1
|
Chen Y, Liu F, Shi S, Xiao S, Gong X. The Integrated Transcriptome Bioinformatics Analysis of Energy Metabolism-Related Profiles for Dorsal Root Ganglion of Neuropathic Pain. Mol Neurobiol 2025; 62:4149-4171. [PMID: 39406937 DOI: 10.1007/s12035-024-04537-2] [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: 05/28/2024] [Accepted: 10/07/2024] [Indexed: 03/05/2025]
Abstract
Neuropathic pain (NP) is a debilitating disease and is associated with energy metabolism alterations. This study aimed to identify energy metabolism-related differentially expressed genes (EMRDEGs) in NP, construct a diagnostic model, and analyze immune cell infiltration and single-cell gene expression characteristics of NP. GSE89224, GSE123919, and GSE134003 were downloaded from the Gene Expression Omnibus. Differentially expressed genes (DEGs) analysis and an intersection with highly energy metabolism-related modules in weighted gene co-expression network analysis (WGCNA) was performed in GSE89224. Least absolute shrinkage and selection operator (LASSO), random forest, and logistic regression were used for model genes selection. NP samples were divided into high- and low-risk groups and different disease subtypes based on risk score of LASSO algorithm and consensus clustering analysis, respectively. Immune cell composition was estimated in different risk groups and NP subtypes. Datasets 134,003 were performed for identification of single-cell DEGs and functional enrichment. Cell-cell communications and pseudo-time analysis to reveal the expression profile of NP. A total of 38 EMRDEGs were obtained and are majorly enriched in metabolism about glioma and inflammation. LASSO, random forest, and logistic regression identified 6 model genes, which were Itpr1, Gng8, Socs3, Fscn1, Cckbr, and Camk1. The nomogram, based on six model genes, had a good predictive ability, concordance, and diagnostic value. The comparisons between different risk groups and NP subtypes identified important pathways and different immune cells component. The immune infiltration results majorly associated with inflammation and energy metabolism. Single-cell analysis revealed cell-cell communications and cells differentiation characteristics of NP. In conclusion, our results not only elucidate the involvement of energy metabolism in NP but also provides a robust diagnostic tool with six model genes. These findings might give insight into the pathogenesis of NP and provide effective therapeutic regimens for the treatment of NP.
Collapse
Affiliation(s)
- Yongmei Chen
- Department of Laboratory, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, Xiangyang City, Hubei, China
| | - Fan Liu
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shengnan Shi
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shugen Xiao
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Xingrui Gong
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China.
| |
Collapse
|
2
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of Na V 1.7-selective inhibitors for pain. Pain 2025; 166:481-501. [PMID: 39928833 PMCID: PMC11808711 DOI: 10.1097/j.pain.0000000000003425] [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: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 plays an important role in pain processing according to genetic data. Those data made Na V 1.7 a popular drug target, especially since its relatively selective expression in nociceptors promised pain relief without the adverse effects associated with broader sodium channel blockade. Despite encouraging preclinical data in rodents, Na V 1.7-selective inhibitors have not yet proven effective in clinical trials. Discrepancies between preclinical and clinical results should raise alarms. We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors. Putting aside species differences, most preclinical studies tested young male rodents with limited genetic variability, inconsistent with the clinical population. Inflammatory pain was the most common preclinical chronic pain model whereas nearly all clinical trials focused on neuropathic pain despite some evidence suggesting Na V 1.7 channels are not essential for neuropathic pain. Preclinical studies almost exclusively measured evoked pain whereas most clinical trials assessed average pain intensity without distinguishing between evoked and spontaneous pain. Nearly all preclinical studies gave a single dose of drug unlike the repeat dosing used clinically, thus precluding preclinical data from demonstrating whether tolerance or other slow processes occur. In summary, preclinical testing of Na V 1.7-selective inhibitors aligned poorly with clinical testing. Beyond issues that have already garnered widespread attention in the pain literature, our results highlight the treatment regimen and choice of pain model as areas for improvement.
Collapse
Affiliation(s)
- Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Russell Smith
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Ahlström FH, Viisanen H, Karhinen L, Velagapudi V, Blomqvist KJ, Lilius TO, Rauhala PV, Kalso EA. Gene expression in the dorsal root ganglion and the cerebrospinal fluid metabolome in polyneuropathy and opioid tolerance in rats. IBRO Neurosci Rep 2024; 17:38-51. [PMID: 38933596 PMCID: PMC11201153 DOI: 10.1016/j.ibneur.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
First-line pharmacotherapy for peripheral neuropathic pain (NP) of diverse pathophysiology consists of antidepressants and gabapentinoids, but only a minority achieve sufficient analgesia with these drugs. Opioids are considered third-line analgesics in NP due to potential severe and unpredictable adverse effects in long-term use. Also, opioid tolerance and NP may have shared mechanisms, raising further concerns about opioid use in NP. We set out to further elucidate possible shared and separate mechanisms after chronic morphine treatment and oxaliplatin-induced and diabetic polyneuropathies, and to identify potential diagnostic markers and therapeutic targets. We analysed thermal nociceptive behaviour, the transcriptome of dorsal root ganglia (DRG) and the metabolome of cerebrospinal fluid (CSF) in these three conditions, in rats. Several genes were differentially expressed, most following oxaliplatin and least after chronic morphine treatment, compared with saline-treated rats. A few genes were differentially expressed in the DRGs in all three models (e.g. Csf3r and Fkbp5). Some, e.g. Alox15 and Slc12a5, were differentially expressed in both diabetic and oxaliplatin models. Other differentially expressed genes were associated with nociception, inflammation, and glial cells. The CSF metabolome was most significantly affected in the diabetic rats. Interestingly, we saw changes in nicotinamide metabolism, which has been associated with opioid addiction and withdrawal, in the CSF of morphine-tolerant rats. Our results offer new hypotheses for the pathophysiology and treatment of NP and opioid tolerance. In particular, the role of nicotinamide metabolism in opioid addiction deserves further study.
Collapse
Affiliation(s)
- Fredrik H.G. Ahlström
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Hanna Viisanen
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Leena Karhinen
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Vidya Velagapudi
- Metabolomics Unit, Institute for Molecular Medicine Finland FIMM, University of Helsinki, P.O. Box 20, FI-00014, Finland
| | - Kim J. Blomqvist
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Tuomas O. Lilius
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Tukholmankatu 8C, 00014, Finland
- Department of Emergency Medicine and Services, University of Helsinki and HUS Helsinki University Hospital, Haartmaninkatu 4, Helsinki 00290, Finland
| | - Pekka V. Rauhala
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- Individualized Drug Therapy Research Programme, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
| | - Eija A. Kalso
- Department of Pharmacology, Faculty of Medicine, Biomedicum 1, University of Helsinki, Haartmaninkatu 8, 00014, Finland
- SleepWell Research Programme, Faculty of Medicine, , University of Helsinki, Haartmaninkatu 3, 00014, Finland
- Department of Anaesthesiology and Intensive Care Medicine, Helsinki University Hospital and University of Helsinki, HUS, Stenbäckinkatu 9, P.O. Box 440, 00029, Finland
| |
Collapse
|
4
|
Mitchell ME, Torrijos G, Cook LF, Mwirigi JM, He L, Shiers S, Price TJ. Interleukin-6 induces nascent protein synthesis in human dorsal root ganglion nociceptors primarily via MNK-eIF4E signaling. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100159. [PMID: 39156884 PMCID: PMC11327947 DOI: 10.1016/j.ynpai.2024.100159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024]
Abstract
Plasticity of dorsal root ganglion (DRG) nociceptors in the peripheral nervous system requires new protein synthesis. This plasticity is believed to be responsible for the physiological changes seen in DRG nociceptors in animal models of chronic pain. Experiments in human DRG (hDRG) neurons also support this hypothesis, but a direct observation of nascent protein synthesis in response to a pain promoting substance, like interleukin-6 (IL-6), has not been measured in these neurons. To fill this gap in knowledge, we used acutely prepared human DRG explants from organ donors. These explants provide a physiologically relevant microenvironment, closer to in vivo conditions, allowing for the examination of functional alterations in DRG neurons reflective of human neuropathophysiology. Using this newly developed assay, we demonstrate upregulation of the target of the MNK1/2 kinases, phosphorylated eIF4E (p-eIF4E), and nascently synthesized proteins in a substantial subset of hDRG neurons following exposure to IL-6. To pinpoint the specific molecular mechanisms driving this IL-6-driven increase in nascent proteins, we used the specific MNK1/2 inhibitor eFT508. Treatment with eFT508 resulted in the inhibition of IL-6-induced increases in p-eIF4E and nascent proteins. Additionally, using TRPV1 as a marker for nociceptors, we found that these effects occurred in a large number of human nociceptors. Our findings provide clear evidence that IL-6 drives nascent protein synthesis in human TRPV1+ nociceptors primarily via MNK1/2-eIF4E signaling. The work links animal findings to human nociception, creates a framework for additional hDRG signaling experiments, and substantiates the continued development of MNK inhibitors for pain.
Collapse
Affiliation(s)
| | | | - Lauren F. Cook
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Juliet M. Mwirigi
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Lucy He
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Theodore J. Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
5
|
Lister KC, Wong C, Uttam S, Parisien M, Stecum P, Brown N, Cai W, Hooshmandi M, Gu N, Amiri M, Beaudry F, Jafarnejad SM, Tavares-Ferreira D, Inturi NN, Mazhar K, Zhao HT, Fitzsimmons B, Gkogkas CG, Sonenberg N, Price TJ, Diatchenko L, Atlasi Y, Mogil JS, Khoutorsky A. Translational control in the spinal cord regulates gene expression and pain hypersensitivity in the chronic phase of neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600539. [PMID: 38979173 PMCID: PMC11230214 DOI: 10.1101/2024.06.24.600539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensitization of spinal nociceptive circuits plays a crucial role in neuropathic pain. This sensitization depends on new gene expression that is primarily regulated via transcriptional and translational control mechanisms. The relative roles of these mechanisms in regulating gene expression in the clinically relevant chronic phase of neuropathic pain are not well understood. Here, we show that changes in gene expression in the spinal cord during the chronic phase of neuropathic pain are substantially regulated at the translational level. Downregulating spinal translation at the chronic phase alleviated pain hypersensitivity. Cell-type-specific profiling revealed that spinal inhibitory neurons exhibited greater changes in translation after peripheral nerve injury compared to excitatory neurons. Notably, increasing translation selectively in all inhibitory neurons or parvalbumin-positive (PV+) interneurons, but not excitatory neurons, promoted mechanical pain hypersensitivity. Furthermore, increasing translation in PV+ neurons decreased their intrinsic excitability and spiking activity, whereas reducing translation in spinal PV+ neurons prevented the nerve injury-induced decrease in excitability. Thus, translational control mechanisms in the spinal cord, particularly in inhibitory neurons, play a role in mediating neuropathic pain hypersensitivity.
