1
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of NaV1.7-selective inhibitors for pain. Pain 2024:00006396-990000000-00751. [PMID: 39446737 DOI: 10.1097/j.pain.0000000000003425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel NaV1.7 plays an important role in pain processing according to genetic data. Those data made NaV1.7 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, NaV1.7-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 NaV1.7-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 NaV1.7 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 NaV1.7-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
|
2
|
St Clair-Glover M, Finol-Urdaneta RK, Maddock M, Wallace E, Miellet S, Wallace G, Yue Z, Dottori M. Efficient fabrication of 3D bioprinted functional sensory neurons using an inducible Neurogenin-2 human pluripotent stem cell line. Biofabrication 2024; 16:045022. [PMID: 39084624 DOI: 10.1088/1758-5090/ad69c4] [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: 12/28/2023] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Three-dimensional (3D) tissue models have gained recognition for their improved ability to mimic the native cell microenvironment compared to traditional two-dimensional models. This progress has been driven by advances in tissue-engineering technologies such as 3D bioprinting, a promising method for fabricating biomimetic living tissues. While bioprinting has succeeded in generating various tissues to date, creating neural tissue models remains challenging. In this context, we present an accelerated approach to fabricate 3D sensory neuron (SN) structures using a transgenic human pluripotent stem cell (hPSC)-line that contains an inducible Neurogenin-2 (NGN2) expression cassette. The NGN2 hPSC line was first differentiated to neural crest cell (NCC) progenitors, then incorporated into a cytocompatible gelatin methacryloyl-based bioink for 3D bioprinting. Upregulated NGN2 expression in the bioprinted NCCs resulted in induced SN (iSN) populations that exhibited specific cell markers, with 3D analysis revealing widespread neurite outgrowth through the scaffold volume. Calcium imaging demonstrated functional activity of iSNs, including membrane excitability properties and voltage-gated sodium channel (NaV) activity. This efficient approach to generate 3D bioprinted iSN structures streamlines the development of neural tissue models, useful for the study of neurodevelopment and disease states and offering translational potential.
Collapse
Affiliation(s)
- Mitchell St Clair-Glover
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marnie Maddock
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Eileen Wallace
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Sara Miellet
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Gordon Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Zhilian Yue
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| |
Collapse
|
3
|
Du L, Zhu J, Liu S, Yang W, Hu X, Zhang W, Cui W, Yang Y, Wang C, Yang Y, Gao T, Zhang C, Zhang R, Lou M, Zhou H, Rao J, Maoying Q, Chu Y, Wang Y, Mi W. Transient receptor potential melastatin 8 contributes to the interleukin-33-mediated cold allodynia in a mouse model of neuropathic pain. Pain 2024:00006396-990000000-00679. [PMID: 39132923 DOI: 10.1097/j.pain.0000000000003346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/17/2024] [Indexed: 08/13/2024]
Abstract
ABSTRACT Cold allodynia is a common complaint of patients suffering from neuropathic pain initiated by peripheral nerve injury. However, the mechanisms that drive neuropathic cold pain remain elusive. In this study, we show that the interleukin (IL)-33/ST2 signaling in the dorsal root ganglion (DRG) is a critical contributor to neuropathic cold pain by interacting with the cold sensor transient receptor potential melastatin 8 (TRPM8). By using the St2-/- mice, we demonstrate that ST2 is required for the generation of nociceptor hyperexcitability and cold allodynia in a mouse model of spared nerve injury (SNI). Moreover, the selective elimination of ST2 function from the Nav1.8-expressing nociceptor markedly suppresses SNI-induced cold allodynia. Consistent with the loss-of-function studies, intraplantar injection of recombinant IL-33 (rIL-33) is sufficient to induce cold allodynia. Mechanistically, ST2 is co-expressed with TRPM8 in both mouse and human DRG neurons and rIL-33-induced Ca2+ influx in mouse DRG neurons through TRPM8. Co-immunoprecipitation assays further reveal that ST2 interacts with TRPM8 in DRG neurons. Importantly, rIL-33-induced cold allodynia is abolished by pharmacological inhibition of TRPM8 and genetic ablation of the TRPM8-expressing neurons. Thus, our findings suggest that the IL-33/ST2 signaling mediates neuropathic cold pain through downstream cold-sensitive TRPM8 channels, thereby identifying a potential analgesic target for the treatment of neuropathic cold pain.
Collapse
Affiliation(s)
- Lixia Du
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Biochemistry, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianyu Zhu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shenbin Liu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xueming Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenwen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenqiang Cui
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yayue Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chenghao Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yachen Yang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tianchi Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruofan Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengping Lou
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong Zhou
- Department of Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Jia Rao
- Department of Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Qiliang Maoying
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Yuxia Chu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Wenli Mi
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Stein C. Effects of pH on opioid receptor activation and implications for drug design. Biophys J 2024:S0006-3495(24)00446-6. [PMID: 38970252 DOI: 10.1016/j.bpj.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
G-protein-coupled receptors are integral membrane proteins that transduce chemical signals from the extracellular matrix into the cell. Traditional drug design has considered ligand-receptor interactions only under normal conditions. However, studies on opioids indicate that such interactions are very different in diseased tissues. In such microenvironments, protons play an important role in structural and functional alterations of both ligands and receptors. The pertinent literature strongly suggests that future drug design should take these aspects into account in order to reduce adverse side effects while preserving desired effects of novel compounds.
Collapse
Affiliation(s)
- Christoph Stein
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Experimental Anaesthesiology, Berlin, Germany.
| |
Collapse
|
5
|
Li H, Deng Z, Yu X, Lin J, Xie Y, Liao W, Ma Y, Zheng Q. Combining dual-view fusion pose estimation and multi-type motion feature extraction to assess arthritis pain in mice. Biomed Signal Process Control 2024; 92:106080. [DOI: 10.1016/j.bspc.2024.106080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
|
6
|
Dasgupta I, Rangineni DP, Abdelsaid H, Ma Y, Bhushan A. Tiny Organs, Big Impact: How Microfluidic Organ-on-Chip Technology Is Revolutionizing Mucosal Tissues and Vasculature. Bioengineering (Basel) 2024; 11:476. [PMID: 38790343 PMCID: PMC11117503 DOI: 10.3390/bioengineering11050476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-chip (OOC) technology has gained importance for biomedical studies and drug development. This technology involves microfluidic devices that mimic the structure and function of specific human organs or tissues. OOCs are a promising alternative to traditional cell-based models and animals, as they provide a more representative experimental model of human physiology. By creating a microenvironment that closely resembles in vivo conditions, OOC platforms enable the study of intricate interactions between different cells as well as a better understanding of the underlying mechanisms pertaining to diseases. OOCs can be integrated with other technologies, such as sensors and imaging systems to monitor real-time responses and gather extensive data on tissue behavior. Despite these advances, OOCs for many organs are in their initial stages of development, with several challenges yet to be overcome. These include improving the complexity and maturity of these cellular models, enhancing their reproducibility, standardization, and scaling them up for high-throughput uses. Nonetheless, OOCs hold great promise in advancing biomedical research, drug discovery, and personalized medicine, benefiting human health and well-being. Here, we review several recent OOCs that attempt to overcome some of these challenges. These OOCs with unique applications can be engineered to model organ systems such as the stomach, cornea, blood vessels, and mouth, allowing for analyses and investigations under more realistic conditions. With this, these models can lead to the discovery of potential therapeutic interventions. In this review, we express the significance of the relationship between mucosal tissues and vasculature in organ-on-chip (OOC) systems. This interconnection mirrors the intricate physiological interactions observed in the human body, making it crucial for achieving accurate and meaningful representations of biological processes within OOC models. Vasculature delivers essential nutrients and oxygen to mucosal tissues, ensuring their proper function and survival. This exchange is critical for maintaining the health and integrity of mucosal barriers. This review will discuss the OOCs used to represent the mucosal architecture and vasculature, and it can encourage us to think of ways in which the integration of both can better mimic the complexities of biological systems and gain deeper insights into various physiological and pathological processes. This will help to facilitate the development of more accurate predictive models, which are invaluable for advancing our understanding of disease mechanisms and developing novel therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA; (I.D.); (D.P.R.); (H.A.); (Y.M.)
| |
Collapse
|
7
|
Xie YF, Yang J, Ratté S, Prescott SA. Similar excitability through different sodium channels and implications for the analgesic efficacy of selective drugs. eLife 2024; 12:RP90960. [PMID: 38687187 PMCID: PMC11060714 DOI: 10.7554/elife.90960] [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: 05/02/2024] Open
Abstract
Nociceptive sensory neurons convey pain-related signals to the CNS using action potentials. Loss-of-function mutations in the voltage-gated sodium channel NaV1.7 cause insensitivity to pain (presumably by reducing nociceptor excitability) but clinical trials seeking to treat pain by inhibiting NaV1.7 pharmacologically have struggled. This may reflect the variable contribution of NaV1.7 to nociceptor excitability. Contrary to claims that NaV1.7 is necessary for nociceptors to initiate action potentials, we show that nociceptors can achieve similar excitability using different combinations of NaV1.3, NaV1.7, and NaV1.8. Selectively blocking one of those NaV subtypes reduces nociceptor excitability only if the other subtypes are weakly expressed. For example, excitability relies on NaV1.8 in acutely dissociated nociceptors but responsibility shifts to NaV1.7 and NaV1.3 by the fourth day in culture. A similar shift in NaV dependence occurs in vivo after inflammation, impacting ability of the NaV1.7-selective inhibitor PF-05089771 to reduce pain in behavioral tests. Flexible use of different NaV subtypes exemplifies degeneracy - achieving similar function using different components - and compromises reliable modulation of nociceptor excitability by subtype-selective inhibitors. Identifying the dominant NaV subtype to predict drug efficacy is not trivial. Degeneracy at the cellular level must be considered when choosing drug targets at the molecular level.
