreased inhibitory postsynaptic inputs could be responsible for the spinal nociceptive hypersensitivity in the putative FM model.
Collapse
Affiliation(s)
- Daisuke Uta
- Department of Applied Pharmacology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Katsuyuki Tsuboshima
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Hisao Nishijo
- System Emotional Science, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Kazue Mizumura
- Department of Physiology, Nihon University School of Dentistry, Tokyo 101-8310, Japan
| | - Toru Taguchi
- Department of Physical Therapy, Faculty of Rehabilitation, Niigata University of Health and Welfare, Niigata 950-3198, Japan; Institute for Human Movement and Medical Sciences (IHMMS), Niigata University of Health and Welfare, Niigata 950-3198, Japan.
| |
Collapse
|
17
|
Foxo1 selectively regulates static mechanical pain by interacting with Nav1.7. Pain 2021; 162:490-502. [PMID: 32868747 DOI: 10.1097/j.pain.0000000000002055] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
ABSTRACT Mechanical allodynia is a debilitating condition for millions of patients with chronic pain. Mechanical allodynia can manifest in distinct forms, including brush-evoked dynamic and filament-evoked static allodynia. In the nervous system, the forkhead protein Foxo1 plays a critical role in neuronal structures and functions. However, the role of Foxo1 in the somatosensory signal remains unclear. Here, we found that Foxo1 selectively regulated static mechanical pain. Foxo1 knockdown decreased sensitivity to static mechanical stimuli in normal rats and attenuated static mechanical allodynia in rat models for neuropathic, inflammatory, and chemotherapy pain. Conversely, Foxo1 overexpression selectively enhanced sensitivity to static mechanical stimuli and provoked static mechanical allodynia. Furthermore, Foxo1 interacted with voltage-gated sodium Nav1.7 channels and increased the Nav1.7 current density by accelerating activation rather than by changing the expression of Nav1.7 in dorsal root ganglia neurons. In addition, the serum level of Foxo1 was found to be increased in chronic pain patients and to be positively correlated with the severity of chronic pain. Altogether, our findings suggest that serum Foxo1 level could be used as a biological marker for prediction and diagnosis of chronic pain. Moreover, selective blockade of Foxo1/Nav1.7 interaction may offer a new therapeutic approach in patients with mechanical pain.
Collapse
|
18
|
Hu R, Zhang J, Liu X, Huang D, Cao YQ. Low-Dose Interleukin-2 and Regulatory T Cell Treatments Attenuate Punctate and Dynamic Mechanical Allodynia in a Mouse Model of Sciatic Nerve Injury. J Pain Res 2021; 14:893-906. [PMID: 33854366 PMCID: PMC8040486 DOI: 10.2147/jpr.s301343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/13/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Nerve injury-induced mechanical hyper-sensitivity, in particular stroking-induced dynamic allodynia, is highly debilitating and difficult to treat. Previous studies indicate that the immunosuppressive regulatory T (Treg) cells modulate the magnitude of punctate mechanical allodynia resulting from sciatic nerve injury. However, whether enhancing Treg-mediated suppression attenuates dynamic allodynia is not known. In the present study, we addressed this knowledge gap by treating mice with low-dose interleukin-2 (ld-IL2) injections or adoptive transfer of Treg cells. Methods Female Swiss Webster mice received daily injections of ld-IL2 (1 μg/mouse, intraperitoneally) either before or after unilateral spared nerve injury (SNI). Male C57BL/6J mice received adoptive transfer of 1 x 106 Treg cells 3 weeks post-SNI. The responses to punctate and dynamic mechanical stimuli on the hindpaw were monitored before and up to 4–6 weeks post-SNI. We also compared the distribution of Treg cells and CD3+ total T cells after SNI and/or ld-IL2 treatment. Results Ld-IL2 pretreatment in female Swiss Webster mice completely blocked the development of SNI-induced dynamic mechanical allodynia and reduced the magnitude of punctate allodynia. Delayed ld-IL2 treatment in female mice significantly attenuated the morphine-resistant punctate and dynamic allodynia at 3–5 weeks post-SNI. Adoptive transfer of Treg cells to male C57BL/6J mice 3 weeks post-SNI effectively reversed the persistent punctate and dynamic allodynia, supporting that the effect of ld-IL2 is mediated through endogenous Treg cells, and is likely independent of mouse strain and sex. Neither ld-IL2 treatment nor Treg transfer affected the basal responses to punctate or brush stimuli. Ld-IL2 significantly increased the frequency of Treg cells among total CD3+ T cells in the injured sciatic nerves but not in the uninjured nerves or the dorsal root ganglia, suggesting the injured nerve as ld-IL2’s site of action. Conclusion Collectively, results from the present study supports Treg as a cellular target and ld-IL2 as a potential therapeutic option for nerve injury-induced persistent punctate and dynamic mechanical allodynia.