Collapse
Affiliation(s)
- Kevin C. Lister
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Marc Parisien
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Patricia Stecum
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Nicole Brown
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Weihua Cai
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Mehdi Hooshmandi
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Ning Gu
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Mehdi Amiri
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Francis Beaudry
- Département de biomédecine vétérinaire, Faculté de médecine vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- Centre de recherche sur le cerveau et l’apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, BT9 7AE, UK
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Nikhil Nageshwar Inturi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Khadijah Mazhar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | | | | | - Christos G. Gkogkas
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, University Campus, 45110 Ioannina, Greece
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Dallas, 75080
| | - Luda Diatchenko
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, BT9 7AE, UK
| | - Jeffrey S. Mogil
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
- Department of Psychology, Faculty of Science, McGill University, Montreal, QC, Canada
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Rouchka EC, de Almeida C, House RB, Daneshmand JC, Chariker JH, Saraswat-Ohri S, Gomes C, Sharp M, Shum-Siu A, Cesarz GM, Petruska JC, Magnuson DSK. Construction of a Searchable Database for Gene Expression Changes in Spinal Cord Injury Experiments. J Neurotrauma 2024; 41:1030-1043. [PMID: 37917105 PMCID: PMC11302316 DOI: 10.1089/neu.2023.0035] [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] [Indexed: 11/03/2023] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition with an estimated 18,000 new cases annually in the United States. The field has accepted and adopted standardized databases such as the Open Data Commons for Spinal Cord Injury (ODC-SCI) to aid in broader analyses, but these currently lack high-throughput data despite the availability of nearly 6000 samples from over 90 studies available in the Sequence Read Archive. This limits the potential for large datasets to enhance our understanding of SCI-related mechanisms at the molecular and cellular level. Therefore, we have developed a protocol for processing RNA-Seq samples from high-throughput sequencing experiments related to SCI resulting in both raw and normalized data that can be efficiently mined for comparisons across studies, as well as homologous discovery across species. We have processed 1196 publicly available RNA-Seq samples from 50 bulk RNA-Seq studies across nine different species, resulting in an SQLite database that can be used by the SCI research community for further discovery. We provide both the database as well as a web-based front-end that can be used to query the database for genes of interest, differential gene expression, genes with high variance, and gene set enrichments.
Collapse
Affiliation(s)
- Eric C. Rouchka
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
- Kentucky IDeA Networks of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
- Bioinformatics Program, University of Louisville, Louisville, Kentucky, USA
| | - Carlos de Almeida
- Translational Neuroscience Program, University of Louisville, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Randi B. House
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, USA
| | | | - Julia H. Chariker
- Kentucky IDeA Networks of Biomedical Research Excellence (KY INBRE) Bioinformatics Core, University of Louisville, Louisville, Kentucky, USA
- Department of Neuroscience Training, University of Louisville, Louisville, Kentucky, USA
| | - Sujata Saraswat-Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Cynthia Gomes
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| | - Morgan Sharp
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Alice Shum-Siu
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Greta M. Cesarz
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Jeffrey C. Petruska
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| | - David S. K. Magnuson
- Translational Neuroscience Program, University of Louisville, Louisville, Kentucky, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
- Department of Neurological Surgery, University of Louisville, Louisville, Kentucky, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Barakat A, Munro G, Heegaard AM. Finding new analgesics: Computational pharmacology faces drug discovery challenges. Biochem Pharmacol 2024; 222:116091. [PMID: 38412924 DOI: 10.1016/j.bcp.2024.116091] [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: 10/02/2023] [Revised: 01/10/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
Despite the worldwide prevalence and huge burden of pain, pain is an undertreated phenomenon. Currently used analgesics have several limitations regarding their efficacy and safety. The discovery of analgesics possessing a novel mechanism of action has faced multiple challenges, including a limited understanding of biological processes underpinning pain and analgesia and poor animal-to-human translation. Computational pharmacology is currently employed to face these challenges. In this review, we discuss the theory, methods, and applications of computational pharmacology in pain research. Computational pharmacology encompasses a wide variety of theoretical concepts and practical methodological approaches, with the overall aim of gaining biological insight through data acquisition and analysis. Data are acquired from patients or animal models with pain or analgesic treatment, at different levels of biological organization (molecular, cellular, physiological, and behavioral). Distinct methodological algorithms can then be used to analyze and integrate data. This helps to facilitate the identification of biological molecules and processes associated with pain phenotype, build quantitative models of pain signaling, and extract translatable features between humans and animals. However, computational pharmacology has several limitations, and its predictions can provide false positive and negative findings. Therefore, computational predictions are required to be validated experimentally before drawing solid conclusions. In this review, we discuss several case study examples of combining and integrating computational tools with experimental pain research tools to meet drug discovery challenges.
Collapse
Affiliation(s)
- Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt.
| | | | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Zhang J, Zhang X, Gao Y, Li L, Bai L, Wang L, Qiao Y, Wang X, Liang Z, Xu JT. Neuralized1-Mediated CPEB3 Ubiquitination in the Spinal Dorsal Horn Contributes to the Pathogenesis of Neuropathic Pain in Rats. ACS Chem Neurosci 2023; 14:3418-3430. [PMID: 37644621 DOI: 10.1021/acschemneuro.3c00313] [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] [Indexed: 08/31/2023] Open
Abstract
Compelling evidence has shown that Neuralized1 (Neurl1) facilitates hippocampal-dependent memory storage by modulating cytoplasmic polyadenylation element-binding protein 3 (CPEB3)-dependent protein synthesis. In the current study, we investigated the role of Neurl1 in the pathogenesis of neuropathic pain and the underlying mechanisms. The neuropathic pain was evaluated by lumbar 5 spinal nerve ligation (SNL) in rats. Immunofluorescence staining, Western blotting, qRT-PCR, and coimmunoprecipitation (Co-IP) were performed to investigate the underlying mechanisms. Our results showed that SNL led to an increase of Neurl1 in the spinal dorsal horn. Spinal microinjection of AAV-EGFP-Neurl1 shRNA alleviated mechanical allodynia; decreased the level of CPEB3 ubiquitination; inhibited the production of GluA1, GluA2, and PSD95; and reduced GluA1-containing AMPA receptors in the membrane of the dorsal horn following SNL. Knockdown of spinal CPEB3 decreased the production of GluA1, GluA2, and PSD95 in the dorsal horn and attenuated abnormal pain after SNL. Overexpression of Neurl1 in the dorsal horn resulted in pain-related hypersensitivity in naïve rats; raised the level of CPEB3 ubiquitination; increased the production of GluA1, GluA2, and PSD95; and augmented GluA1-containing AMPA receptors in the membrane in the dorsal horn. Moreover, spinal Neurl1 overexpression-induced mechanical allodynia in naïve rats was partially reversed by repeated intrathecal injections of CPEB3 siRNA. Collectively, our results suggest that SNL-induced upregulation of Neurl1 through CPEB3 ubiquitination-dependent production of GluA1, GluA2, and PSD95 in the dorsal horn contributes to the pathogenesis of neuropathic pain in rats. Targeting spinal Neurl1 might be a promising therapeutic strategy for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yan Gao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liren Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Liying Bai
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital, Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Li Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Yiming Qiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Xueli Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Zongyi Liang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ji-Tian Xu
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
- Neuroscience Research Institute, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| |
Collapse
|
9
|
Borgonetti V, Galeotti N. Posttranscriptional Regulation of Gene Expression Participates in the Myelin Restoration in Mouse Models of Multiple Sclerosis: Antisense Modulation of HuR and HuD ELAV RNA Binding Protein. Mol Neurobiol 2023; 60:2661-2677. [PMID: 36696009 PMCID: PMC10039839 DOI: 10.1007/s12035-023-03236-8] [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: 07/26/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023]
Abstract
Neuropathic pain is the most difficult-to-treat pain syndrome in multiple sclerosis. Evidence relates neuropathic pain to demyelination, which often originates from unresolved neuroinflammation or altered immune response. Posttranscriptional regulation of gene expression might play a fundamental role in the regulation of these processes. The ELAV RNA-binding proteins HuR and HuD are involved in the promotion of inflammatory phenomena and in neuronal development and maintenance, respectively. Thus, the aim of this study was to investigate the role of HuR and HuD in demyelination-associated neuropathic pain in the mouse experimental autoimmune encephalomyelitis (EAE) model. HuR resulted overexpressed in the spinal cord of MOG35-55-EAE and PLP139-151-EAE mice and was detected in CD11b + cells. Conversely, HuD was largely downregulated in the MOG-EAE spinal cord, along with GAP43 and neurofilament H, while in PLP-EAE mice, HuD and neuronal markers remained unaltered. Intranasal antisense oligonucleotide (ASO) delivery to knockdown HuR, increased myelin basic protein expression, and Luxol Fast Blue staining in both EAE models, an indication of increased myelin content. These effects temporally coincided with attenuation of pain hypersensitivity. Anti-HuR ASO increased the expression of HuD in GAP43-expressing cells and promoted a HuD-mediated neuroprotective activity in MOG-EAE mice, while in PLP-EAE mice, HuR silencing dampened pro-inflammatory responses mediated by spinal microglia activation. In conclusion, anti-HuR ASO showed myelin protection at analgesic doses with multitarget mechanisms, and it deserves further consideration as an innovative agent to counteract demyelination in neuropathic pain states.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, I-50139, Florence, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, I-50139, Florence, Italy.