Collapse
Affiliation(s)
- Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
| | - Steven A Prescott
- Neurosciences and Mental Health, The Hospital for Sick ChildrenTorontoCanada
- Institute of Biomedical Engineering, University of TorontoTorontoCanada
- Department of Physiology, University of TorontoTorontoCanada
| |
Collapse
|
8
|
Liu PW, Zhang H, Werley CA, Pichler M, Ryan SJ, Lewarch CL, Jacques J, Grooms J, Ferrante J, Li G, Zhang D, Bremmer N, Barnett A, Chantre R, Elder AE, Cohen AE, Williams LA, Dempsey GT, McManus OB. A phenotypic screening platform for chronic pain therapeutics using all-optical electrophysiology. Pain 2024; 165:922-940. [PMID: 37963235 PMCID: PMC10950549 DOI: 10.1097/j.pain.0000000000003090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
ABSTRACT Chronic pain associated with osteoarthritis (OA) remains an intractable problem with few effective treatment options. New approaches are needed to model the disease biology and to drive discovery of therapeutics. We present an in vitro model of OA pain, where dorsal root ganglion (DRG) sensory neurons were sensitized by a defined mixture of disease-relevant inflammatory mediators, here called Sensitizing PAin Reagent Composition or SPARC. Osteoarthritis-SPARC components showed synergistic or additive effects when applied in combination and induced pain phenotypes in vivo. To measure the effect of OA-SPARC on neural firing in a scalable format, we used a custom system for high throughput all-optical electrophysiology. This system enabled light-based membrane voltage recordings from hundreds of neurons in parallel with single cell and single action potential resolution and a throughput of up to 500,000 neurons per day. A computational framework was developed to construct a multiparameter OA-SPARC neuronal phenotype and to quantitatively assess phenotype reversal by candidate pharmacology. We screened ∼3000 approved drugs and mechanistically focused compounds, yielding data from over 1.2 million individual neurons with detailed assessment of functional OA-SPARC phenotype rescue and orthogonal "off-target" effects. Analysis of confirmed hits revealed diverse potential analgesic mechanisms including ion channel modulators and other mechanisms including MEK inhibitors and tyrosine kinase modulators. Our results suggest that the Raf-MEK-ERK axis in DRG neurons may integrate the inputs from multiple upstream inflammatory mediators found in osteoarthritis patient joints, and MAPK pathway activation in DRG neurons may contribute to chronic pain in patients with osteoarthritis.
Collapse
Affiliation(s)
- Pin W. Liu
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | | | | | | | | | - Guangde Li
- Quiver Bioscience, Cambridge, MA, United States
| | - Dawei Zhang
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | | | | | | |
Collapse
|
9
|
Bang S, Jiang C, Xu J, Chandra S, McGinnis A, Luo X, He Q, Li Y, Wang Z, Ao X, Parisien M, Fernandes de Araujo LO, Esfahan SJ, Zhang Q, Tonello R, Berta T, Diatchenko L, Ji RR. Satellite glial GPR37L1 regulates maresin and potassium channel signaling for pain control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.03.569787. [PMID: 38106084 PMCID: PMC10723316 DOI: 10.1101/2023.12.03.569787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
G protein coupled receptor 37-like 1 (GPR37L1) is an orphan GPCR and its function remains largely unknown. Here we report that GPR37L1 transcript is highly expressed compared to all known GPCRs in mouse and human dorsal root ganglia (DRGs) and selectively expressed in satellite glial cells (SGCs). Peripheral neuropathy following diabetes and chemotherapy by streptozotocin and paclitaxel resulted in downregulations of surface GPR37L1 in mouse and human DRGs. Transgenic mice with Gpr37l1 deficiency exhibited impaired resolution of neuropathic pain symptom (mechanical allodynia), whereas overexpression of Gpr37l1 in mouse DRGs can reverse neuropathic pain. Notably, GPR37L1 is co-expressed and coupled with potassium channels in SGCs. We found striking species differences in potassium channel expression in SGCs, with predominant expression of KCNJ10 and KCNJ3 in mouse and human SGCs, respectively. GPR37L1 regulates the surface expression and function of KCNJ10 and KCNJ3. We identified the pro-resolving lipid mediator maresin 1 (MaR1) as a GPR37L1 ligand. MaR1 increases KCNJ10/KCNJ3-mediated potassium influx in SGCs via GPR37L1. MaR1 protected chemotherapy-induced suppression of KCNJ13/KCNJ10 expression and function in SGCs. Finally, genetic analysis revealed that the GPR37L1-E296K variant is associated with increased chronic pain risk by destabilizing the protein. Thus, GPR37L1 in SGCs offers a new target for neuropathy protection and pain control.
Collapse
|
10
|
Lawn T, Howard MA, Turkheimer F, Misic B, Deco G, Martins D, Dipasquale O. From neurotransmitters to networks: Transcending organisational hierarchies with molecular-informed functional imaging. Neurosci Biobehav Rev 2023; 150:105193. [PMID: 37086932 PMCID: PMC10390343 DOI: 10.1016/j.neubiorev.2023.105193] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/01/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
The human brain exhibits complex interactions across micro, meso-, and macro-scale organisational principles. Recent synergistic multi-modal approaches have begun to link micro-scale information to systems level dynamics, transcending organisational hierarchies and offering novel perspectives into the brain's function and dysfunction. Specifically, the distribution of micro-scale properties (such as receptor density or gene expression) can be mapped onto macro-scale measures from functional MRI to provide novel neurobiological insights. Methodological approaches to enrich functional imaging analyses with molecular information are rapidly evolving, with several streams of research having developed relatively independently, each offering unique potential to explore the trans-hierarchical functioning of the brain. Here, we address the three principal streams of research - spatial correlation, molecular-enriched network, and in-silico whole brain modelling analyses - to provide a critical overview of the different sources of molecular information, how this information can be utilised within analyses of fMRI data, the merits and pitfalls of each methodology, and, through the use of key examples, highlight their promise to shed new light on key domains of neuroscientific inquiry.
Collapse
Affiliation(s)
- Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Matthew A Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Bratislav Misic
- Montreal Neurological Institute and Hospital, McGill University, Montreal, Québec, Canada
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Ramon Trias Fargas 25-27, Barcelona 08005, Spain; Department of Neuropsychology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
11
|
Kumar V, Kingsley D, Perikamana SM, Mogha P, Goodwin CR, Varghese S. Self-assembled innervated vasculature-on-a-chip to study nociception. Biofabrication 2023; 15:10.1088/1758-5090/acc904. [PMID: 36996841 PMCID: PMC10152403 DOI: 10.1088/1758-5090/acc904] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/30/2023] [Indexed: 04/01/2023]
Abstract
Nociceptor sensory neurons play a key role in eliciting pain. An active crosstalk between nociceptor neurons and the vascular system at the molecular and cellular level is required to sense and respond to noxious stimuli. Besides nociception, interaction between nociceptor neurons and vasculature also contributes to neurogenesis and angiogenesis.In vitromodels of innervated vasculature can greatly help delineate these roles while facilitating disease modeling and drug screening. Herein, we report the development of a microfluidic-assisted tissue model of nociception in the presence of microvasculature. The self-assembled innervated microvasculature was engineered using endothelial cells and primary dorsal root ganglion (DRG) neurons. The sensory neurons and the endothelial cells displayed distinct morphologies in presence of each other. The neurons exhibited an elevated response to capsaicin in the presence of vasculature. Concomitantly, increased transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor expression was observed in the DRG neurons in presence of vascularization. Finally, we demonstrated the applicability of this platform for modeling nociception associated with tissue acidosis. While not demonstrated here, this platform could also serve as a tool to study pain resulting from vascular disorders while also paving the way towards the development of innervated microphysiological models.
Collapse
Affiliation(s)
- Vardhman Kumar
- Department of Biomedical Engineering, Duke University, Durham NC
| | - David Kingsley
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
| | | | - Pankaj Mogha
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
| | - C Rory Goodwin
- Department of Neurosurgery, Spine Division, Duke University Medical Center, Durham, NC
| | - Shyni Varghese
- Department of Biomedical Engineering, Duke University, Durham NC
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
- Department of Mechanical Engineering and Material Science, Duke University, Durham NC
| |
Collapse
|
12
|
Shin SM, Lauzadis J, Itson-Zoske B, Cai Y, Fan F, Natarajan GK, Kwok WM, Puopolo M, Hogan QH, Yu H. Targeting intrinsically disordered regions facilitates discovery of calcium channels 3.2 inhibitory peptides for adeno-associated virus-mediated peripheral analgesia. Pain 2022; 163:2466-2484. [PMID: 35420557 PMCID: PMC9562599 DOI: 10.1097/j.pain.0000000000002650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/19/2022] [Accepted: 03/23/2022] [Indexed: 11/27/2022]
Abstract
ABSTRACT Ample data support a prominent role of peripheral T-type calcium channels 3.2 (Ca V 3.2) in generating pain states. Development of primary sensory neuron-specific inhibitors of Ca V 3.2 channels is an opportunity for achieving effective analgesic therapeutics, but success has been elusive. Small peptides, especially those derived from natural proteins as inhibitory peptide aptamers (iPAs), can produce highly effective and selective blockade of specific nociceptive molecular pathways to reduce pain with minimal off-target effects. In this study, we report the engineering of the potent and selective iPAs of Ca V 3.2 from the intrinsically disordered regions (IDRs) of Ca V 3.2 intracellular segments. Using established prediction algorithms, we localized the IDRs in Ca V 3.2 protein and identified several Ca V 3.2iPA candidates that significantly reduced Ca V 3.2 current in HEK293 cells stably expressing human wide-type Ca V 3.2. Two prototype Ca V 3.2iPAs (iPA1 and iPA2) derived from the IDRs of Ca V 3.2 intracellular loops 2 and 3, respectively, were expressed selectively in the primary sensory neurons of dorsal root ganglia in vivo using recombinant adeno-associated virus (AAV), which produced sustained inhibition of calcium current conducted by Ca V 3.2/T-type channels and significantly attenuated both evoked and spontaneous pain behavior in rats with neuropathic pain after tibial nerve injury. Recordings from dissociated sensory neurons showed that AAV-mediated Ca V 3.2iPA expression suppressed neuronal excitability, suggesting that Ca V 3.2iPA treatment attenuated pain by reversal of injury-induced neuronal hypersensitivity. Collectively, our results indicate that Ca V 3.2iPAs are promising analgesic leads that, combined with AAV-mediated delivery in anatomically targeted sensory ganglia, have the potential to be a selective peripheral Ca V 3.2-targeting strategy for clinical treatment of pain.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justas Lauzadis
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yongsong Cai
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Fan Fan
- Department of Pharmacology and Toxicology, The University of Mississippi Medical Center, Jackson, MS, United States
| | - Gayathri K. Natarajan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Wai-Meng Kwok
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michelino Puopolo
- Department of Anesthesiology, Stony Brook University, Stony Brook, NY, United States
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
13
|
Vamvakas A, Lawn T, Veronese M, Williams SCR, Tsougos I, Howard MA. Neurotransmitter receptor densities are associated with changes in regional Cerebral blood flow during clinical ongoing pain. Hum Brain Mapp 2022; 43:5235-5249. [PMID: 35796178 PMCID: PMC9812236 DOI: 10.1002/hbm.25999] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/07/2022] [Accepted: 06/19/2022] [Indexed: 01/15/2023] Open
Abstract
Arterial spin labelling (ASL) plays an increasingly important role in neuroimaging pain research but does not provide molecular insights regarding how regional cerebral blood flow (rCBF) relates to underlying neurotransmission. Here, we integrate ASL with positron emission tomography (PET) and brain transcriptome data to investigate the molecular substrates of rCBF underlying clinically relevant pain states. Two data sets, representing acute and chronic ongoing pain respectively, were utilised to quantify changes in rCBF; one examining pre-surgical versus post-surgical pain, and the second comparing patients with painful hand Osteoarthritis to a group of matched controls. We implemented a whole-brain spatial correlation analysis to explore associations between change in rCBF (ΔCBF) and neurotransmitter receptor distributions derived from normative PET templates. Additionally, we utilised transcriptomic data from the Allen Brain Atlas to inform distributions of receptor expression. Both datasets presented significant correlations of ΔCBF with the μ-opioid and dopamine-D2 receptor expressions, which play fundamental roles in brain activity associated with pain experiences. ΔCBF also correlated with the gene expression distributions of several receptors involved in pain processing. Overall, this is the first study illustrating the molecular basis of ongoing pain ASL indices and emphasises the potential of rCBF as a biomarker in pain research.