Collapse
Affiliation(s)
- Rong Hu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pain Management, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jintao Zhang
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xuemei Liu
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Dong Huang
- Department of Pain Management, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Yu-Qing Cao
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA.,Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
Prevalence and characteristics of cutaneous allodynia in probable migraine. Sci Rep 2021; 11:2467. [PMID: 33510340 PMCID: PMC7844001 DOI: 10.1038/s41598-021-82080-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/15/2021] [Indexed: 12/31/2022] Open
Abstract
Cutaneous allodynia (CA) is a pain in response to non-nociceptive stimulation and a marker of central sensitisation. Probable migraine (PM) is a migraine subtype that fulfils all but one criterion of migraine. Headache intensity and the disability of individuals with PM are similar or lower than individuals with migraine. This study compared CA prevalence and characteristics of PM and migraine using a nationally representative sample in Korea. The Allodynia Symptom Checklist-12 (ASC-12) was used to assess CA (ASC-12 score ≥ 3). PM and migraine prevalence were 11.6% and 5.0%, respectively. CA prevalence did not significantly differ between PM and migraine (14.5% vs. 16.0%, p = 0.701). Participants with PM with CA reported a higher monthly headache frequency (3.3 ± 4.3 vs. 1.8 ± 3.6, p = 0.044), more severe headache intensity (Visuals Analogue Scale, 6.0 [4.0–7.0] vs. 5.0 [3.0–6.0], p = 0.002), and higher impact of headache (Headache Impact Test-6, 56.3 ± 7.2 vs. 48.3 ± 8.0, p < 0.001) than those without CA. Multiple regression analyses revealed that headache frequency and intensity, anxiety, and depression were significant factors for CA in participants with PM. In conclusion, CA prevalence among participants with PM and migraine were comparable. Anxiety, depression, and headache frequency and intensity were significant factors for CA in participants with PM.
Collapse
|
20
|
Terayama R, Uchibe K. Reorganization of synaptic inputs to spinal dorsal horn neurons in neuropathic pain. Int J Neurosci 2021; 132:1210-1216. [PMID: 33428497 DOI: 10.1080/00207454.2021.1873980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Peripheral nerve injuries produce a variety of negative structural and functional changes in the central terminal sites of damaged axons, as well as the injured primary afferents. Such changes have been shown to be involved in the development of neuropathic pain, which includes abnormal pain sensations such as allodynia and hyperalgesia. Since the spinal dorsal horn is the first central site where signals from peripheral sensory nerves are transmitted and shows a variety of changes after peripheral nerve injury or chronic inflammation of peripheral tissues, it is one of the most important sites contributing to the mechanisms underlying the development of neuropathic pain. The functional disruption of inhibitory interneurons and glial activation in the spinal dorsal horn after peripheral nerve injury cause reorganization of neuronal circuits and changes in the excitability of second-order neurons. These events are involved in the development or maintenance of neuropathic pain. Here, we describe the interactions of primary afferents, interneurons, and glial cells that may cause reorganization of synaptic inputs to spinal dorsal horn neurons after peripheral nerve injury.