| |
Collapse
|
10
|
Lückemeyer DD, Prudente AS, de Amorim Ferreira M, da Silva AM, Tonello R, Junior SJM, do Prado CSH, de Castro Júnior CJ, Gomez MV, Calixto JB, Ferreira J. Critical Pronociceptive Role of Family 2 Voltage-Gated Calcium Channels in a Novel Mouse Model of HIV-Associated Sensory Neuropathy. Mol Neurobiol 2023; 60:2954-2968. [PMID: 36754911 DOI: 10.1007/s12035-023-03244-8] [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: 07/11/2022] [Accepted: 01/13/2023] [Indexed: 02/10/2023]
Abstract
Some people living with HIV present painful sensory neuropathy (HIV-SN) that is pharmacoresistant, sex-associated, and a major source of morbidity. Since the specific mechanisms underlying HIV-SN are not well understood, the aim of our study was to characterize a novel model of painful HIV-SN by combining the HIV-1 gp120 protein and the antiretroviral stavudine (d4T) in mice and to investigate the pronociceptive role of the family 2 voltage-gated calcium channel (VGCC) α1 subunit (Cav2.X channels) in such a model. HIV-SN was induced in male and female C57BL/6 mice by administration of gp120 and/or d4T and detected by a battery of behavior tests and by immunohistochemistry. The role of Cav2.X channels was assessed by the treatment with selective blockers and agonists as well as by mRNA detection. Repeated administration with gp120 and/or d4T produced long-lasting touch-evoked painful-like behaviors (starting at 6 days, reaching a maximum on day 13, and lasting up to 28 days after treatment started), with a greater intensity in female mice treated with the combination of gp120 + d4T. Moreover, gp120 + d4T treatment reduced the intraepidermal nerve fibers and well-being of female mice, without altering other behaviors. Mechanistically, gp120 + d4T treatment induced Cav2.1, 2.2, and 2.3 transcriptional increases in the dorsal root ganglion and the Cav2.X agonist-induced nociception. Accordingly, intrathecal selective Cav2.2 blockade presented longer and better efficacy in reversing the hyperalgesia induced by gp120 + d4T treatment compared with Cav2.1 or Cav2.3, but also presented the worst safety (inducing side effects at effective doses). We conclude that the family 2 calcium channels (Cav2.X) exert a critical pronociceptive role in a novel mouse model of HIV-SN.
Collapse
Affiliation(s)
- Debora Denardin Lückemeyer
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Arthur Silveira Prudente
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Marcella de Amorim Ferreira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Ana Merian da Silva
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Raquel Tonello
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sérgio José Macedo Junior
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
- Centro de Inovação E Ensaios Pré-Clínicos, Florianópolis, SC, Brazil
| | | | | | - Marcus Vinicius Gomez
- Instituto de Educação E Pesquisa da Santa Casa de Belo Horizonte, Belo Horizonte, MG, Brazil
| | | | - Juliano Ferreira
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| |
Collapse
|
11
|
Tu Y, Muley MM, Beggs S, Salter MW. Microglia-independent peripheral neuropathic pain in male and female mice. Pain 2022; 163:e1129-e1144. [PMID: 35384869 PMCID: PMC9578531 DOI: 10.1097/j.pain.0000000000002643] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT The dominant view in the field of pain is that peripheral neuropathic pain is driven by microglia in the somatosensory processing region of the spinal dorsal horn. Here, to the contrary, we discovered a form of neuropathic pain that is independent of microglia. Mice in which the nucleus pulposus (NP) of the intervertebral disc was apposed to the sciatic nerve developed a constellation of neuropathic pain behaviours: hypersensitivity to mechanical, cold, and heat stimuli. However, NP application caused no activation of spinal microglia nor was pain hypersensitivity reversed by microglial inhibition. Rather, NP-induced pain hypersensitivity was dependent on cells within the NP which recruited macrophages to the adjacent nerve. Eliminating macrophages systemically or locally prevented NP-induced pain hypersensitivity. Pain hypersensitivity was also prevented by genetically disrupting the neurotrophin brain-derived neurotrophic factor selectively in macrophages. Moreover, the behavioural phenotypes as well as the molecular mechanisms of NP-induced pain hypersensitivity were not different between males and females. Our findings reveal a previously unappreciated mechanism for by which a discrete peripheral nerve lesion may produce pain hypersensitivity, which may help to explain the limited success of microglial inhibitors on neuropathic pain in human clinical trials.
Collapse
Affiliation(s)
- YuShan Tu
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Milind M. Muley
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto Centre for the Study of Pain, Toronto, ON, Canada
| | - Simon Beggs
- Developmental Neurosciences, UCL GOSH Institute of Child Health, London, United Kingdom
| | - Michael W. Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada
- University of Toronto Centre for the Study of Pain, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Zheng Q, Dong X, Green DP, Dong X. Peripheral mechanisms of chronic pain. MEDICAL REVIEW 2022; 2:251-270. [PMID: 36067122 PMCID: PMC9381002 DOI: 10.1515/mr-2022-0013] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Abstract
Acutely, pain serves to protect us from potentially harmful stimuli, however damage to the somatosensory system can cause maladaptive changes in neurons leading to chronic pain. Although acute pain is fairly well controlled, chronic pain remains difficult to treat. Chronic pain is primarily a neuropathic condition, but studies examining the mechanisms underlying chronic pain are now looking beyond afferent nerve lesions and exploring new receptor targets, immune cells, and the role of the autonomic nervous system in contributing chronic pain conditions. The studies outlined in this review reveal how chronic pain is not only confined to alterations in the nervous system and presents findings on new treatment targets and for this debilitating disease.
Collapse
Affiliation(s)
- Qin Zheng
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Dustin P. Green
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Howard Hughes Medical Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
13
|
Paracha M, Thakar A, Darling RA, Wulff SS, Rule DC, Nair S, Brown TE. Role of cathepsin K in the expression of mechanical hypersensitivity following intra-plantar inflammation. Sci Rep 2022; 12:7108. [PMID: 35501334 PMCID: PMC9061763 DOI: 10.1038/s41598-022-11043-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/14/2022] [Indexed: 11/26/2022] Open
Abstract
Persistent/chronic inflammatory pain involves multiple pathophysiological mechanisms and is far more complex than acute/momentary pain. Current therapeutics for chronic inflammatory pain are often not effective because the etiology responsible for the pain is not addressed by traditional pharmacological treatments. Cathepsin K is a cysteine protease that has mostly been studied in the context of bone and joint disorders. Previous work by others has shown that inhibition of cathepsin K activity reduces osteoarthritis-associated nociception in joints. However, the role of cathepsin K in cutaneous inflammation is understudied. We assessed the effectiveness of genetic deletion or pharmacological inhibition of cathepsin K in male mice on the expression of nocifensive behaviors after formalin injection or mechanical and thermal hypersensitivity after injection of complete Freund’s adjuvant (CFA) into the mouse hind paw. Our data demonstrate that cathepsin K knockout mice (Ctsk−/−) have a reduction in nocifensive behaviors in the formalin test. In addition, Ctsk−/− do not develop mechanical hypersensitivity after CFA injection for up to 7 days. Moreover, we found that inhibition of cathepsin K reduced mechanical hypersensitivity after CFA injection and mRNA levels, protein levels, and cathepsin K activity levels were elevated after CFA injection. Based upon our data, cathepsin K is indicated to play a role in the expression of chemically-induced cutaneous hypersensitivity, as Ctsk−/− mice do not develop mechanical hypersensitivity and show a reduction in nocifensive behaviors. Further research is needed to determine whether attenuating cathepsin K activity may generate a clinically relevant therapeutic.
Collapse
|
14
|
Zheng Q, Xie W, Lückemeyer DD, Lay M, Wang XW, Dong X, Limjunyawong N, Ye Y, Zhou FQ, Strong JA, Zhang JM, Dong X. Synchronized cluster firing, a distinct form of sensory neuron activation, drives spontaneous pain. Neuron 2022; 110:209-220.e6. [PMID: 34752775 PMCID: PMC8776619 DOI: 10.1016/j.neuron.2021.10.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/01/2021] [Accepted: 10/13/2021] [Indexed: 01/21/2023]
Abstract
Spontaneous pain refers to pain occurring without external stimuli. It is a primary complaint in chronic pain conditions and remains difficult to treat. Moreover, the mechanisms underlying spontaneous pain remain poorly understood. Here we employed in vivo imaging of dorsal root ganglion (DRG) neurons and discovered a distinct form of abnormal spontaneous activity following peripheral nerve injury: clusters of adjacent DRG neurons firing synchronously and sporadically. The level of cluster firing correlated directly with nerve injury-induced spontaneous pain behaviors. Furthermore, we demonstrated that cluster firing is triggered by activity of sympathetic nerves, which sprout into DRGs after injury, and identified norepinephrine as a key neurotransmitter mediating this unique firing. Chemogenetic and pharmacological manipulations of sympathetic activity and norepinephrine receptors suggest that they are necessary and sufficient for DRG cluster firing and spontaneous pain behavior. Therefore, blocking sympathetically mediated cluster firing may be a new paradigm for treating spontaneous pain.