Collapse
Affiliation(s)
- Alexandros Vamvakas
- Medical Physics Department, Medical SchoolUniversity of ThessalyLarisaGreece
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Timothy Lawn
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Steven C. R. Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Ioannis Tsougos
- Medical Physics Department, Medical SchoolUniversity of ThessalyLarisaGreece
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Matthew A. Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
14
|
Li Y, Li T, Du Y, Wu S. Structure-based drug discovery facilitates future painkiller development. Clin Transl Med 2022; 12:e1120. [PMID: 36412509 PMCID: PMC9680165 DOI: 10.1002/ctm2.1120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Yuqing Li
- Department of Experiment & ResearchSouth China Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Teng Li
- Department of Experiment & ResearchSouth China Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Yang Du
- School of MedicineKobilka Institute of Innovative Drug DiscoveryThe Chinese University of Hong KongShenzhenGuangdongChina
| | - Song Wu
- Department of Experiment & ResearchSouth China Hospital of Shenzhen UniversityShenzhenGuangdongChina
| |
Collapse
|
15
|
Abdus-Saboor I, Luo W. Measuring Mouse Somatosensory Reflexive Behaviors with High-speed Videography, Statistical Modeling, and Machine Learning. NEUROMETHODS 2022; 178:441-456. [PMID: 35783537 PMCID: PMC9249079 DOI: 10.1007/978-1-0716-2039-7_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Objectively measuring and interpreting an animal's sensory experience remains a challenging task. This is particularly true when using preclinical rodent models to study pain mechanisms and screen for potential new pain treatment reagents. How to determine their pain states in a precise and unbiased manner is a hurdle that the field will need to overcome. Here, we describe our efforts to measure mouse somatosensory reflexive behaviors with greatly improved precision by high-speed video imaging. We describe how coupling sub-second ethograms of reflexive behaviors with a statistical reduction method and supervised machine learning can be used to create a more objective quantitative mouse "pain scale." Our goal is to provide the readers with a protocol of how to integrate some of the new tools described here with currently used mechanical somatosensory assays, while discussing the advantages and limitations of this new approach.
Collapse
Affiliation(s)
- Ishmail Abdus-Saboor
- Department of Biology, University of Pennsylvania, 3740 Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Wenqin Luo
- Department of Neuroscience, University of Pennsylvania, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA
| |
Collapse
|
16
|
Sisignano M, Gribbon P, Geisslinger G. Drug Repurposing to Target Neuroinflammation and Sensory Neuron-Dependent Pain. Drugs 2022; 82:357-373. [PMID: 35254645 PMCID: PMC8899787 DOI: 10.1007/s40265-022-01689-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
Abstract
Around 20% of the American population have chronic pain and estimates in other Western countries report similar numbers. This represents a major challenge for global health care systems. Additional problems for the treatment of chronic and persistent pain are the comparably low efficacy of existing therapies, the failure to translate effects observed in preclinical pain models to human patients and related setbacks in clinical trials from previous attempts to develop novel analgesics. Drug repurposing offers an alternative approach to identify novel analgesics as it can bypass various steps of classical drug development. In recent years, several approved drugs were attributed analgesic properties. Here, we review available data and discuss recent findings suggesting that the approved drugs minocycline, fingolimod, pioglitazone, nilotinib, telmisartan, and others, which were originally developed for the treatment of different pathologies, can have analgesic, antihyperalgesic, or neuroprotective effects in preclinical and clinical models of inflammatory or neuropathic pain. For our analysis, we subdivide the drugs into substances that can target neuroinflammation or substances that can act on peripheral sensory neurons, and highlight the proposed mechanisms. Finally, we discuss the merits and challenges of drug repurposing for the development of novel analgesics.
Collapse
Affiliation(s)
- Marco Sisignano
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany. .,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany. .,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, ScreeningPort, Schnackenburgallee 114, 22525, Hamburg, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- pharmazentrum frankfurt/ZAFES, Institute of Clinical Pharmacology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| |
Collapse
|
17
|
Iseppon F, Linley JE, Wood JN. Calcium imaging for analgesic drug discovery. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 11:100083. [PMID: 35079661 PMCID: PMC8777277 DOI: 10.1016/j.ynpai.2021.100083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022]
Abstract
Somatosensation and pain are complex phenomena involving a rangeofspecialised cell types forming different circuits within the peripheral and central nervous systems. In recent decades, advances in the investigation of these networks, as well as their function in sensation, resulted from the constant evolution of electrophysiology and imaging techniques to allow the observation of cellular activity at the population level both in vitro and in vivo. Genetically encoded indicators of neuronal activity, combined with recent advances in DNA engineering and modern microscopy, offer powerful tools to dissect and visualise the activity of specific neuronal subpopulations with high spatial and temporal resolution. In recent years various groups developed in vivo imaging techniques to image calcium transients in the dorsal root ganglia, the spinal cord and the brain of anesthetised and awake, behaving animals to address fundamental questions in both the physiology and pathophysiology of somatosensation and pain. This approach, besides giving unprecedented details on the circuitry of innocuous and painful sensation, can be a very powerful tool for pharmacological research, from the characterisation of new potential drugs to the discovery of new, druggable targets within specific neuronal subpopulations. Here we summarise recent developments in calcium imaging for pain research, discuss technical challenges and advances, and examine the potential positive impact of this technique in early preclinical phases of the analgesic drug discovery process.
Collapse
Affiliation(s)
- Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John E. Linley
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
18
|
Martins D, Veronese M, Turkheimer FE, Howard MA, Williams SCR, Dipasquale O. A candidate neuroimaging biomarker for detection of neurotransmission-related functional alterations and prediction of pharmacological analgesic response in chronic pain. Brain Commun 2021; 4:fcab302. [PMID: 35169702 PMCID: PMC8833258 DOI: 10.1093/braincomms/fcab302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/10/2021] [Accepted: 12/21/2021] [Indexed: 01/29/2023] Open
Abstract
Chronic pain is a world-wide clinical challenge. Response to analgesic treatment is limited and difficult to predict. Functional MRI has been suggested as a potential solution. However, while most analgesics target specific neurotransmission pathways, functional MRI-based biomarkers are not specific for any neurotransmitter system, limiting our understanding of how they might contribute to predict treatment response. Here, we sought to bridge this gap by applying Receptor-Enriched Analysis of Functional Connectivity by Targets to investigate whether neurotransmission-enriched functional connectivity mapping can provide insights into the brain mechanisms underlying chronic pain and inter-individual differences in analgesic response after a placebo or duloxetine. We performed secondary analyses of two openly available resting-state functional MRI data sets of 56 patients with chronic knee osteoarthritis pain who underwent pre-treatment brain scans in two clinical trials. Study 1 (n = 17) was a 2-week single-blinded placebo pill trial. Study 2 (n = 39) was a 3-month double-blinded randomized trial comparing placebo to duloxetine, a dual serotonin–noradrenaline reuptake inhibitor. Across two independent studies, we found that patients with chronic pain present alterations in the functional circuit related to the serotonin transporter, when compared with age-matched healthy controls. Placebo responders in Study 1 presented with higher pre-treatment functional connectivity enriched by the dopamine transporter compared to non-responders. Duloxetine responders presented with higher pre-treatment functional connectivity enriched by the serotonin and noradrenaline transporters when compared with non-responders. Neurotransmission-enriched functional connectivity mapping might hold promise as a new mechanistic-informed biomarker for functional brain alterations and prediction of response to pharmacological analgesia in chronic pain.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Federico E. Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Matthew A. Howard
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Steve C. R. Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
19
|
Marques F, Falquet L, Vandewyer E, Beets I, Glauser DA. Signaling via the FLP-14/FRPR-19 neuropeptide pathway sustains nociceptive response to repeated noxious stimuli in C. elegans. PLoS Genet 2021; 17:e1009880. [PMID: 34748554 PMCID: PMC8601619 DOI: 10.1371/journal.pgen.1009880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/18/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
In order to thrive in constantly changing environments, animals must adaptively respond to threatening events. Noxious stimuli are not only processed according to their absolute intensity, but also to their context. Adaptation processes can cause animals to habituate at different rates and degrees in response to permanent or repeated stimuli. Here, we used a forward genetic approach in Caenorhabditis elegans to identify a neuropeptidergic pathway, essential to prevent fast habituation and maintain robust withdrawal responses to repeated noxious stimuli. This pathway involves the FRPR-19A and FRPR-19B G-protein coupled receptor isoforms produced from the frpr-19 gene by alternative splicing. Loss or overexpression of each or both isoforms can impair withdrawal responses caused by the optogenetic activation of the polymodal FLP nociceptor neuron. Furthermore, we identified FLP-8 and FLP-14 as FRPR-19 ligands in vitro. flp-14, but not flp-8, was essential to promote withdrawal response and is part of the same genetic pathway as frpr-19 in vivo. Expression and cell-specific rescue analyses suggest that FRPR-19 acts both in the FLP nociceptive neurons and downstream interneurons, whereas FLP-14 acts from interneurons. Importantly, genetic impairment of the FLP-14/FRPR-19 pathway accelerated the habituation to repeated FLP-specific optogenetic activation, as well as to repeated noxious heat and harsh touch stimuli. Collectively, our data suggest that well-adjusted neuromodulation via the FLP-14/FRPR-19 pathway contributes to promote nociceptive signals in C. elegans and counteracts habituation processes that otherwise tend to rapidly reduce aversive responses to repeated noxious stimuli.