Collapse
Affiliation(s)
- Ryuji Terayama
- Department of Maxillofacial Anatomy and Neuroscience, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kenta Uchibe
- Department of Maxillofacial Anatomy and Neuroscience, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
21
|
Liu F, Yajima T, Wang M, Shen JF, Ichikawa H, Sato T. Effects of trigeminal nerve injury on the expression of galanin and its receptors in the rat trigeminal ganglion. Neuropeptides 2020; 84:102098. [PMID: 33069139 DOI: 10.1016/j.npep.2020.102098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 11/24/2022]
Abstract
In the spinal nervous system, the expression of galanin (GAL) and galanin receptors (GALRs) that play important roles in the transmission and modulation of nociceptive information can be affected by nerve injury. However, in the trigeminal nervous system, the effects of trigeminal nerve injury on the expression of GAL are controversy in the previous studies. Besides, little is known about the effects of trigeminal nerve injury on the expression of GALRs. In the present study, the effects of trigeminal nerve injury on the expression of GAL and GALRs in the rat trigeminal ganglion (TG) were investigated by using quantitative real-time reverse transcription-polymerase chain reaction and immunohistochemistry. To identify the nerve-injured and nerve-uninjured TG neurons, activating transcription factor 3 (ATF3, the nerve-injured neuron marker) was stained by immunofluorescence. The levels of GAL mRNA in the rostral half and caudal half of the TG dramatically increased after transection of infraorbital nerve (ION) and inferior alveolar nerve (IAN), respectively. Immunohistochemical labeling of GAL and ATF3 revealed that GAL level was elevated in both injured and adjacent uninjured small and medium-sized TG neurons after ION/IAN transection. In addition, the levels of GAL2R-like immunoreactivity were reduced in both injured and adjacent uninjured TG neurons after ION/IAN transection, while levels of GAL1R and GAL3R-like immunoreactivity remained unchanged. Furthermore, the number of small to medium-sized TG neurons co-expressing GAL- and GAL1R/GAL2R/GAL3R-like immunoreactivity was significantly increased after ION/IAN transection. In line with previous studies in other spinal neuron systems, these results suggest that GAL and GALRs play functional roles in orofacial neuropathic pain and trigeminal nerve regeneration after trigeminal nerve injury.
Collapse
Affiliation(s)
- Fei Liu
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo machi, Sendai 980-8575, Japan; State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department II of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province 610041, China.
| | - Takehiro Yajima
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo machi, Sendai 980-8575, Japan
| | - Min Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department II of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department II of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Hiroyuki Ichikawa
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo machi, Sendai 980-8575, Japan
| | - Tadasu Sato
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo machi, Sendai 980-8575, Japan
| |
Collapse
|
22
|
Mustonen L, Aho T, Harno H, Kalso E. Static mechanical allodynia in post-surgical neuropathic pain after breast cancer treatments. Scand J Pain 2020; 20:683-691. [PMID: 32697763 DOI: 10.1515/sjpain-2020-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/19/2020] [Indexed: 11/15/2022]
Abstract
Objectives Static mechanical allodynia (SMA), i. e., pain caused by normally non-painful static pressure, is a prevalent manifestation of neuropathic pain (NP). Although SMA may significantly affect the patient's daily life, it is less well studied in the clinical context. We aimed to characterize SMA in women with chronic post-surgical NP (CPSNP) after breast cancer surgery. Our objective was to improve understanding of the clinical picture of this prevalent pain condition. This is a substudy of a previously published larger cohort of patients with intercostobrachial nerve injury after breast cancer surgery (Mustonen et al. Pain. 2019;160:246-56). Methods We studied SMA in 132 patients with CPSNP after breast cancer surgery. The presence, location, and intensity of SMA were assessed at clinical sensory examination. The patients gave self-reports of pain with the Brief Pain Inventory (BPI). We studied the association of SMA to type of surgery, oncological treatments, BMI, other pains, and psychological factors. General pain sensitivity was assessed by the cold pressor test. Results SMA was prevalent (84%) in this cohort whereas other forms of allodynia were scarce (6%). Moderate-to-severe SMA was frequently observed even in patients who reported mild pain in BPI. Breast and the side of chest were the most common locations of SMA. SMA was associated with breast surgery type, but not with psychological factors. Severe SMA, but not self-reported pain, was associated with lower cold pain tolerance. Conclusions SMA is prevalent in post-surgical NP after breast cancer surgery and it may represent a distinct NP phenotype. High intensities of SMA may signal the presence of central sensitization. Implications SMA should be considered when examining and treating patients with post-surgical NP after breast cancer surgery.