Collapse
Affiliation(s)
- Qin Zheng
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Debora D Lückemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mark Lay
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Yaqing Ye
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA.
| |
Collapse
|
15
|
Wistrom E, Chase R, Smith PR, Campbell ZT. A compendium of validated pain genes. WIREs Mech Dis 2022; 14:e1570. [PMID: 35760453 PMCID: PMC9787016 DOI: 10.1002/wsbm.1570] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022]
Abstract
The development of novel pain therapeutics hinges on the identification and rigorous validation of potential targets. Model organisms provide a means to test the involvement of specific genes and regulatory elements in pain. Here we provide a list of genes linked to pain-associated behaviors. We capitalize on results spanning over three decades to identify a set of 242 genes. They support a remarkable diversity of functions spanning action potential propagation, immune response, GPCR signaling, enzymatic catalysis, nucleic acid regulation, and intercellular signaling. Making use of existing tissue and single-cell high-throughput RNA sequencing datasets, we examine their patterns of expression. For each gene class, we discuss archetypal members, with an emphasis on opportunities for additional experimentation. Finally, we discuss how powerful and increasingly ubiquitous forward genetic screening approaches could be used to improve our ability to identify pain genes. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Eric Wistrom
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Rebecca Chase
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Patrick R. Smith
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA
| | - Zachary T. Campbell
- Department of Biological SciencesUniversity of Texas at DallasRichardsonTexasUSA,Center for Advanced Pain StudiesUniversity of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
16
|
Molecular Changes in the Dorsal Root Ganglion during the Late Phase of Peripheral Nerve Injury-induced Pain in Rodents: A Systematic Review. Anesthesiology 2021; 136:362-388. [PMID: 34965284 DOI: 10.1097/aln.0000000000004092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
The dorsal root ganglion is widely recognized as a potential target to treat chronic pain. A fundamental understanding of quantitative molecular and genomic changes during the late phase of pain is therefore indispensable. The authors performed a systematic literature review on injury-induced pain in rodent dorsal root ganglions at minimally 3 weeks after injury. So far, slightly more than 300 molecules were quantified on the protein or messenger RNA level, of which about 60 were in more than one study. Only nine individual sequencing studies were performed in which the most up- or downregulated genes varied due to heterogeneity in study design. Neuropeptide Y and galanin were found to be consistently upregulated on both the gene and protein levels. The current knowledge regarding molecular changes in the dorsal root ganglion during the late phase of pain is limited. General conclusions are difficult to draw, making it hard to select specific molecules as a focus for treatment.
Collapse
|
17
|
Chen D, Yu W, Aitken L, Gunn-Moore F. Willin/FRMD6: A Multi-Functional Neuronal Protein Associated with Alzheimer's Disease. Cells 2021; 10:cells10113024. [PMID: 34831245 PMCID: PMC8616527 DOI: 10.3390/cells10113024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
The FERM domain-containing protein 6 (FRMD6), also known as Willin, is an upstream regulator of Hippo signaling that has recently been shown to modulate actin cytoskeleton dynamics and mechanical phenotype of neuronal cells through ERK signaling. Physiological functions of Willin/FRMD6 in the nervous system include neuronal differentiation, myelination, nerve injury repair, and vesicle exocytosis. The newly established neuronal role of Willin/FRMD6 is of particular interest given the mounting evidence suggesting a role for Willin/FRMD6 in Alzheimer's disease (AD), including a series of genome wide association studies that position Willin/FRMD6 as a novel AD risk gene. Here we describe recent findings regarding the role of Willin/FRMD6 in the nervous system and its actions in cellular perturbations related to the pathogenesis of AD.
Collapse
|
18
|
Keane L, Antignano I, Riechers SP, Zollinger R, Dumas AA, Offermann N, Bernis ME, Russ J, Graelmann F, McCormick PN, Esser J, Tejera D, Nagano A, Wang J, Chelala C, Biederbick Y, Halle A, Salomoni P, Heneka MT, Capasso M. mTOR-dependent translation amplifies microglia priming in aging mice. J Clin Invest 2021; 131:132727. [PMID: 33108356 DOI: 10.1172/jci132727] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/14/2020] [Indexed: 12/20/2022] Open
Abstract
Microglia maintain homeostasis in the brain. However, with age, they become primed and respond more strongly to inflammatory stimuli. We show here that microglia from aged mice had upregulated mTOR complex 1 signaling controlling translation, as well as protein levels of inflammatory mediators. Genetic ablation of mTOR signaling showed a dual yet contrasting effect on microglia priming: it caused an NF-κB-dependent upregulation of priming genes at the mRNA level; however, mice displayed reduced cytokine protein levels, diminished microglia activation, and milder sickness behavior. The effect on translation was dependent on reduced phosphorylation of 4EBP1, resulting in decreased binding of eIF4E to eIF4G. Similar changes were present in aged human microglia and in damage-associated microglia, indicating that upregulation of mTOR-dependent translation is an essential aspect of microglia priming in aging and neurodegeneration.
Collapse
Affiliation(s)
- Lily Keane
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Centre for Tumour Microenvironment and
| | | | | | | | | | - Nina Offermann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Maria E Bernis
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jenny Russ
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | | | - Julia Esser
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dario Tejera
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ai Nagano
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Claude Chelala
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | - Annett Halle
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Paolo Salomoni
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Melania Capasso
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Centre for Tumour Microenvironment and
| |
Collapse
|
19
|
Tran PV, Johns ME, McAdams B, Abrahante JE, Simone DA, Banik RK. Global transcriptome analysis of rat dorsal root ganglia to identify molecular pathways involved in incisional pain. Mol Pain 2021; 16:1744806920956480. [PMID: 32909881 PMCID: PMC7493244 DOI: 10.1177/1744806920956480] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
To develop non-opioid therapies for postoperative incisional pain, we must understand its underlying molecular mechanisms. In this study, we assessed global gene expression changes in dorsal root ganglia neurons in a model of incisional pain to identify pertinent molecular pathways. Male, Sprague-Dawley rats underwent infiltration of 1% capsaicin or vehicle into the plantar hind paw (n = 6-9/group) 30 min before plantar incision. Twenty-four hours after incision or sham (control) surgery, lumbar L4-L6 dorsal root ganglias were collected from rats pretreated with vehicle or capsaicin. RNA was isolated and sequenced by next generation sequencing. The genes were then annotated to functional networks using a knowledge-based database, Ingenuity Pathway Analysis. In rats pretreated with vehicle, plantar incision caused robust hyperalgesia, up-regulated 36 genes and downregulated 90 genes in dorsal root ganglias one day after plantar incision. Capsaicin pretreatment attenuated pain behaviors, caused localized denervation of the dermis and epidermis, and prevented the incision-induced changes in 99 of 126 genes. The pathway analyses showed altered gene networks related to increased pro-inflammatory and decreased anti-inflammatory responses in dorsal root ganglias. Insulin-like growth factor signaling was identified as one of the major gene networks involved in the development of incisional pain. Expression of insulin-like growth factor -2 and IGFBP6 in dorsal root ganglia were independently validated with quantitative real-time polymerase chain reaction. We discovered a distinct subset of dorsal root ganglia genes and three key signaling pathways that are altered 24 h after plantar incision but are unchanged when incision was made after capsaicin infiltration in the skin. Further exploration of molecular mechanisms of incisional pain may yield novel therapeutic targets.
Collapse
Affiliation(s)
- Phu V Tran
- Department of Pediatrics, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Malcolm E Johns
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Brian McAdams
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Juan E Abrahante
- Informatics Institute, University of Minnesota, Minneapolis, MN, USA
| | - Donald A Simone
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Ratan K Banik
- Department of Anesthesiology, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
Wang SM, Goguadze N, Kimura Y, Yasui Y, Pan B, Wang TY, Nakamura Y, Lin YT, Hogan QH, Wilson KL, Su TP, Wu HE. Genomic Action of Sigma-1 Receptor Chaperone Relates to Neuropathic Pain. Mol Neurobiol 2021; 58:2523-2541. [PMID: 33459966 PMCID: PMC8128747 DOI: 10.1007/s12035-020-02276-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022]
Abstract
Sigma-1 receptors (Sig-1Rs) are endoplasmic reticulum (ER) chaperones implicated in neuropathic pain. Here we examine if the Sig-1R may relate to neuropathic pain at the level of dorsal root ganglia (DRG). We focus on the neuronal excitability of DRG in a "spare nerve injury" (SNI) model of neuropathic pain in rats and find that Sig-1Rs likely contribute to the genesis of DRG neuronal excitability by decreasing the protein level of voltage-gated Cav2.2 as a translational inhibitor of mRNA. Specifically, during SNI, Sig-1Rs translocate from ER to the nuclear envelope via a trafficking protein Sec61β. At the nucleus, the Sig-1R interacts with cFos and binds to the promoter of 4E-BP1, leading to an upregulation of 4E-BP1 that binds and prevents eIF4E from initiating the mRNA translation for Cav2.2. Interestingly, in Sig-1R knockout HEK cells, Cav2.2 is upregulated. In accordance with those findings, we find that intra-DRG injection of Sig-1R agonist (+)pentazocine increases frequency of action potentials via regulation of voltage-gated Ca2+ channels. Conversely, intra-DRG injection of Sig-1R antagonist BD1047 attenuates neuropathic pain. Hence, we discover that the Sig-1R chaperone causes neuropathic pain indirectly as a translational inhibitor.
Collapse
MESH Headings
- Animals
- Calcium Channels, N-Type/genetics
- Calcium Channels, N-Type/metabolism
- Endoplasmic Reticulum/metabolism
- Eukaryotic Initiation Factor-4E/metabolism
- Ganglia, Spinal/metabolism
- Gene Expression Regulation
- Genome
- HEK293 Cells
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Nerve Tissue/injuries
- Nerve Tissue/pathology
- Neuralgia/genetics
- Nuclear Envelope/metabolism
- Promoter Regions, Genetic/genetics
- Protein Biosynthesis
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Caps/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptors, sigma/agonists
- Receptors, sigma/genetics
- Receptors, sigma/metabolism
- SEC Translocation Channels/metabolism
- Transcription, Genetic
- Sigma-1 Receptor
- Rats
Collapse
Affiliation(s)
- Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Tzu-Yun Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Psychiatry, College of Medicine, National Cheng Kung University, Tainan City, 70101, Taiwan
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
- Department of Pharmacology, Graduate School of Biomedical & Health Science, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Yu-Ting Lin
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH/DHHS, Suite 3512, 333 Cassell Drive, Baltimore, MD, 21224, USA
| |
Collapse
|
21
|
Barragan-Iglesias P, Kunder N, Wanghzou A, Black B, Ray PR, Lou TF, de la Peña JB, Atmaramani R, Shukla T, Pancrazio JJ, Price TJ, Campbell ZT. A peptide encoded within a 5' untranslated region promotes pain sensitization in mice. Pain 2021; 162:1864-1875. [PMID: 33449506 PMCID: PMC8119312 DOI: 10.1097/j.pain.0000000000002191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022]
Abstract
ABSTRACT Translational regulation permeates neuronal function. Nociceptors are sensory neurons responsible for the detection of harmful stimuli. Changes in their activity, termed plasticity, are intimately linked to the persistence of pain. Although inhibitors of protein synthesis robustly attenuate pain-associated behavior, the underlying targets that support plasticity are largely unknown. Here, we examine the contribution of protein synthesis in regions of RNA annotated as noncoding. Based on analyses of previously reported ribosome profiling data, we provide evidence for widespread translation in noncoding transcripts and regulatory regions of mRNAs. We identify an increase in ribosome occupancy in the 5' untranslated regions of the calcitonin gene-related peptide (CGRP/Calca). We validate the existence of an upstream open reading frame (uORF) using a series of reporter assays. Fusion of the uORF to a luciferase reporter revealed active translation in dorsal root ganglion neurons after nucleofection. Injection of the peptide corresponding to the calcitonin gene-related peptide-encoded uORF resulted in pain-associated behavioral responses in vivo and nociceptor sensitization in vitro. An inhibitor of heterotrimeric G protein signaling blocks both effects. Collectively, the data suggest pervasive translation in regions of the transcriptome annotated as noncoding in dorsal root ganglion neurons and identify a specific uORF-encoded peptide that promotes pain sensitization through GPCR signaling.