Collapse
Affiliation(s)
- Filipe Marques
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Elke Vandewyer
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Isabel Beets
- Neural Signaling and Circuit Plasticity Group, Department of Biology, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
20
|
Caudle RM, Smith MT, Romero-Sandoval EA. Editorial: Verification of Animal Pain Models by Reverse Translation. Front Pharmacol 2021; 12:778880. [PMID: 34690791 PMCID: PMC8531501 DOI: 10.3389/fphar.2021.778880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robert M Caudle
- Department of Oral and Maxillofacial Surgery, University of Florida, Gainesville, FL, United States
| | - Maree Therese Smith
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | | |
Collapse
|
21
|
Automated home-cage monitoring as a potential measure of sickness behaviors and pain-like behaviors in LPS-treated mice. PLoS One 2021; 16:e0256706. [PMID: 34449819 PMCID: PMC8396795 DOI: 10.1371/journal.pone.0256706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The use of endotoxin, such as lipopolysaccharide (LPS) as a model of sickness behavior, has attracted recent attention. To objectively investigate sickness behavior along with its pain-like behaviors in LPS-treated mice, the behavioral measurement requires accurate methods, which reflects clinical relevance. While reflexive pain response tests have been used for decades for pain assessment, its accuracy and clinical relevance remain problematic. Hence, we used automated home-cage monitoring LABORAS to evaluate spontaneous locomotive behaviors in LPS-induced mice. LPS-treated mice displayed sickness behaviors including pain-like behaviors in automated home-cage monitoring characterized by decreased mobile behaviors (climbing, locomotion, rearing) and increased immobility compared to that of the control group in both short- and long-term locomotive assessments. Here, in short-term measurement, both in the open-field test and automated home-cage monitoring, mice demonstrated impaired locomotive behaviors. We also assessed 24 h long-term locomotor activity in the home-cage system, which profiled the diurnal behaviors of LPS-stimulated mice. The results demonstrated significant behavioral impairment in LPS-stimulated mice compared to the control mice in both light and dark phases. However, the difference is more evident in the dark phase compared to the light phase owing to the nocturnal activity of mice. In addition, the administration of indomethacin as a pharmacological intervention improved sickness behaviors in the open-field test as well as automated home-cage monitoring, confirming that automated home-cage monitoring could be potentially useful in pharmacological screening. Together, our results demonstrate that automated home-cage monitoring could be a feasible alternative to conventional methods, such as the open-field test and combining several behavioral assessments may provide a better understanding of sickness behavior and pain-like behaviors in LPS-treated mice.
Collapse
|
22
|
Bagdas D, Sevdar G, Gul Z, Younis R, Cavun S, Tae HS, Ortells MO, Arias HR, Gurun MS. (E)-3-furan-2-yl-N-phenylacrylamide (PAM-4) decreases nociception and emotional manifestations of neuropathic pain in mice by α7 nicotinic acetylcholine receptor potentiation. Neurol Res 2021; 43:1056-1068. [PMID: 34281483 DOI: 10.1080/01616412.2021.1949684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Clinical intervention of pain is often accompanied by changes in affective behaviors, so both assays of affective and sensorial aspects of nociception play an important role in the development of novel analgesics. Although positive allosteric modulation (PAM) of α7 nicotinic acetylcholine receptors (nAChRs) has been recognized as a novel approach for the relief of sensorial aspects of pain, their effects on affective components of pain remain unclear. Therefore, we investigated whether PAM-4, a highly selective α7-nAChR PAM, attenuates inflammatory and neuropathic pain, as well as the concomitant depressive/anxiety comorbidities. The anti-nociceptive activity of PAM-4 was assessed in mice using the formalin test and chronic constriction injury (CCI)-induced neuropathic pain model. The anxiolytic- and antidepressant-like activity of PAM-4 was evaluated using the marble burying test and forced swimming test. Acute systemic administration of PAM-4 dose-dependently reversed formalin-induced paw licking behavior and CCI-induced mechanical allodynia without development of any motor impairment. PAM-4 reversed the decreased swimming time and number of buried marbles in CCI-treated mice, suggesting that this ligand attenuates chronic pain-induced depression-like behavior and anxiogenic-like effects. The effects of PAM-4 were inhibited by the α7-selective antagonist methyllycaconitine, indicating molecular mechanism mediated by α7-nAChRs. Indeed, electrophysiological recordings showed the PAM-4 enhances human α7 nAChRs with higher potency and efficacy compared to rat α7 nAChRs. These findings suggest that PAM-4 reduces both sensorial and affective behaviors induced by chronic pain in mice by α7-nAChR potentiation. PAM-4 deserves further investigations for the management of chronic painful conditions with comorbidities.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Psychiatry, School of Medicine, Yale University, New Haven, USA
| | - Gulce Sevdar
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Zulfiye Gul
- Department of Pharmacology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Rabha Younis
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Sinan Cavun
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, Australia
| | - Marcelo O Ortells
- Facultad de Medicina, Universidad de Morón, Morón, and CONICET, Moron, Argentina
| | - Hugo R Arias
- Department of Pharmacology and Physiology, Oklahoma State University College of Osteopathic Medicine, Tahlequah, OK, USA
| | - Mine Sibel Gurun
- Department of Pharmacology, Faculty of Medicine, Bursa Uludag University, Bursa, Turkey
| |
Collapse
|
23
|
Abstract
In this review, the state of the art for compounds affecting the endocannabinoid (eCB) system is described with a focus on the treatment of pain. Amongst directly acting CB receptor ligands, clinical experience with ∆9 -tetrahydracannabinol and medical cannabis in chronic non-cancer pain indicates that there are differences between the benefits perceived by patients and the at best modest effect seen in meta-analyses of randomized controlled trials. The reason for this difference is not known but may involve differences in the type of patients that are recruited, the study conditions that are chosen and the degree to which biases such as reporting bias are operative. Other directly acting CB receptor ligands such as biased agonists and allosteric receptor modulators have not yet reached the clinic. Amongst indirectly acting compounds targeting the enzymes responsible for the synthesis and catabolism of the eCBs anandamide and 2-arachidonoylglycerol, fatty acid amide hydrolase (FAAH) inhibitors have been investigated clinically but were per se not useful for the treatment of pain, although they may be useful for the treatment of post-traumatic stress disorder and cannabis use disorder. Dual-acting compounds targeting this enzyme and other targets such as cyclooxygenase-2 or transient potential vanilloid receptor 1 may be a way forward for the treatment of pain.
Collapse
Affiliation(s)
- C J Fowler
- From the, Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
24
|
Draxler P, Moen A, Galek K, Boghos A, Ramazanova D, Sandkühler J. Spontaneous, Voluntary, and Affective Behaviours in Rat Models of Pathological Pain. FRONTIERS IN PAIN RESEARCH 2021; 2:672711. [PMID: 35295455 PMCID: PMC8915731 DOI: 10.3389/fpain.2021.672711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
In pain patients affective and motivational reactions as well as impairment of daily life activities dominate the clinical picture. In contrast, many rodent pain models have been established on the basis of mechanical hypersensitivity testing. Up to today most rodent studies on pain still rely on reflexive withdrawal responses only. This discrepancy has likely contributed to the low predictive power of preclinical pain models for novel therapies. Here, we used a behavioural test array for rats to behaviourally evaluate five aetiologically distinct pain models consisting of inflammatory-, postsurgical-, cephalic-, neuropathic- and chemotherapy-induced pain. We assessed paralleling clinical expressions and comorbidities of chronic pain with an array of behavioural tests to assess anxiety, social interaction, distress, depression, and voluntary/spontaneous behaviours. Pharmacological treatment of the distinct pain conditions was performed with pathology-specific and clinically efficacious analgesics as gabapentin, sumatriptan, naproxen, and codeine. We found that rats differed in their manifestation of symptoms depending on the pain model and that pathology-specific analgesics also reduced the associated behavioural parameters. Based on all behavioural test performed, we screened for tests that can discriminate experimental groups on the basis of reflexive as well as non-sensory, affective parameters. Together, we propose a set of non-evoked behaviours with a comparable predictive power to mechanical threshold testing for each pain model.