Collapse
Affiliation(s)
- Laura Mustonen
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Neurocenter, Neurology, University of Helsinki, and Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Tommi Aho
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Harno
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Neurocenter, Neurology, University of Helsinki, and Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Eija Kalso
- Division of Pain Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
23
|
Jessri M, Sultan AS, Tavares T, Schug S. Central mechanisms of pain in orofacial pain patients: Implications for management. J Oral Pathol Med 2020; 49:476-483. [PMID: 32539196 DOI: 10.1111/jop.13062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Central sensitization (CS) is a form of neuroplasticity characterized by changes in the neural sensitivity, responsiveness, and/or output that are not contingent on peripheral input nor activity-dependent. CS is characterized by activation of unmyelinated C-fibers resulting in a cascade of events at molecular and cellular levels which eventuate into generation of synaptic currents at rest. CS, therefore, contributes to heightened generalized pain sensitivity, further complicates the process of reaching a diagnosis, and increases the possibility of treatment failure. BODY: Trigeminal nerve is the main sensory supplier of the anterior part of the head, including the intraoral structures. Primary afferent nociceptors of the trigeminal nerve and low threshold mechanoreceptors synapse with wide dynamic range (WDR) neurons in the pons. This multifaceted network of nerve interactions which is further complicated by the modulatory circuits that can suppress or heighten the activity of WDR neurons is one of the main contributors to CS. The importance of CS in orofacial pain disorders is emphasized in the context of chronic pain development. As for all chronic pain conditions, it is crucial to consider the biopsychosocial aspects of chronic orofacial pain in managing this diverse group of conditions. This review highlights current understanding of the biopsychosocial model and central mechanisms contributing to the pathogenesis of chronic orofacial pain.
Collapse
Affiliation(s)
- Maryam Jessri
- UWA Dental School, The University of Western Australia, Nedlands, WA, Australia
| | - Ahmed S Sultan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Tiffany Tavares
- Department of Oral Pathology, Radiology, and Medicine, University of Missouri, Kansas City, MO, USA
| | - Stephan Schug
- Anaesthesiology and Pain Medicine, UWA Medical School, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
24
|
Optogenetic Inhibition of CGRPα Sensory Neurons Reveals Their Distinct Roles in Neuropathic and Incisional Pain. J Neurosci 2019; 38:5807-5825. [PMID: 29925650 DOI: 10.1523/jneurosci.3565-17.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/29/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023] Open
Abstract
Cutaneous somatosensory neurons convey innocuous and noxious mechanical, thermal, and chemical stimuli from peripheral tissues to the CNS. Among these are nociceptive neurons that express calcitonin gene-related peptide-α (CGRPα). The role of peripheral CGRPα neurons (CANs) in acute and injury-induced pain has been studied using diphtheria toxin ablation, but their functional roles remain controversial. Because ablation permanently deletes a neuronal population, compensatory changes may ensue that mask the physiological or pathophysiological roles of CANs, particularly for injuries that occur after ablation. Therefore, we sought to define the role of intact CANs in vivo under baseline and injury conditions by using noninvasive transient optogenetic inhibition. We assessed pain behavior longitudinally from acute to chronic time points. We generated adult male and female mice that selectively express the outward rectifying proton pump archaerhodopsin-3 (Arch) in CANs, and inhibited their peripheral cutaneous terminals in models of neuropathic (spared nerve injury) and inflammatory (skin-muscle incision) pain using transdermal light activation of Arch. After nerve injury, brief activation of Arch reversed the chronic mechanical, cold, and heat hypersensitivity, alleviated the spontaneous pain, and reversed the sensitized mechanical currents in primary afferent somata. In contrast, Arch inhibition of CANs did not alter incision-induced hypersensitivity. Instead, incision-induced mechanical and heat hypersensitivity was alleviated by peripheral blockade of CGRPα peptide-receptor signaling. These results reveal that CANs have distinct roles in the time course of pain during neuropathic and incisional injuries and suggest that targeting peripheral CANs or CGRPα peptide-receptor signaling could selectively treat neuropathic or postoperative pain, respectively.SIGNIFICANCE STATEMENT The contribution of sensory afferent CGRPα neurons (CANs) to neuropathic and inflammatory pain is controversial. Here, we left CANs intact during neuropathic and perioperative incision injury by using transient transdermal optogenetic inhibition of CANs. We found that peripheral CANs are required for neuropathic mechanical, cold, and heat hypersensitivity, spontaneous pain, and sensitization of mechanical currents in afferent somata. However, they are dispensable for incisional pain transmission. In contrast, peripheral pharmacological inhibition of CGRPα peptide-receptor signaling alleviated the incisional mechanical and heat hypersensitivity, but had no effect on neuropathic pain. These results show that CANs have distinct roles in neuropathic and incisional pain and suggest that their targeting via novel peripheral treatments may selectively alleviate neuropathic versus incisional pain.