Collapse
Affiliation(s)
- Paulino Barragan-Iglesias
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
- Department of Physiology and Pharmacology, Center for Basic
Sciences, Autonomous University of Aguascalientes, Aguascalientes, 20130,
Mexico
| | - Nikesh Kunder
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Andi Wanghzou
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
| | - Bryan Black
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
| | - Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
| | - Tzu-Fang Lou
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Rahul Atmaramani
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
| | - Tarjani Shukla
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Joseph J. Pancrazio
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| | - Zachary T. Campbell
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
22
|
Belair DG, Sudak K, Connelly K, Collins ND, Kopytek SJ, Kolaja KL. Investigation Into the Role of ERK in Tyrosine Kinase Inhibitor-Induced Neuropathy. Toxicol Sci 2021; 181:160-174. [PMID: 33749749 DOI: 10.1093/toxsci/kfab033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common and debilitating adverse event that can alter patient treatment options and halt candidate drug development. A case study is presented here describing the preclinical and clinical development of CC-90003, a small molecule extracellular signal-regulated kinase (ERK)1/2 inhibitor investigated as an oncology therapy. In a Phase Ia clinical trial, CC-90003 elicited adverse drug-related neuropathy and neurotoxicity that contributed to discontinued development of CC-90003 for oncology therapy. Preclinical evaluation of CC-90003 in dogs revealed clinical signs and electrophysiological changes consistent with peripheral neuropathy that was reversible. Mice did not exhibit signs of neuropathy upon daily dosing with CC-90003, supporting that rodents generally poorly predict CIPN. We sought to investigate the mechanism of CC-90003-induced peripheral neuropathy using a phenotypic in vitro assay. Translating preclinical neuropathy findings to humans proves challenging as no robust in vitro models of CIPN exist. An approach was taken to examine the influence of CIPN-associated drugs on human-induced pluripotent stem cell-derived peripheral neuron (hiPSC-PN) electrophysiology on multielectrode arrays (MEAs). The MEA assay with hiPSC-PNs was sensitive to CIPN-associated drugs cisplatin, sunitinib, colchicine, and importantly, to CC-90003 in concordance with clinical neuropathy incidence. Biochemical data together with in vitro MEA data for CC-90003 and 12 of its structural analogs, all having similar ERK inhibitory activity, revealed that CC-90003 disrupted in vitro neuronal electrophysiology likely via on-target ERK inhibition combined with off-target kinase inhibition and translocator protein inhibition. This approach could prove useful for assessing CIPN risk and interrogating mechanisms of drug-induced neuropathy.
Collapse
Affiliation(s)
- David G Belair
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| | - Katelyn Sudak
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| | - Kimberly Connelly
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| | - Nathaniel D Collins
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| | - Stephan J Kopytek
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| | - Kyle L Kolaja
- Nonclinical Safety, Bristol Myers Squibb (formerly Celgene), Summit, New Jersey 07901, USA
| |
Collapse
|
23
|
Niehaus JK, Taylor-Blake B, Loo L, Simon JM, Zylka MJ. Spinal macrophages resolve nociceptive hypersensitivity after peripheral injury. Neuron 2021; 109:1274-1282.e6. [PMID: 33667343 DOI: 10.1016/j.neuron.2021.02.018] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/28/2020] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Peripheral nerve injury induces long-term pro-inflammatory responses in spinal cord glial cells that facilitate neuropathic pain, but the identity of endogenous cells that resolve spinal inflammation has not been determined. Guided by single-cell RNA sequencing (scRNA-seq), we found that MRC1+ spinal cord macrophages proliferated and upregulated the anti-inflammatory mediator Cd163 in mice following superficial injury (SI; nerve intact), but this response was blunted in nerve-injured animals. Depleting spinal macrophages in SI animals promoted microgliosis and caused mechanical hypersensitivity to persist. Conversely, expressing Cd163 in spinal macrophages increased Interleukin 10 expression, attenuated micro- and astrogliosis, and enduringly alleviated mechanical and thermal hypersensitivity in nerve-injured animals. Our data indicate that MRC1+ spinal macrophages actively restrain glia to limit neuroinflammation and resolve mechanical pain following a superficial injury. Moreover, we show that spinal macrophages from nerve-injured animals mount a dampened anti-inflammatory response but can be therapeutically coaxed to promote long-lasting recovery of neuropathic pain.
Collapse
Affiliation(s)
- Jesse K Niehaus
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bonnie Taylor-Blake
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lipin Loo
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
24
|
Notartomaso S, Scarselli P, Mascio G, Liberatore F, Mazzon E, Mammana S, Gugliandolo A, Cruccu G, Bruno V, Nicoletti F, Battaglia G. N-Acetylcysteine causes analgesia in a mouse model of painful diabetic neuropathy. Mol Pain 2021; 16:1744806920904292. [PMID: 32009537 PMCID: PMC6997966 DOI: 10.1177/1744806920904292] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
N-Acetylcysteine, one of the most prescribed antioxidant drugs, enhances pain
threshold in rodents and humans by activating mGlu2 metabotropic glutamate
receptors. Here, we assessed the analgesic activity of N-acetylcysteine in the
streptozotocin model of painful diabetic neuropathy and examined the effect of
N-acetylcysteine on proteins that are involved in mechanisms of nociceptive
sensitization. Mice with blood glucose levels ≥250 mg/dl in response to a single
intraperitoneal (i.p.) injection of streptozotocin (200 mg/kg) were used for the
assessment of mechanical pain thresholds. Systemic treatment with
N-acetylcysteine (100 mg/kg, i.p., either single injection or daily injections
for seven days) caused analgesia in diabetic mice. N-acetylcysteine-induced
analgesia was abrogated by the Sxc− inhibitors, sulfasalazine (8 mg/kg, i.p.), erastin (30 mg/kg,
i.p.), and sorafenib (10 mg/kg, i.p.), or by the mGlu2/3 receptor antagonist,
LY341495 (1 mg/kg, i.p.). Repeated administrations of N-acetylcysteine in
diabetic mice reduced ERK1/2 phosphorylation in the dorsal region of the lumbar
spinal cord. The analgesic activity of N-acetylcysteine was occluded by the MEK
inhibitor, PD0325901 (25 mg/kg, i.p.), the TRPV1 channel blocker, capsazepine
(40 mg/kg, i.p.), or by a cocktail of NMDA and mGlu5 metabotropic glutamate
receptor antagonists (memantine, 25 mg/kg, plus MTEP, 5 mg/kg,
both i.p.). These findings offer the first demonstration that N-acetylcysteine
relieves pain associated with diabetic neuropathy and holds promise for the use
of N-acetylcysteine as an add-on drug in diabetic patients.
Collapse
Affiliation(s)
| | - Pamela Scarselli
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Giada Mascio
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | | | | | - Santa Mammana
- IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | | | - Giorgio Cruccu
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Valeria Bruno
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Ferdinando Nicoletti
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Giuseppe Battaglia
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| |
Collapse
|
25
|
Zhang S, Chen Y, Wang Y, Zhang P, Chen G, Zhou Y. Insights Into Translatomics in the Nervous System. Front Genet 2021; 11:599548. [PMID: 33408739 PMCID: PMC7779767 DOI: 10.3389/fgene.2020.599548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Most neurological disorders are caused by abnormal gene translation. Generally, dysregulation of elements involved in the translational process disrupts homeostasis in neurons and neuroglia. Better understanding of how the gene translation process occurs requires detailed analysis of transcriptomic and proteomic profile data. However, a lack of strictly direct correlations between mRNA and protein levels limits translational investigation by combining transcriptomic and proteomic profiling. The much better correlation between proteins and translated mRNAs than total mRNAs in abundance and insufficiently sensitive proteomics approach promote the requirement of advances in translatomics technology. Translatomics which capture and sequence the mRNAs associated with ribosomes has been effective in identifying translational changes by genetics or projections, ribosome stalling, local translation, and transcript isoforms in the nervous system. Here, we place emphasis on the main three translatomics methods currently used to profile mRNAs attached to ribosome-nascent chain complex (RNC-mRNA). Their prominent applications in neurological diseases including glioma, neuropathic pain, depression, fragile X syndrome (FXS), neurodegenerative disorders are outlined. The content reviewed here expands our understanding on the contributions of aberrant translation to neurological disease development.