Collapse
Affiliation(s)
- Peter Draxler
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Aurora Moen
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Karolina Galek
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Ani Boghos
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Dariga Ramazanova
- Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS) Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Jürgen Sandkühler
- Division of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Siracusa R, Monaco F, D’Amico R, Genovese T, Cordaro M, Interdonato L, Gugliandolo E, Peritore AF, Crupi R, Cuzzocrea S, Impellizzeri D, Fusco R, Di Paola R. Epigallocatechin-3-Gallate Modulates Postoperative Pain by Regulating Biochemical and Molecular Pathways. Int J Mol Sci 2021; 22:ijms22136879. [PMID: 34206850 PMCID: PMC8268037 DOI: 10.3390/ijms22136879] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 01/11/2023] Open
Abstract
Treating postoperative (PO) pain is a clinical challenge. Inadequate PO pain management can lead to worse outcomes, for example chronic post-surgical pain. Therefore, acquiring new information on the PO pain mechanism would increase the therapeutic options available. In this paper, we evaluated the role of a natural substance, epigallocatechin-3-gallate (EGCG), on pain and neuroinflammation induced by a surgical procedure in an animal model of PO pain. We performed an incision of the hind paw and EGCG was administered for five days. Mechanical allodynia, thermal hyperalgesia, and motor dysfunction were assessed 24 h, and three and five days after surgery. At the same time points, animals were sacrificed, and sera and lumbar spinal cord tissues were harvested for molecular analysis. EGCG administration significantly alleviated hyperalgesia and allodynia, and reduced motor disfunction. From the molecular point of view, EGCG reduced the activation of the WNT pathway, reducing WNT3a, cysteine-rich domain frizzled (FZ)1 and FZ8 expressions, and both cytosolic and nuclear β-catenin expression, and the noncanonical β-catenin–independent signaling pathways, reducing the activation of the NMDA receptor subtype NR2B (pNR2B), pPKC and cAMP response element-binding protein (pCREB) expressions at all time points. Additionally, EGCG reduced spinal astrocytes and microglia activation, cytokines overexpression and nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) pathway, downregulating inducible nitric oxide synthase (iNOS) activation, cyclooxygenase 2 (COX-2) expression, and prostaglandin E2 (PGE2) levels. Thus, EGCG administration managing the WNT/β-catenin signaling pathways modulates PO pain related neurochemical and inflammatory alterations.
Collapse
Affiliation(s)
- Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Francesco Monaco
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy; (F.M.); (M.C.)
| | - Ramona D’Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Tiziana Genovese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy; (F.M.); (M.C.)
| | - Livia Interdonato
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (E.G.); (R.C.)
| | - Alessio Filippo Peritore
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Rosalia Crupi
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy; (E.G.); (R.C.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
- Correspondence: ; Tel.: +39-090-676-5208
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy; (R.S.); (R.D.); (T.G.); (L.I.); (A.F.P.); (S.C.); (R.F.); (R.D.P.)
| |
Collapse
|
26
|
Nonsurgical mouse model of endometriosis-associated pain that responds to clinically active drugs. Pain 2021; 161:1321-1331. [PMID: 32132396 DOI: 10.1097/j.pain.0000000000001832] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endometriosis is an estrogen-dependent inflammatory disease that affects approximately 10% of women. Debilitating pelvic or abdominal pain is one of its major clinical features. Current animal models of endometriosis-associated pain require surgery either to implant tissue or to remove the ovaries. Moreover, existing models do not induce spontaneous pain, which is the primary symptom of patients with chronic pain, including endometriosis. A lack of models that accurately recapitulate the disease phenotype must contribute to the high failure rate of clinical trials for analgesic drugs directed at chronic pain, including those for endometriosis. We set out to establish a murine model of endometriosis-associated pain. Endometriosis was induced nonsurgically by injecting a dissociated uterine horn into a recipient mouse. The induced lesions exhibited histological features that resemble human lesions along with an increase in proinflammatory cytokines and recruitment of immune cells. We also observed the presence of calcitonin gene-related peptide-, TRPA1-, and TRPV1-expressing nerve fibers in the lesions. This model induced mechanical allodynia, spontaneous abdominal pain, and changes in thermal selection behavior that indicate discomfort. These behavioral changes were reduced by drugs used clinically for endometriosis, specifically letrozole (aromatase inhibitor) and danazol (androgen). Endometriosis also induced neuronal changes as evidenced by activation of the NF-κB signaling pathway in TRPA1- and TRPV1-expressing dorsal root ganglion neurons. In conclusion, we have established a model of endometriosis-associated pain that responds to clinically active drugs and can, therefore, be used to identify novel therapies.
Collapse
|
27
|
Effects of Curcumin and Its Different Formulations in Preclinical and Clinical Studies of Peripheral Neuropathic and Postoperative Pain: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22094666. [PMID: 33925121 PMCID: PMC8125634 DOI: 10.3390/ijms22094666] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/11/2022] Open
Abstract
Lesion or disease of the somatosensory system leads to the development of neuropathic pain. Peripheral neuropathic pain encompasses damage or injury of the peripheral nervous system. On the other hand, 10–15% of individuals suffer from acute postoperative pain followed by persistent pain after undergoing surgeries. Antidepressants, anticonvulsants, baclofen, and clonidine are used to treat peripheral neuropathy, whereas opioids are used to treat postoperative pain. The negative effects associated with these drugs emphasize the search for alternative therapeutics with better efficacy and fewer side effects. Curcumin, a polyphenol isolated from the roots of Curcuma longa, possesses antibacterial, antioxidant, and anti-inflammatory properties. Furthermore, the low bioavailability and fast metabolism of curcumin have led to the advent of various curcumin formulations. The present review provides a comprehensive analysis on the effects of curcumin and its formulations in preclinical and clinical studies of neuropathic and postoperative pain. Based on the positive outcomes from both preclinical and clinical studies, curcumin holds the promise of mitigating or preventing neuropathic and postoperative pain conditions. However, more clinical studies with improved curcumin formulations are required to involve its use as adjuvant to neuropathic and postoperative drugs.
Collapse
|
28
|
Nicotinamide adenine dinucleotide phosphate oxidase 2-derived reactive oxygen species contribute to long-term potentiation of C-fiber-evoked field potentials in spinal dorsal horn and persistent mirror-image pain following high-frequency stimulus of the sciatic nerve. Pain 2021; 161:758-772. [PMID: 32195784 DOI: 10.1097/j.pain.0000000000001761] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
High-frequency stimulation (HFS) of the sciatic nerve has been reported to produce long-term potentiation (LTP) and long-lasting pain hypersensitivity in rats. However, the central underlying mechanism remains unclear. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) belongs to a group of electron-transporting transmembrane enzymes that produce reactive oxygen species (ROS). Here, we found that NOX2 was upregulated in the lumbar spinal dorsal horn after HFS of the left sciatic nerve, which induced bilateral pain and spinal LTP in both male and female rats. Blocking NOX2 with blocking peptide or shRNA prevented the development of bilateral mechanical allodynia, the induction of spinal LTP, and the phosphorylation of N-methyl-d-aspartate (NMDA) receptor 2B (GluN2B) and nuclear factor kappa-B (NF-κB) p65 after HFS. Moreover, NOX2 shRNA reduced the frequency and amplitude of both spontaneous excitatory postsynaptic currents and miniature excitatory postsynaptic currents in laminar II neurons. Furthermore, 8-hydroxyguanine (8-OHG), an oxidative stress marker, was increased in the spinal dorsal horn. Spinal application of ROS scavenger, Phenyl-N-tert-butylnitrone (PBN), depressed the already established spinal LTP. Spinal application of H2O2, one ROS, induced LTP and bilateral mechanical allodynia, increased the frequency and amplitude of spontaneous excitatory postsynaptic currents in laminar II neurons, and phosphorylated GluN2B and p65 in the dorsal horn. This study provided electrophysiological and behavioral evidence that NOX2-derived ROS in the spinal cord contributed to persistent mirror-image pain by enhancing the synaptic transmission, which was mediated by increasing presynaptic glutamate release and activation of NMDA receptor and NF-κB in the spinal dorsal horn.
Collapse
|
29
|
Cho C, Deol HK, Martin LJ. Bridging the Translational Divide in Pain Research: Biological, Psychological and Social Considerations. Front Pharmacol 2021; 12:603186. [PMID: 33935700 PMCID: PMC8082136 DOI: 10.3389/fphar.2021.603186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 02/22/2021] [Indexed: 12/29/2022] Open
Abstract
A gap exists between translating basic science research into effective pain therapies in humans. While preclinical pain research has primarily used animal models to understand biological processes, a lesser focus has been toward using animal models to fully consider other components of the pain experience, such as psychological and social influences. Herein, we provide an overview of translational studies within pain research by breaking them down into purely biological, psychological and social influences using a framework derived from the biopsychosocial model. We draw from a wide landscape of studies to illustrate that the pain experience is highly intricate, and every attempt must be made to address its multiple components and interactors to aid in fully understanding its complexity. We highlight our work where we have developed animal models to assess the cognitive and social effects on pain modulation while conducting parallel experiments in people that provide proof-of-importance for human pain modulation. In some instances, human pain research has sparked the development of novel animal models, with these animal models used to better understand the complexity of phenomena considered to be uniquely human such as placebo responses and empathy.
Collapse
Affiliation(s)
- Chulmin Cho
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Harashdeep K Deol
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Loren J Martin
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| |
Collapse
|
30
|
Liao YH, Wang B, Chen MX, Liu Y, Ao LJ. LIFU Alleviates Neuropathic Pain by Improving the KCC 2 Expression and Inhibiting the CaMKIV-KCC 2 Pathway in the L4-L5 Section of the Spinal Cord. Neural Plast 2021; 2021:6659668. [PMID: 33953740 PMCID: PMC8057881 DOI: 10.1155/2021/6659668] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/13/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
Effective treatment remains lacking for neuropathic pain (NP), a type of intractable pain. Low-intensity focused ultrasound (LIFU), a noninvasive, cutting-edge neuromodulation technique, can effectively enhance inhibition of the central nervous system (CNS) and reduce neuronal excitability. We investigated the effect of LIFU on NP and on the expression of potassium chloride cotransporter 2 (KCC2) in the spinal cords of rats with peripheral nerve injury (PNI) in the lumbar 4-lumbar 5 (L4-L5) section. In this study, rats received PNI surgery on their right lower legs followed by LIFU stimulation of the L4-L5 section of the spinal cord for 4 weeks, starting 3 days after surgery. We used the 50% paw withdraw threshold (PWT50) to evaluate mechanical allodynia. Western blotting (WB) and immunofluorescence (IF) were used to calculate the expression of phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2), calcium/calmodulin-dependent protein kinase type IV (CaMKIV), phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB), and KCC2 in the L4-L5 portion of the spinal cord after the last behavioral tests. We found that PWT50 decreased (P < 0.05) 3 days post-PNI surgery in the LIFU- and LIFU+ groups and increased (P < 0.05) after 4 weeks of LIFU stimulation. The expression of p-CREB and CaMKIV decreased (P < 0.05) and that of KCC2 increased (P < 0.05) after 4 weeks of LIFU stimulation, but that of p-ERK1/2 (P > 0.05) was unaffected. Our study showed that LIFU could effectively alleviate NP behavior in rats with PNI by increasing the expression of KCC2 on spinal dorsal corner neurons. A possible explanation is that LIFU could inhibit the activation of the CaMKIV-KCC2 pathway.