Collapse
|
25
|
Chen Y, Shi Y, Wang G, Li Y, Cheng L, Wang Y. Memantine selectively prevented the induction of dynamic allodynia by blocking Kir2.1 channel and inhibiting the activation of microglia in spinal dorsal horn of mice in spared nerve injury model. Mol Pain 2019; 15:1744806919838947. [PMID: 30845882 PMCID: PMC6487752 DOI: 10.1177/1744806919838947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Memantine is one of the important clinical medications in treating moderate to severe Alzheimer disease. The effect of memantine on preventing or treating punctate allodynia has been thoroughly studied but not on the induction of dynamic allodynia. The aim of this study is to investigate whether memantine could prevent the induction of dynamic allodynia and its underlying spinal mechanisms. Results (1) In in vivo spared nerve injury pain model, pretreatment with memantine at a lower dose (10 nmol, intrathecal; memantine-10) selectively prevented the induction of dynamic allodynia but not the punctate allodynia. (2) Pretreatment with either MK801-10 (MK801-10 nmol, intrathecal) or higher dose of memantine (30 nmol, intrathecal; memantine-30) prevented the induction of both dynamic and punctate allodynia. (3) Memantine-10 showed significant effect on the inhibition of the spared nerve injury-induced overactivation of microglia in spinal dorsal horn. (4) In contrast, in complete freund′s adjuvant (CFA) model, memantine-10 neither affected the CFA injection-induced activation of microglia in spinal dorsal horn nor the induction of dynamic allodynia. (5) Immunohistological studies showed Kir2.1 channel distributed widely and co-localized with microglia in the spinal dorsal horn of mice. (6) Pretreatment with either minocycline, a microglia inhibitor, or ML133, a Kir2.1 inhibitor, both selectively prevented the overactivation of microglia in spinal dorsal horn and the induction of dynamic allodynia following spared nerve injury. Conclusion The selective inhibitory effect on the induction of dynamic allodynia in spared nerve injury model by low dose of the memantine (memantine-10) was tightly correlated with the blockade of microglia Kir2.1 channel to suppress the microglia activation.
Collapse
Affiliation(s)
- Yangyang Chen
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiqian Shi
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoxiang Wang
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yimei Li
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Longzhen Cheng
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- 1 Neurology Department, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Abstract
The lionfish (Pterois volitans) is a venomous invasive species found in the Caribbean and Northwestern Atlantic. It poses a growing health problem because of the increase in frequency of painful stings, for which no treatment or antidote exists, and the long-term disability caused by the pain. Understanding the venom's algogenic properties can help identify better treatment for these envenomations. In this study, we provide the first characterization of the pain and inflammation caused by lionfish venom and examine the mechanisms through which it causes pain using a combination of in vivo and in vitro approaches including behavioral, physiological, calcium imaging, and electrophysiological testing. Intraplantar injections of the venom produce a significant increase in pain behavior, as well as a marked increase in mechanical sensitivity for up to 24 hours after injection. The algogenic substance(s) are heat-labile peptides that cause neurogenic inflammation at the site of injection and induction of Fos and microglia activation in the superficial layers of the dorsal horn. Finally, calcium imaging and electrophysiology experiments show that the venom acts predominantly on nonpeptidergic, TRPV1-negative, nociceptors, a subset of neurons implicated in sensing mechanical pain. These data provide the first characterization of the pain and inflammation caused by lionfish venom, as well as the first insight into its possible cellular mechanism of action.
Collapse
|