Collapse
Affiliation(s)
- Shuxia Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjie Wang
- Key Laboratory of Elemene Anti-Cancer Medicine of Zhejiang Province and Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou, China
| | - Piao Zhang
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Yousuf MS, Shiers SI, Sahn JJ, Price TJ. Pharmacological Manipulation of Translation as a Therapeutic Target for Chronic Pain. Pharmacol Rev 2021; 73:59-88. [PMID: 33203717 PMCID: PMC7736833 DOI: 10.1124/pharmrev.120.000030] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysfunction in regulation of mRNA translation is an increasingly recognized characteristic of many diseases and disorders, including cancer, diabetes, autoimmunity, neurodegeneration, and chronic pain. Approximately 50 million adults in the United States experience chronic pain. This economic burden is greater than annual costs associated with heart disease, cancer, and diabetes combined. Treatment options for chronic pain are inadequately efficacious and riddled with adverse side effects. There is thus an urgent unmet need for novel approaches to treating chronic pain. Sensitization of neurons along the nociceptive pathway causes chronic pain states driving symptoms that include spontaneous pain and mechanical and thermal hypersensitivity. More than a decade of preclinical research demonstrates that translational mechanisms regulate the changes in gene expression that are required for ongoing sensitization of nociceptive sensory neurons. This review will describe how key translation regulation signaling pathways, including the integrated stress response, mammalian target of rapamycin, AMP-activated protein kinase (AMPK), and mitogen-activated protein kinase-interacting kinases, impact the translation of different subsets of mRNAs. We then place these mechanisms of translation regulation in the context of chronic pain states, evaluate currently available therapies, and examine the potential for developing novel drugs. Considering the large body of evidence now published in this area, we propose that pharmacologically manipulating specific aspects of the translational machinery may reverse key neuronal phenotypic changes causing different chronic pain conditions. Therapeutics targeting these pathways could eventually be first-line drugs used to treat chronic pain disorders. SIGNIFICANCE STATEMENT: Translational mechanisms regulating protein synthesis underlie phenotypic changes in the sensory nervous system that drive chronic pain states. This review highlights regulatory mechanisms that control translation initiation and how to exploit them in treating persistent pain conditions. We explore the role of mammalian/mechanistic target of rapamycin and mitogen-activated protein kinase-interacting kinase inhibitors and AMPK activators in alleviating pain hypersensitivity. Modulation of eukaryotic initiation factor 2α phosphorylation is also discussed as a potential therapy. Targeting specific translation regulation mechanisms may reverse changes in neuronal hyperexcitability associated with painful conditions.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Stephanie I Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - James J Sahn
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| |
Collapse
|
27
|
Kumar S, Vinayak M. Quercetin Ameliorates CFA-Induced Chronic Inflammatory Hyperalgesia via Modulation of ROS-Mediated ERK1/2 Signaling and Inhibition of Spinal Glial Activation In Vivo. Neuromolecular Med 2020; 22:517-533. [DOI: 10.1007/s12017-020-08609-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 08/11/2020] [Indexed: 01/26/2023]
|
28
|
Qiu S, Liu B, Mo Y, Wang X, Zhong L, Han X, Mi F. MiR-101 promotes pain hypersensitivity in rats with chronic constriction injury via the MKP-1 mediated MAPK pathway. J Cell Mol Med 2020; 24:8986-8997. [PMID: 32656992 PMCID: PMC7417728 DOI: 10.1111/jcmm.15532] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 05/30/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
This study was performed to characterize the effect of microRNA‐101 (miR‐101) on the pain hypersensitivity in CCI rat models with the involvement of mitogen‐activated protein kinase phosphatase 1 (MKP‐1) in spinal cord microglial cells. The mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) in the developed CCI models were determined to assess the hypersensitivity of rats to mechanical stimulation and thermal pain. To assess inflammation, the levels of interleukin (IL)‐1β, IL‐6 and tumour necrosis factor‐α (TNF‐α) in the spinal dorsal horns of CCI rats and lipopolysaccharide (LPS)‐activated microglial cells were examined. miR‐101 and MKP‐1 gain‐ and loss‐of‐function experiments were conducted in in vivo and in vitro settings to examine the roles of miR‐101 and MKP‐1 in CCI hypersensitivity and inflammation. The results showed that miR‐101 was highly expressed in the spinal dorsal horn and microglial cells of CCI rat models. Furthermore, overexpression of miR‐101 promoted the pain hypersensitivity in CCI rat models by reducing MWT and TWL. The overexpression of miR‐101 also promoted inflammation in LPS‐exposed microglial cells, as indicated by increased levels of IL‐1β, IL‐6 and TNF‐α. MiR‐101 was shown to target MKP‐1, inhibiting its expression. Moreover, miR‐101 promoted pain hypersensitivity in CCI rat models by inhibiting MKP‐1 expression and activating the mitogen‐activated protein kinase (MAPK) signalling pathway. Taken together, miR‐101 could potentially promote hypersensitivity and inflammatory response of microglial cells and aggravate neuropathic pain in CCI rat models by inhibiting MKP‐1 in the MAPK signalling pathway.
Collapse
Affiliation(s)
- Shuang Qiu
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Benjuan Liu
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Yanshuai Mo
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Xueqin Wang
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Lina Zhong
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Xiao Han
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| | - Fuli Mi
- Department of Anesthesiology, Linyi People's Hospital, Linyi, China
| |
Collapse
|
29
|
Li Y, Guo X, Sun L, Xiao J, Su S, Du S, Li Z, Wu S, Liu W, Mo K, Xia S, Chang Y, Denis D, Tao Y. N 6-Methyladenosine Demethylase FTO Contributes to Neuropathic Pain by Stabilizing G9a Expression in Primary Sensory Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902402. [PMID: 32670741 PMCID: PMC7341103 DOI: 10.1002/advs.201902402] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/27/2020] [Indexed: 05/23/2023]
Abstract
Nerve injury-induced change in gene expression in primary sensory neurons of dorsal root ganglion (DRG) is critical for neuropathic pain genesis. N6-methyladenosine (m6A) modification of RNA represents an additional layer of gene regulation. Here, it is reported that peripheral nerve injury increases the expression of the m6A demethylase fat-mass and obesity-associated proteins (FTO) in the injured DRG via the activation of Runx1, a transcription factor that binds to the Fto gene promoter. Mimicking this increase erases m6A in euchromatic histone lysine methyltransferase 2 (Ehmt2) mRNA (encoding the histone methyltransferase G9a) and elevates the level of G9a in DRG and leads to neuropathic pain symptoms. Conversely, blocking this increase reverses a loss of m6A sites in Ehmt2 mRNA and destabilizes the nerve injury-induced G9a upregulation in the injured DRG and alleviates nerve injury-associated pain hypersensitivities. FTO contributes to neuropathic pain likely through stabilizing nerve injury-induced upregulation of G9a, a neuropathic pain initiator, in primary sensory neurons.
Collapse
Affiliation(s)
- Yize Li
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Xinying Guo
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Linlin Sun
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Jifang Xiao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Songxue Su
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
| | - Shibin Du
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Zhen Li
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Shaogen Wu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Weili Liu
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Kai Mo
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Shangzhou Xia
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Yun‐Juan Chang
- The Office of Advanced Research ComputingRutgers, The State University of New Jersey 185 S. Orange Ave., MSB C‐630NewarkNJ07103USA
| | - Daniel Denis
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
| | - Yuan‐Xiang Tao
- Department of AnesthesiologyNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E594NewarkNJ07103USA
- Department of Physiology, Pharmacology & NeuroscienceNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
- Department of Cell Biology & Molecular MedicineNew Jersey Medical School, RutgersThe State University of New Jersey185 S. Orange Ave., MSB E661NewarkNJ07103USA
| |
Collapse
|
30
|
Hill RZ, Bautista DM. Getting in Touch with Mechanical Pain Mechanisms. Trends Neurosci 2020; 43:311-325. [DOI: 10.1016/j.tins.2020.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/14/2020] [Accepted: 03/04/2020] [Indexed: 01/10/2023]
|
31
|
Peirs C, Dallel R, Todd AJ. Recent advances in our understanding of the organization of dorsal horn neuron populations and their contribution to cutaneous mechanical allodynia. J Neural Transm (Vienna) 2020; 127:505-525. [PMID: 32239353 PMCID: PMC7148279 DOI: 10.1007/s00702-020-02159-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023]
Abstract
The dorsal horns of the spinal cord and the trigeminal nuclei in the brainstem contain neuron populations that are critical to process sensory information. Neurons in these areas are highly heterogeneous in their morphology, molecular phenotype and intrinsic properties, making it difficult to identify functionally distinct cell populations, and to determine how these are engaged in pathophysiological conditions. There is a growing consensus concerning the classification of neuron populations, based on transcriptomic and transductomic analyses of the dorsal horn. These approaches have led to the discovery of several molecularly defined cell types that have been implicated in cutaneous mechanical allodynia, a highly prevalent and difficult-to-treat symptom of chronic pain, in which touch becomes painful. The main objective of this review is to provide a contemporary view of dorsal horn neuronal populations, and describe recent advances in our understanding of on how they participate in cutaneous mechanical allodynia.
Collapse
Affiliation(s)
- Cedric Peirs
- Université Clermont Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France.
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK.
| | - Radhouane Dallel
- Université Clermont Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, Clermont-Ferrand, F-63000, France
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Andrew J Todd
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
32
|
Lyu C, Lyu GW, Mulder J, Martinez A, Shi TJS. G Protein-Gated Inwardly Rectifying Potassium Channel Subunit 3 is Upregulated in Rat DRGs and Spinal Cord After Peripheral Nerve Injury. J Pain Res 2020; 13:419-429. [PMID: 32110090 PMCID: PMC7034995 DOI: 10.2147/jpr.s233744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background G protein-gated inwardly rectifying potassium (GIRK) channels are involved in the regulation of neuronal excitability. Four GIRK subunits (GIRK1-4) are expressed in rat dorsal root ganglia (DRGs). Recently, we have characterized the expression of GIRK1 and −2, and both are downregulated in rat DRGs and spinal cord after a complete sciatic nerve transection (axotomy). Here, we aimed to study the neurochemical characteristics of GIRK3, and its regulation in rat DRGs and spinal cord induced by nerve injury. Methods A sciatic nerve axotomy was performed to study the influences of injury on GIRK3 expression in DRGs and spinal cord. A dorsal root rhizotomy and a sciatic nerve crush were employed to study the axonal transport of GIRK3 protein, respectively. Immunohistochemistry analysis was employed for investigating the neurochemical characteristics of GIRK3. Results In control DRGs, ~18% of neuron profiles (NPs) were GIRK3-positive (+), and ~41%, ~48% and ~45% of GIRK3+ NPs were CGRP+, IB4+ and NF200+, respectively. GIRK3-like immunoreactivity was observed in glabrous skin of hind paws and axons originating from DRG neurons. Fourteen days after axotomy, more than one-third of DRG NPs were GIRK3+, and among these ~51% and ~56% coexpressed galanin and neuropeptide Y, respectively. In control animals, a small group of interneurons found in the dorsal horn was GIRK3+. In addition, GIRK3+ processes could be observed in superficial laminae of spinal dorsal horn. After nerve injury, the intensity of GIRK3-like immunoreactivity in the superficial layers was increased. Evidence based on rhizotomy and sciatic nerve crush indicated both anterograde and retrograde transport of GIRK3. Conclusion Our study demonstrates that GIRK3 is expressed in sensory neurons and spinal cord. GIRK3 has both anterograde and retrograde axonal transport. GIRK3 expression can be regulated by peripheral nerve injury.