Collapse
Affiliation(s)
- Ye-Hui Liao
- School of Rehabilitation, Kunming Medical University, Kunming, 650500 Yunnan Province, China
| | - Bin Wang
- School of Rehabilitation, Kunming Medical University, Kunming, 650500 Yunnan Province, China
| | - Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, 650500 Yunnan Province, China
| | - Yao Liu
- School of Rehabilitation, Kunming Medical University, Kunming, 650500 Yunnan Province, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, 650500 Yunnan Province, China
| |
Collapse
|
31
|
Single cell transcriptomics of primate sensory neurons identifies cell types associated with chronic pain. Nat Commun 2021; 12:1510. [PMID: 33686078 PMCID: PMC7940623 DOI: 10.1038/s41467-021-21725-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/08/2021] [Indexed: 01/24/2023] Open
Abstract
Distinct types of dorsal root ganglion sensory neurons may have unique contributions to chronic pain. Identification of primate sensory neuron types is critical for understanding the cellular origin and heritability of chronic pain. However, molecular insights into the primate sensory neurons are missing. Here we classify non-human primate dorsal root ganglion sensory neurons based on their transcriptome and map human pain heritability to neuronal types. First, we identified cell correlates between two major datasets for mouse sensory neuron types. Machine learning exposes an overall cross-species conservation of somatosensory neurons between primate and mouse, although with differences at individual gene level, highlighting the importance of primate data for clinical translation. We map genomic loci associated with chronic pain in human onto primate sensory neuron types to identify the cellular origin of chronic pain. Genome-wide associations for chronic pain converge on two different neuronal types distributed between pain disorders that display different genetic susceptibilities, suggesting both unique and shared mechanisms between different pain conditions. The contribution of distinct types of dorsal root ganglion neurons to chronic pain is unclear. Here, the authors molecularly profile non-human primate sensory neurons and show that genome-wide associations converge on two neuronal types with different genetic susceptibilities for chronic pain.
Collapse
|
32
|
A new hypertonic saline assay for analgesic screening in mice: effects of animal strain, sex, and diurnal phase. Can J Anaesth 2021; 68:672-682. [PMID: 33598887 DOI: 10.1007/s12630-021-01923-5] [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: 08/11/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 10/22/2022] Open
Abstract
PURPOSE There exists a pressing need for the identification of novel analgesics. We recently reported on a new preclinical assay for rapid analgesic screening based on intraplantar (i.pl.) injection of 10% hypertonic saline (HS) in female outbred (CD-1) mice. Herein, we characterized the HS assay's performance in inbred (C57BL/6) mice, sensitivity to sex differences, and effects of diurnal rhythm phase. METHODS In randomized, controlled, blinded in vivo animal experiments, we studied nociceptive responses induced by i.pl. HS in C57BL/6 (vs CD-1) mice of both sexes (n = 240) and determined diurnal rhythm phase effects in female animals. We established the HS assay's sensitivity to morphine by constructing dose-response curves and calculating half-maximal inhibitory doses (ID50s). RESULTS The injection of i.pl. HS produced nociceptive (licking and biting) responses in all C57BL/6 mice tested. In both C57BL/6 and CD-1 mice, the mean (95% confidence interval [CI]) response magnitudes were greater in females vs males (C57BL/6: 87 sec [64 to 110] vs 45 sec [29 to 61]; difference in means, 42 sec; 95% CI, 17 to 68; P < 0.001; n = 10/group; CD-1: 110 sec [95 to 126] vs 53 sec [32 to 74]; difference in means, 57 sec; 95% CI, 34 to 79; P < 0.001; n = 10/group). The mean (95% CI) nociceptive responses were greater at 24:00 hr than at 12:00 hr in C57BL/6 mice (64 sec [40 to 88] vs 37 sec [24 to 51]; difference in means, 27 sec; 95% CI, 7 to 47; P = 0.007; n = 10/group), but not in CD-1 mice (P = 0.97). Intravenous morphine dose-dependently attenuated nociceptive responses of both C57BL/6 and CD-1 mice (ID50, 0.6 and 2.5 mg·kg-1, respectively; P = 0.41). CONCLUSION These findings in inbred and outbred mice solidify the utility of the HS assay as an effective, rapid, robust, and versatile preclinical tool for analgesic screening.
Collapse
|
33
|
Shen H, Gardner AM, Vyas J, Ishida R, Tawfik VL. Modeling Complex Orthopedic Trauma in Rodents: Bone, Muscle and Nerve Injury and Healing. Front Pharmacol 2021; 11:620485. [PMID: 33597884 PMCID: PMC7882733 DOI: 10.3389/fphar.2020.620485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Orthopedic injury can occur from a variety of causes including motor vehicle collision, battlefield injuries or even falls from standing. Persistent limb pain is common after orthopedic injury or surgery and presents a unique challenge, as the initiating event may result in polytrauma to bone, muscle, and peripheral nerves. It is imperative that we understand the tissue-specific and multicellular response to this unique type of injury in order to best develop targeted treatments that improve healing and regeneration. In this Mini Review we will first discuss current rodent models of orthopedic trauma/complex orthotrauma. In the second section, we will focus on bone-specific outcomes including imaging modalities, biomechanical testing and immunostaining for markers of bone healing/turnover. In the third section, we will discuss muscle-related pathology including outcome measures of fibrosis, muscle regeneration and tensile strength measurements. In the fourth section, we will discuss nervous system-related pathology including outcome measures of pain-like responses, both reflexive and non-reflexive. In all sections we will consider parallels between preclinical outcome measures and the functional and mechanistic findings of the human condition.
Collapse
Affiliation(s)
- Huaishuang Shen
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Aysha M Gardner
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Juhee Vyas
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Ryosuke Ishida
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Department of Anesthesiology, Shimane University, Shimane, Japan
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
34
|
Bagdas D, Gul Z, Meade JA, Cam B, Cinkilic N, Gurun MS. Pharmacologic Overview of Chlorogenic Acid and its Metabolites in Chronic Pain and Inflammation. Curr Neuropharmacol 2020; 18:216-228. [PMID: 31631820 PMCID: PMC7327949 DOI: 10.2174/1570159x17666191021111809] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/03/2019] [Accepted: 10/16/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Natural phenolic compounds in medicinal herbs and dietary plants are antioxidants which play therapeutic or preventive roles in different pathological situations, such as oxidative stress and inflammation. One of the most studied phenolic compounds in the last decade is chlorogenic acid (CGA), which is a potent antioxidant found in certain foods and drinks. OBJECTIVE This review focuses on the anti-inflammatory and antinociceptive bioactivities of CGA, and the putative mechanisms of action are described. Ethnopharmacological reports related to these bioactivities are also reviewed. MATERIALS AND METHODS An electronic literature search was conducted by authors up to October 2019. Original articles were selected. RESULTS CGA has been shown to reduce inflammation and modulate inflammatory and neuropathic pain in animal models. CONCLUSION The consensus of the literature search was that systemic CGA may facilitate pain management via bolstering antioxidant defenses against inflammatory insults.
Collapse
Affiliation(s)
- Deniz Bagdas
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, United States.,Yale Tobacco Center of Regulatory Science, Yale University, New Haven, CT, United States
| | - Zulfiye Gul
- Department of Pharmacology, Faculty of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Julie A Meade
- Department of Pharmacology & Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Betul Cam
- Department of Physiology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Nilufer Cinkilic
- Department of Biology, Faculty of Science and Arts, Uludag University, Bursa, Turkey
| | - Mine Sibel Gurun
- Department of Pharmacology, Faculty of Medicine, Uludag University, Bursa, Turkey
| |
Collapse
|
35
|
Shen M, Chen M, Liang T, Wang S, Xue Y, Bertz R, Xie XQ, Feng Z. Pain Chemogenomics Knowledgebase (Pain-CKB) for Systems Pharmacology Target Mapping and Physiologically Based Pharmacokinetic Modeling Investigation of Opioid Drug-Drug Interactions. ACS Chem Neurosci 2020; 11:3245-3258. [PMID: 32966035 DOI: 10.1021/acschemneuro.0c00372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
More than 50 million adults in America suffer from chronic pain. Opioids are commonly prescribed for their effectiveness in relieving many types of pain. However, excessive prescribing of opioids can lead to abuse, addiction, and death. Non-steroidal anti-inflammatory drugs (NSAIDs), another major class of analgesic, also have many problematic side effects including headache, dizziness, vomiting, diarrhea, nausea, constipation, reduced appetite, and drowsiness. There is an urgent need for the understanding of molecular mechanisms that underlie drug abuse and addiction to aid in the design of new preventive or therapeutic agents for pain management. To facilitate pain related small-molecule signaling pathway studies and the prediction of potential therapeutic target(s) for the treatment of pain, we have constructed a comprehensive platform of a pain domain-specific chemogenomics knowledgebase (Pain-CKB) with integrated data mining computing tools. Our new computing platform describes the chemical molecules, genes, proteins, and signaling pathways involved in pain regulation. Pain-CKB is implemented with a friendly user interface for the prediction of the relevant protein targets and analysis and visualization of the outputs, including HTDocking, TargetHunter, BBB predictor, and Spider Plot. Combining these with other novel tools, we performed three case studies to systematically demonstrate how further studies can be conducted based on the data generated from Pain-CKB and its algorithms and tools. First, systems pharmacology target mapping was carried out for four FDA approved analgesics in order to identify the known target and predict off-target interactions. Subsequently, the target mapping outcomes were applied to build physiologically based pharmacokinetic (PBPK) models for acetaminophen and fentanyl to explore the drug-drug interaction (DDI) between this pair of drugs. Finally, pharmaco-analytics was conducted to explore the detailed interaction pattern of acetaminophen reactive metabolite and its hepatotoxicity target, thioredoxin reductase.