Collapse
Affiliation(s)
- Chuang Lyu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, People's Republic of China
| | - Gong-Wei Lyu
- Department of Neurology, 1st Hospital of Harbin Medical University, Harbin 150001, People's Republic of China
| | - Jan Mulder
- Department of Neuroscience, Karolinska Institutet, Stockholm SE-171 77, Sweden.,Science for Life Laboratory, Karolinska Institutet, Stockholm SE-171 65, Sweden
| | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| | - Tie-Jun Sten Shi
- Department of Biomedicine, University of Bergen, Bergen 5009, Norway
| |
Collapse
|
33
|
Wolbert J, Cheng MI, Meyer zu Horste G, Su MA. Deciphering immune mechanisms in chronic inflammatory demyelinating polyneuropathies. JCI Insight 2020; 5:132411. [PMID: 32051341 DOI: 10.1172/jci.insight.132411] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disease of the peripheral nerves that presents with either chronic progression or relapsing disease. Recent studies in samples from patients with CIDP and mouse models have delineated how defects in central (thymic) and peripheral (extrathymic) immune tolerance mechanisms can cause PNS autoimmunity. Notably, nerve parenchymal cells actively contribute to local autoimmunity and also control disease outcome. Here, we outline how emerging technologies increasingly enable an integrated view of how immune cells and PNS parenchymal cells communicate in CIDP. We also relate the known heterogeneity of clinical presentation with specific underlying mechanisms. For example, a severe subtype of CIDP with tremor is associated with pathogenic IgG4 autoantibodies against nodal and paranodal proteins. An improved understanding of pathogenic mechanisms in CIDP will form the basis for more effective mechanism-based therapies.
Collapse
Affiliation(s)
- Jolien Wolbert
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Mandy I Cheng
- Department of Microbiology Immunology and Medical Genetics and
| | - Gerd Meyer zu Horste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Maureen A Su
- Department of Microbiology Immunology and Medical Genetics and.,Department of Pediatrics, UCLA, Los Angeles, California, USA
| |
Collapse
|
34
|
Expression and regulation of FRMD6 in mouse DRG neurons and spinal cord after nerve injury. Sci Rep 2020; 10:1880. [PMID: 32024965 PMCID: PMC7002571 DOI: 10.1038/s41598-020-58261-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/10/2020] [Indexed: 12/26/2022] Open
Abstract
FRMD6, a member of the group of FERM-domain proteins, is involved both in communication between cells, interactions with extracellular matrix, cellular apoptotic and regenerative mechanisms. FRMD6 was first discovered in the rodent sciatic nerve, and in the present immunohistochemical study we investigated the distribution of FRMD6 in the dorsal root ganglia (DRGs), sciatic nerve and spinal cord following sciatic nerve injury. FRMD6-immunoreactivity was found in the cytoplasm, nucleus or both, and in a majority of DRG neurons. FRMD6-immunoreactivity co-existed with several well-known neuronal markers, including calcitonin gene-related peptide, isolectin B4 and neurofilament 200 in mouse DRGs. After peripheral nerve injury, the FRMD6 mRNA levels and the overall percentage of FRMD6-positive neuron profiles (NPs) were decreased in ipsilateral lumbar DRGs, the latter mainly affecting small size neurons with cytoplasmic localization. Conversely, the proportion of NPs with nuclear FRMD6-immunoreactivity was significantly increased. In the sciatic nerve, FRMD6-immunoreactivity was observed in non-neuronal cells and in axons, and accumulated proximally to a ligation of the nerve. In the spinal cord FRMD6-immunoreactivity was detected in neurons in both dorsal and ventral horns, and was upregulated in ipsilateral dorsal horn after peripheral nerve axotomy. Our results demonstrate that FRMD6 is strictly regulated by peripheral nerve injury at the spinal level.
Collapse
|
35
|
Lyu C, Xia S, Lyu GW, Dun XP, Zheng K, Su J, Barde S, Xu ZQD, Hökfelt T, Shi TJS. A preliminary study on DRGs and spinal cord of a galanin receptor 2-EGFP transgenic mouse. Neuropeptides 2020; 79:102000. [PMID: 31864679 DOI: 10.1016/j.npep.2019.102000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 01/31/2023]
Abstract
The neuropeptide galanin functions via three G-protein coupled receptors, Gal1-3-R. Both Gal1-R and 2-R are involved in pain signaling at the spinal level. Here a Gal2-R-EGFP transgenic (TG) mouse was generated and studied in pain tests and by characterizing Gal2-R expression in both sensory ganglia and spinal cord. After peripheral spared nerve injury, mechanical allodynia developed and was ipsilaterally similar between wild type (WT) and TG mice. A Gal2-R-EGFP-positive signal was primarily observed in small and medium-sized dorsal root ganglion (DRG) neurons and in spinal interneurons and processes. No significant difference in size distribution of DRG neuronal profiles was found between TG and WT mice. Both percentage and fluorescence intensity of Gal2-R-EGFP-positive neuronal profiles were overall significantly upregulated in ipsilateral DRGs as compared to contralateral DRGs. There was an ipsilateral reduction in substance P-positive and calcitonin gene-related peptide (CGRP)-positive neuronal profiles, and this reduction was more pronounced in TG as compared to WT mice. Moreover, Gal2-R-EGFP partly co-localized with three pain-related neuropeptides, CGRP, neuropeptide Y and galanin, both in intact and injured DRGs, and with galanin also in local neurons in the superficial dorsal horn. Taken together, the present results provide novel information on the localization and phenotype of DRG and spinal neurons expressing the second galanin receptor, Gal2-R, and on phenotypic changes following peripheral nerve injury. Gal2-R may also be involved in autoreceptor signaling.
Collapse
Affiliation(s)
- Chuang Lyu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin 150069, PR China.
| | - Sheng Xia
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Gong-Wei Lyu
- Department of Neurology, 1st Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Xin-Peng Dun
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Kang Zheng
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Jie Su
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Neurobiology, Capital Medical University, Beijing 100069, PR China
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Tie-Jun Sten Shi
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway.
| |
Collapse
|
36
|
Stevens AM, Saleem M, Deal B, Janjic J, Pollock JA. Targeted cyclooxygenase-2 inhibiting nanomedicine results in pain-relief and differential expression of the RNA transcriptome in the dorsal root ganglia of injured male rats. Mol Pain 2020; 16:1744806920943309. [PMID: 32762277 PMCID: PMC7543154 DOI: 10.1177/1744806920943309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/18/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic constriction injury of the sciatic nerve in rats causes peripheral neuropathy leading to pain-like behaviors commonly seen in humans. Neuropathy is a leading cause of neuropathic pain, which involves a complex cellular and molecular response in the peripheral nervous system with interactions between neurons, glia, and infiltrating immune cells. In this study, we utilize a nonsteroidal anti-inflammatory drug -loaded nanoemulsion to deliver the cyclooxygenase-2 inhibitor, Celecoxib, directly to circulating monocytes following nerve injury, which provides long-lasting pain relief. However, it is not fully understood how cyclooxygenase-2 inhibition in a macrophage traveling to the site of injury impacts gene expression in the dorsal root ganglia. To elucidate aspects of the molecular mechanisms underlying pain-like behavior in chronic constriction injury, as well as subsequent pain relief with treatment, we employ RNAseq transcriptome profiling of the dorsal root ganglia associated with the injured sciatic nerve in rats. Using high throughput RNA sequencing in this way provides insight into the molecular mechanisms involved in this neuroinflammatory response. We compare the transcriptome from the dorsal root ganglias of the following study groups: chronic constriction injury animals administered with cyclooxygenase-2 inhibiting celecoxib-loaded nanoemulsion, chronic constriction injury animals administered with vehicle treatment, a drug-free nanoemulsion, and a group of naïve, unoperated and untreated rats. The results show an extensive differential expression of 115 genes. Using the protein annotation through evolutionary relationship classification system, we have revealed pain-related signaling pathways and underlying biological mechanisms involved in the neuroinflammatory response. Quantitative polymerase chain reaction validation confirms expression changes for several genes. This study shows that by directly inhibiting cyclooxygenase-2 activity in infiltrating macrophages at the injured sciatic nerve, there is an associated change in the transcriptome in the cell bodies of the dorsal root ganglia.