Collapse
Affiliation(s)
- Mingzhe Shen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Maozi Chen
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Tianjian Liang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Siyi Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Ying Xue
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Richard Bertz
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, and Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
36
|
The Protective Effects of Pre- and Post-Administration of Micronized Palmitoylethanolamide Formulation on Postoperative Pain in Rats. Int J Mol Sci 2020; 21:ijms21207700. [PMID: 33080989 PMCID: PMC7589788 DOI: 10.3390/ijms21207700] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Postoperative pain (PO) is a common form of acute pain. Inadequate PO treatment is an important health problem, as it leads to worse outcomes, such as chronic post-surgical pain. Therefore, it is necessary to acquire new knowledge on PO mechanisms to develop therapeutic options with greater efficacy than those available today and to lower the risk of adverse effects. For this reason, we evaluated the ability of micronized palmitoylethanolamide (PEA-m) to resolve the pain and inflammatory processes activated after incision of the hind paw in an animal model of PO. Methods: The animals were subjected to surgical paw incision and randomized into different groups. PEA-m was administered orally at 10 mg/kg at different time points before or after incision. Results: Our research demonstrated that the pre- and post-treatment with PEA-m reduced the activation of mast cells at the incision site and the expression of its algogenic mediator nerve growth factor (NGF) in the lumbar spinal cord. Furthermore, again at the spinal level, it was able to decrease the activation of phospho-extracellular signal-regulated kinases (p-ERK), ionized calcium binding adaptor molecule 1 (Iba1), glial fibrillary acidic protein (GFAP), and the expression of brain-derived neurotrophic factor (BDNF). PEA-m also reduced the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) spinal pathway, showing a protective effect in a rat model of PO. Conclusion: The results obtained reinforce the idea that PEA-m may be a potential treatment for the control of pain and inflammatory processes associated with PO. In addition, pre- and post-treatment with PEA-m is more effective than treatment alone after the surgery and this limits the time of taking the compound and the abuse of analgesics.
Collapse
|
37
|
Jones JM, Foster W, Twomey CR, Burdge J, Ahmed OM, Pereira TD, Wojick JA, Corder G, Plotkin JB, Abdus-Saboor I. A machine-vision approach for automated pain measurement at millisecond timescales. eLife 2020; 9:e57258. [PMID: 32758355 PMCID: PMC7434442 DOI: 10.7554/elife.57258] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/05/2020] [Indexed: 12/28/2022] Open
Abstract
Objective and automatic measurement of pain in mice remains a barrier for discovery in neuroscience. Here, we capture paw kinematics during pain behavior in mice with high-speed videography and automated paw tracking with machine and deep learning approaches. Our statistical software platform, PAWS (Pain Assessment at Withdrawal Speeds), uses a univariate projection of paw position over time to automatically quantify seven behavioral features that are combined into a single, univariate pain score. Automated paw tracking combined with PAWS reveals a behaviorally divergent mouse strain that displays hypersensitivity to mechanical stimuli. To demonstrate the efficacy of PAWS for detecting spinally versus centrally mediated behavioral responses, we chemogenetically activated nociceptive neurons in the amygdala, which further separated the pain-related behavioral features and the resulting pain score. Taken together, this automated pain quantification approach will increase objectivity in collecting rigorous behavioral data, and it is compatible with other neural circuit dissection tools for determining the mouse pain state.
Collapse
Affiliation(s)
- Jessica M Jones
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - William Foster
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Colin R Twomey
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Justin Burdge
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Osama M Ahmed
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Talmo D Pereira
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Jessica A Wojick
- Departments of Psychiatry and Neuroscience, University of PennsylvaniaPhiladelphiaUnited States
| | - Gregory Corder
- Departments of Psychiatry and Neuroscience, University of PennsylvaniaPhiladelphiaUnited States
| | - Joshua B Plotkin
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | | |
Collapse
|
38
|
Central IRAK-4 kinase inhibition for the treatment of pain following nerve injury in rats. Brain Behav Immun 2020; 88:781-790. [PMID: 32439472 DOI: 10.1016/j.bbi.2020.05.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/10/2020] [Accepted: 05/10/2020] [Indexed: 11/20/2022] Open
Abstract
There is ample evidence for the role of the immune system in developing chronic pain following peripheral nerve injury. Especially Toll-like receptors (TLRs) and their associated signaling components and pro-inflammatory cytokines such as IL-1β, induced after injury, are involved in nociceptive processes and believed to contribute to the manifestation of chronic neuropathic pain states. Whereas the inhibition of the kinase function of IRAK-4, a central kinase downstream of TLRs and IL-1 receptors (IL-1Rs), seems efficacious in various chronic inflammatory and autoimmune models, it's role in regulating chronic neuropathic pain remained elusive to date. Here, we examined whether pharmacological inhibition of IRAK-4 kinase activity using PF-06650833 and BMS-986147, two clinical-stage kinase inhibitors, is effective for controlling persistent pain following nerve injury. Both inhibitors potently inhibited TLR-triggered cytokine release in human peripheral blood mononuclear cell (PBMC) as well as human and rat whole blood cultures. BMS-986147 showing favorable pharmacokinetic (PK) properties, significantly inhibited R848-triggered plasma TNF levels in a rat in vivo cytokine release model after single oral dosing. However, BMS-986147 dose dependently reversed cold allodynia in a rat chronic constriction injury (CCI) model following intrathecal administration only, supporting the notion that central neuro-immune modulation is beneficial for treating chronic neuropathic pain. Although both inhibitors were efficacious in inhibiting IL-1β- or TLR-triggered cytokine release in rat dorsal root ganglion cultures, only partial efficacy was reached in IL-1β-stimulated human glial cultures indicating that inhibiting IRAK-4́'s kinase function might be partially dispensable for human IL-1β driven neuroinflammation. Overall, our data demonstrate that IRAK-4 inhibitors could provide therapeutic benefit in chronic pain following nerve injury, and the central driver for efficacy in the neuropathic pain model as well as potential side effects of centrally available IRAK-4 inhibitors warrant further investigation to develop effective analgesia for patients in high unmet medical need.
Collapse
|
39
|
Hughes DI, Todd AJ. Central Nervous System Targets: Inhibitory Interneurons in the Spinal Cord. Neurotherapeutics 2020; 17:874-885. [PMID: 33029722 PMCID: PMC7641291 DOI: 10.1007/s13311-020-00936-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Pain is a percept of critical importance to our daily survival. In most cases, it serves both an adaptive function by helping us respond appropriately in a potentially hostile environment and also a protective role by alerting us to tissue damage. Normally, it is evoked by the activation of peripheral nociceptive nerve endings and the subsequent relay of information to distinct cortical and sub-cortical regions, but under pathological conditions that result in chronic pain, it can become spontaneous. Given that one in three chronic pain patients do not respond to the treatments currently available, the need for more effective analgesics is evident. Two principal obstacles to the development of novel analgesic therapies are our limited understanding of how neuronal circuits that comprise these pain pathways transmit and modulate sensory information under normal circumstances and how these circuits change under pathological conditions leading to chronic pain states. In this review, we focus on the role of inhibitory interneurons in setting pain thresholds and, in particular, how disinhibition in the spinal dorsal horn can lead to aberrant sensory processing associated with chronic pain states.
Collapse
Affiliation(s)
- David I Hughes
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland.
| | - Andrew J Todd
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland
| |
Collapse
|
40
|
Fried NT, Chamessian A, Zylka MJ, Abdus-Saboor I. Improving pain assessment in mice and rats with advanced videography and computational approaches. Pain 2020; 161:1420-1424. [PMID: 32102021 PMCID: PMC7302333 DOI: 10.1097/j.pain.0000000000001843] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/30/2020] [Accepted: 02/13/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Nathan T. Fried
- Department of Biology, Rutgers University-Camden, Camden, NJ, United States
| | - Alexander Chamessian
- Departments of Neurology and
- Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark J. Zylka
- Department of Cell Biology and Physiology, UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ishmail Abdus-Saboor
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
41
|
Fattori V, Rasquel-Oliveira FS, Artero NA, Ferraz CR, Borghi SM, Casagrande R, Verri WA. Diosmin Treats Lipopolysaccharide-Induced Inflammatory Pain and Peritonitis by Blocking NF-κB Activation in Mice. JOURNAL OF NATURAL PRODUCTS 2020; 83:1018-1026. [PMID: 32083866 DOI: 10.1021/acs.jnatprod.9b00887] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Gram-negative bacterial infections induce inflammation and pain. Lipopolysaccharide (LPS) is a pathogen-associated molecular pattern and the major constituent of Gram-negative bacterial cell walls. Diosmin is a citrus flavonoid with antioxidant and anti-inflammatory activities. Here we investigated the efficacy of diosmin in a nonsterile model of inflammatory pain and peritonitis induced by LPS. Diosmin reduced in a dose-dependent manner LPS-induced inflammatory mechanical hyperalgesia, thermal hyperalgesia, and neutrophil recruitment to the paw (myeloperoxidase activity). Diosmin also normalized changes in paw weight distribution assessed by static weight bearing as a nonreflexive method of pain measurement. Moreover, treatment with diosmin inhibited LPS-induced peritonitis as observed by a reduction of leukocyte recruitment and oxidative stress. Diosmin reduced LPS-induced total ROS production (DCFDA assay) and superoxide anion production (NBT assay and NBT-positive cells). We also observed a reduction of LPS-induced oxidative stress and cytokine production (IL-1β, TNF-α, and IL-6) in the paw. Furthermore, we demonstrated that diosmin inhibited LPS-induced NF-κB activation in peritoneal exudate. Thus, we demonstrated, using a model of nonsterile inflammation induced by LPS, that diosmin is a promising molecule for the treatment of inflammation and pain.