Collapse
Affiliation(s)
- Andrea M Stevens
- Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, USA
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA, USA
- Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, USA
| | - Muzamil Saleem
- Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, USA
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA, USA
- Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, USA
| | - Brooke Deal
- Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, USA
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA, USA
- Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, USA
| | - Jelena Janjic
- Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, USA
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - John A Pollock
- Bayer School of Natural and Environmental Sciences, Duquesne University, Pittsburgh, PA, USA
- Department of Biological Sciences, Duquesne University, Pittsburgh, PA, USA
- Chronic Pain Research Consortium, Duquesne University, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Kumar S, Vinayak M. NADPH oxidase1 inhibition leads to regression of central sensitization during formalin induced acute nociception via attenuation of ERK1/2-NFκB signaling and glial activation. Neurochem Int 2019; 134:104652. [PMID: 31891736 DOI: 10.1016/j.neuint.2019.104652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/02/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022]
Abstract
Role of NADPH oxidase1 in the development of inflammatory pain has been demonstrated by gene knockout studies. Nevertheless, pharmacological inhibition of NOX1 is a requisite approach for therapeutic utility. Recently, we have reported the anti-nociceptive effect of newly identified NOX1 specific inhibitor ML171 (2-acetylphenothiazine). Inhibition of NOX1 resulted in attenuation of nociceptive sensitization during acute inflammatory pain via inhibition of ROS generation and its downstream ERK1/2 activation. However, glial activation accompanying inflammation is closely related to the initiation and maintenance of pain. Peripheral nociceptive inputs activate the primary afferents via release of various chemical mediators which are potentially capable of mediating signals from neuron to glia in DRG and subsequently in spinal cord dorsal horn. The subsequent interactions between neuron and glia contribute to pain hypersensitivity. Thus, the present study was focused to investigate the effect of ML171 on ERK1/2 signaling, glial activation, and crosstalk between neuron and glia in a mouse model of formalin induced acute nociception. Thus, the present study was focused to investigate the effect of ML171 on ERK1/2 signaling, glial activation, and crosstalk between neuron and glia in DRG and dorsal horn of the spinal cord of lumbar region (L3-L5) in a mouse model of formalin induced acute nociception. Intraperitoneal administration of ML171 decreased nociceptive behavioral responses, i.e. the flinch and lick counts, in formalin induced nociceptive mice. Immunofluorescence and Western blot analysis demonstrated decreased levels of nociceptive mediators like p-ERK1/2, p-NFκB p65, Iba1 and GFAP in DRG as well as in spinal cord dorsal horn; supporting anti-nociceptive potential of ML171. Further, co-localization studies showed the neuron-glia crosstalk in tissue dependent manner. ERK1/2 was found to be activated in glia and NFκB in neurons in DRG; whereas in case of spinal cord ERK1/2 was activated in neurons and NFκB in astrocytes. Decrease in nociceptive behavioral response and activation of nociceptive mediators after intraperitoneal administration of ML171 strongly advocate anti-nociceptive potential of ML171. This is the first report demonstrating modulation of ERK1/2-NFκB signaling pathway, glial activation and regulation of neuron-glia crosstalk by NADPH oxidase1 inhibition towards its anti-nociceptive action.
Collapse
Affiliation(s)
- Sanjay Kumar
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Manjula Vinayak
- Department of Zoology, Biochemistry and Molecular Biology Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
38
|
Ren J, Liu N, Sun N, Zhang K, Yu L. Mesenchymal Stem Cells and their Exosomes: Promising Therapeutics for Chronic Pain. Curr Stem Cell Res Ther 2019; 14:644-653. [PMID: 31512998 DOI: 10.2174/1574888x14666190912162504] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/27/2019] [Accepted: 06/17/2019] [Indexed: 12/13/2022]
Abstract
Chronic pain is a common condition that seriously affects the quality of human life with
variable etiology and complicated symptoms; people who suffer from chronic pain may experience
anxiety, depression, insomnia, and other harmful emotions. Currently, chronic pain treatments are nonsteroidal
anti-inflammatory drugs and opioids; these drugs are demonstrated to be insufficient and
cause severe side effects. Therefore, research into new therapeutic strategies for chronic pain is a top
priority. In recent years, stem cell transplantation has been demonstrated to be a potent alternative for
the treatment of chronic pain. Mesenchymal stem cells (MSCs), a type of pluripotent stem cell, exhibit
multi-directional differentiation, promotion of stem cell implantation, and immune regulation; they
have also been shown to exert analgesic effects in several chronic pain models. Exosomes produced by
MSCs have been demonstrated to relieve painful symptoms with fewer side effects. In this review, we
summarize the therapeutic use of MSCs in various chronic pain studies. We also discuss ways to enhance
the treatment effect of MSCs. We predict in the future, cell-free therapies for chronic pain will
develop from exosomes secreted by MSCs.
Collapse
Affiliation(s)
- Jinxuan Ren
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Kehan Zhang
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lina Yu
- Department of Anesthesiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Differences between Dorsal Root and Trigeminal Ganglion Nociceptors in Mice Revealed by Translational Profiling. J Neurosci 2019; 39:6829-6847. [PMID: 31253755 DOI: 10.1523/jneurosci.2663-18.2019] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/08/2023] Open
Abstract
Nociceptors located in the trigeminal ganglion (TG) and DRG are the primary sensors of damaging or potentially damaging stimuli for the head and body, respectively, and are key drivers of chronic pain states. While nociceptors in these two tissues show a high degree of functional similarity, there are important differences in their development lineages, their functional connections to the CNS, and recent genome-wide analyses of gene expression suggest that they possess some unique genomic signatures. Here, we used translating ribosome affinity purification to comprehensively characterize and compare mRNA translation in Scn10a-positive nociceptors in the TG and DRG of male and female mice. This unbiased method independently confirms several findings of differences between TG and DRG nociceptors described in the literature but also suggests preferential utilization of key signaling pathways. Most prominently, we provide evidence that translational efficiency in mechanistic target of rapamycin (mTOR)-related genes is higher in the TG compared with DRG, whereas several genes associated with the negative regulator of mTOR, AMP-activated protein kinase, have higher translational efficiency in DRG nociceptors. Using capsaicin as a sensitizing stimulus, we show that behavioral responses are greater in the TG region and this effect is completely reversible with mTOR inhibition. These findings have implications for the relative capacity of these nociceptors to be sensitized upon injury. Together, our data provide a comprehensive, comparative view of transcriptome and translatome activity in TG and DRG nociceptors that enhances our understanding of nociceptor biology.SIGNIFICANCE STATEMENT The DRG and trigeminal ganglion (TG) provide sensory information from the body and head, respectively. Nociceptors in these tissues are critical first neurons in the pain pathway. Injury to peripheral neurons in these tissues can cause chronic pain. Interestingly, clinical and preclinical findings support the conclusion that injury to TG neurons is more likely to cause chronic pain and chronic pain in the TG area is more intense and more difficult to treat. We used translating ribosome affinity purification technology to gain new insight into potential differences in the translatomes of DRG and TG neurons. Our findings demonstrate previously unrecognized differences between TG and DRG nociceptors that provide new insight into how injury may differentially drive plasticity states in nociceptors in these two tissues.
Collapse
|
40
|
Price TJ, Ray PR. Recent advances toward understanding the mysteries of the acute to chronic pain transition. CURRENT OPINION IN PHYSIOLOGY 2019; 11:42-50. [PMID: 32322780 DOI: 10.1016/j.cophys.2019.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic pain affects up to a third of the population. Ongoing epidemiology studies suggest that the impact of chronic pain on the population is accelerating [1]. While advances have been made in understanding how chronic pain develops, there are still many important mysteries about how acute pain transitions to a chronic state. In this review, I summarize recent developments in the field with a focus on several areas of emerging research that are likely to have an important impact on the field. These include mechanisms of cellular plasticity that drive chronic pain, evidence of pervasive sex differential mechanisms in chronic pain and the profound impact that next generation sequencing technologies are having on this area of research.
Collapse
Affiliation(s)
- Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
41
|
de la Peña JBI, Song JJ, Campbell ZT. RNA control in pain: Blame it on the messenger. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1546. [PMID: 31090211 DOI: 10.1002/wrna.1546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/12/2022]
Abstract
mRNA function is meticulously controlled. We provide an overview of the integral role that posttranscriptional controls play in the perception of painful stimuli by sensory neurons. These specialized cells, termed nociceptors, precisely regulate mRNA polarity, translation, and stability. A growing body of evidence has revealed that targeted disruption of mRNAs and RNA-binding proteins robustly diminishes pain-associated behaviors. We propose that the use of multiple independent regulatory paradigms facilitates robust temporal and spatial precision of protein expression in response to a range of pain-promoting stimuli. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation RNA Turnover and Surveillance > Regulation of RNA Stability.
Collapse
Affiliation(s)
- June Bryan I de la Peña
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Jane J Song
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| |
Collapse
|
42
|
Uttam S, Wong C, Price TJ, Khoutorsky A. eIF4E-Dependent Translational Control: A Central Mechanism for Regulation of Pain Plasticity. Front Genet 2018; 9:470. [PMID: 30459806 PMCID: PMC6232926 DOI: 10.3389/fgene.2018.00470] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Translational control of gene expression has emerged as a key mechanism in regulating different forms of long-lasting neuronal plasticity. Maladaptive plastic reorganization of peripheral and spinal nociceptive circuits underlies many chronic pain states and relies on new gene expression. Accordingly, downregulation of mRNA translation in primary afferents and spinal dorsal horn neurons inhibits tissue injury-induced sensitization of nociceptive pathways, supporting a central role for translation dysregulation in the development of persistent pain. Translation is primarily regulated at the initiation stage via the coordinated activity of translation initiation factors. The mRNA cap-binding protein, eukaryotic translation initiation factor 4E (eIF4E), is involved in the recruitment of the ribosome to the mRNA cap structure, playing a central role in the regulation of translation initiation. eIF4E integrates inputs from the mTOR and ERK signaling pathways, both of which are activated in numerous painful conditions to regulate the translation of a subset of mRNAs. Many of these mRNAs are involved in the control of cell growth, proliferation, and neuroplasticity. However, the full repertoire of eIF4E-dependent mRNAs in the nervous system and their translation regulatory mechanisms remain largely unknown. In this review, we summarize the current evidence for the role of eIF4E-dependent translational control in the sensitization of pain circuits and present pharmacological approaches to target these mechanisms. Understanding eIF4E-dependent translational control mechanisms and their roles in aberrant plasticity of nociceptive circuits might reveal novel therapeutic targets to treat persistent pain states.
Collapse
Affiliation(s)
- Sonali Uttam
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Calvin Wong
- Department of Anesthesia, McGill University, Montreal, QC, Canada
| | - Theodore J. Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Center for Advanced Pain Studies, The University of Texas at Dallas, Richardson, TX, United States
| | - Arkady Khoutorsky
- Department of Anesthesia, McGill University, Montreal, QC, Canada
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Starobova H, S. W. A. H, Lewis RJ, Vetter I. Transcriptomics in pain research: insights from new and old technologies. Mol Omics 2018; 14:389-404. [DOI: 10.1039/c8mo00181b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Physiological and pathological pain involves a complex interplay of multiple cell types and signaling pathways.
Collapse
Affiliation(s)
- H. Starobova
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - Himaya S. W. A.
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - R. J. Lewis
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| | - I. Vetter
- Centre for Pain Research
- Institute for Molecular Bioscience
- University of Queensland
- St Lucia
- Australia
| |
Collapse
|