Collapse
Affiliation(s)
- Victor Fattori
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Fernanda S Rasquel-Oliveira
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Nayara A Artero
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Camila R Ferraz
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Sergio M Borghi
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
- Center for Research in Health Sciences, University of Northern Paraná, Londrina 86057-970, Paraná, Brazil
| | - Rubia Casagrande
- Departament of Pharmaceutical Sciences, Center of Health Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| | - Waldiceu A Verri
- Departament of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, Paraná, Brazil
| |
Collapse
|
42
|
Ma X, Wang H, Song T, Wang W, Zhang Z. lncRNA MALAT1 contributes to neuropathic pain development through regulating miR-129-5p/HMGB1 axis in a rat model of chronic constriction injury. Int J Neurosci 2020; 130:1215-1224. [PMID: 32065547 DOI: 10.1080/00207454.2020.1731508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Backgrounds: Mounting studies pay attention to the functional roles of long non-coding RNAs (lncRNAs) in many human diseases including neuropathic pain. LncRNA MALAT1 has been indicated to serve as a critical mediator in neuropathic pain with unclear mechanisms. The present study aims to explore the functional roles of MALAT1 in neuropathic pain progression and the related mechanisms.Methods: Bilateral sciatic nerves were ligated to induce chronic constriction injury (CCI) in order to establish the neuropathic pain rat model followed by behavioral tests, RT-qPCR, Western blotting, and ELISA. Dual luciferase activity assay was performed to determine the binding effect between MALAT1 or HMGB1 and miR-129-5p.Results: The mRNA levels of MALAT1 were significantly enhanced in CCI rats. MALAT1 inhibition repressed the development of neuropathic pain and neuroinflammation. Additionally, miR-129-5p was decreased and HMGB1 was increased, both of which could be rectified by MALAT1 inhibition. Meanwhile, MALAT1 targeted miR-129-5p/HMGB1 axis. Finally, miR-129-5p suppression attenuated the inhibitory effect of MALAT1 inhibition on neuropathic pain and neuroinflammation development in CCI rats.Conclusion: The present study demonstrates that MALAT1 might modulate neuropathic pain via targeting miR-129-5p/HMGB1 axis. These findings may lead to a promising and efficacious clinical approach for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Xiaojing Ma
- Department of Anesthesiology, The First Hospital of Shijiazhuang, Shijiazhuang, China
| | - Hong Wang
- Department of Anesthesiology, The First Hospital of Shijiazhuang, Shijiazhuang, China
| | - Tieying Song
- Department of Anesthesiology, The First Hospital of Shijiazhuang, Shijiazhuang, China
| | - Wenli Wang
- Department of Gynaecology, Maternal and Child Health Care Hospital of Shijiazhuang, Shijiazhuang, China
| | - Zaiwang Zhang
- Department of Anesthesiology, The Bethune International Peace Hospital of P.L.A, Shijiazhuang, China
| |
Collapse
|
43
|
Pahapill PA, Chen G, Arocho-Quinones EV, Nencka AS, Li SJ. Functional connectivity and structural analysis of trial spinal cord stimulation responders in failed back surgery syndrome. PLoS One 2020; 15:e0228306. [PMID: 32074111 PMCID: PMC7029839 DOI: 10.1371/journal.pone.0228306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 01/13/2020] [Indexed: 01/02/2023] Open
Abstract
Background Chronic pain has been associated with alterations in brain structure and function that appear dependent on pain phenotype. Functional connectivity (FC) data on chronic back pain (CBP) is limited and based on heterogeneous pain populations. We hypothesize that failed back surgery syndrome (FBSS) patients being considered for spinal cord stimulation (SCS) therapy have altered resting state (RS) FC cross-network patterns that 1) specifically involve emotion and reward/aversion functions and 2) are related to pain scores. Methods RS functional MRI (fMRI) scans were obtained for 10 FBSS patients who are being considered for but who have not yet undergone implantation of a permanent SCS device and 12 healthy age-matched controls. Seven RS networks were analyzed including the striatum (STM). The Wilcoxon signed-rank test evaluated differences in cross-network FC strength (FCS). Differences in periaqueductal grey (PAG) FC were assessed with seed-based analysis. Results Cross-network FCS was decreased (p<0.05) between the STM and all other networks in these FBSS patients. There was a negative linear relationship (R2 = 0.76, p<0.0022) between STMFCS index and pain scores. The PAG showed decreased FC with network elements and amygdala but increased FC with the sensorimotor cortex and cingulate gyrus. Conclusions Decreased FC between STM and other RS networks in FBSS has not been previously reported. This STMFCS index may represent a more objective measure of chronic pain specific to FBSS which may help guide patient selection for SCS and subsequent management.
Collapse
Affiliation(s)
- Peter A. Pahapill
- Department of Neurosurgery, U.S. Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Guangyu Chen
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Elsa V. Arocho-Quinones
- Department of Neurosurgery, U.S. Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, United States of America
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
- * E-mail:
| | - Andrew S. Nencka
- Center for Imaging Research, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Shi-Jiang Li
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
44
|
Woolf CJ. Capturing Novel Non-opioid Pain Targets. Biol Psychiatry 2020; 87:74-81. [PMID: 31399256 PMCID: PMC6898770 DOI: 10.1016/j.biopsych.2019.06.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/29/2022]
Abstract
The relatively high efficacy of opioids, which have associated risks of addiction, tolerance, and dependence, for the management of acute and terminal pain has been a major driver of the opioid crisis, together with the availability, overprescription, and diversion of these drugs. Eliminating opioids without an effective replacement is, however, no solution, as it substitutes one major problem with another. To deal successfully with the opioid crisis, we need to discover novel analgesics whose mechanisms do not involve the mu opioid receptor but that have high analgesic potency and low risk of adverse effects, particularly no abuse liability. The question is how to achieve this. There are several necessary elements; first, we need to understand the nature of pain and the mechanisms responsible for it, and second, we need to adopt novel and unbiased approaches to the identification and validation of pain targets.
Collapse
Affiliation(s)
- Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
45
|
Haight ES, Forman TE, Cordonnier SA, James ML, Tawfik VL. Microglial Modulation as a Target for Chronic Pain: From the Bench to the Bedside and Back. Anesth Analg 2019; 128:737-746. [PMID: 30883419 DOI: 10.1213/ane.0000000000004033] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
With a widespread opioid epidemic and profound biopsychosocial implications, chronic pain is a multifaceted public health issue requiring urgent attention. The treatment of chronic pain is particularly important to anesthesiologists given our unique role as perioperative physicians and pain medicine specialists. The present review details the recent shift from a neuronal theory of chronic pain to one that includes complex neuron-glia interactions. In particular, we highlight microglia, the myeloid-lineage cells of the central nervous system, as initiators of a postinjury neuroimmune response that contributes to the acute to chronic pain transition. We discuss ever-advancing preclinical studies, wherein significant success has been made through pharmacologic and genetic modulation of microglia, and we emphasize where these approaches have made the transition to the clinical realm. Furthermore, we highlight the most current, novel efforts to visualize glial activation in vivo using positron emission tomography and improve the diagnosis of chronic pain through radiotracer binding of specific targets, like the 18 kDa translocator protein in microglia and myeloid-lineage cells. Our rapidly advancing knowledge about microglia and their involvement in pain suggests that the era of glial-targeted therapeutics is just beginning so long as we refocus our attention on optimizing preclinical studies using a clinically informed approach, before translation.
Collapse
Affiliation(s)
- Elena S Haight
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| | - Thomas E Forman
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California.,University of Colorado School of Medicine, Denver, Colorado
| | - Stephanie A Cordonnier
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California.,College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio
| | - Michelle L James
- Departments of Radiology and Neurology, Stanford University School of Medicine, Stanford, California
| | - Vivianne L Tawfik
- From the Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
46
|
Bán EG, Brassai A, Vizi ES. The role of the endogenous neurotransmitters associated with neuropathic pain and in the opioid crisis: The innate pain-relieving system. Brain Res Bull 2019; 155:129-136. [PMID: 31816407 DOI: 10.1016/j.brainresbull.2019.12.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/22/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022]
Abstract
Neuropathic pain is a chronic pain caused by central and peripheral nerve injury, long-term diabetes or treatment with chemotherapy drugs, and it is dissimilar to other chronic pain conditions. Chronic pain usually seriously affects the quality of life, and its drug treatment may result in increased costs of social and medical care. As in the USA and Canada, in Europe, the demand for pain-relieving medicines used in chronic pain has also significantly increased, but most European countries are not experiencing an opioid crisis. In this review, the role of various endogenous transmitters (noradrenaline, dopamine, serotonin, met- and leu-enkephalins, β-endorphin, dynorphins, cannabinoids, ATP) and various receptors (α2, μ, etc.) in the innate pain-relieving system will be discussed. Furthermore, the modulation of pain processing pathways by transmitters, focusing on neuropathic pain and the role of the sympathetic nervous system in the side effects of excessive opioid treatment, will be explained.
Collapse
Affiliation(s)
- E Gy Bán
- Dept. ME1, Faculty of Medicine in English, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology of Târgu-Mureș, Marosvásárhely, Romania
| | - A Brassai
- Dept. ME1, Faculty of Medicine in English, "George Emil Palade" University of Medicine, Pharmacy, Science and Technology of Târgu-Mureș, Marosvásárhely, Romania
| | - E S Vizi
- Institute of Experimental Medicine, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
47
|
Lee KY, Ratté S, Prescott SA. Excitatory neurons are more disinhibited than inhibitory neurons by chloride dysregulation in the spinal dorsal horn. eLife 2019; 8:e49753. [PMID: 31742556 PMCID: PMC6887484 DOI: 10.7554/elife.49753] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/18/2019] [Indexed: 01/22/2023] Open
Abstract
Neuropathic pain is a debilitating condition caused by the abnormal processing of somatosensory input. Synaptic inhibition in the spinal dorsal horn plays a key role in that processing. Mechanical allodynia - the misperception of light touch as painful - occurs when inhibition is compromised. Disinhibition is due primarily to chloride dysregulation caused by hypofunction of the potassium-chloride co-transporter KCC2. Here we show, in rats, that excitatory neurons are disproportionately affected. This is not because chloride is differentially dysregulated in excitatory and inhibitory neurons, but, rather, because excitatory neurons rely more heavily on inhibition to counterbalance strong excitation. Receptive fields in both cell types have a center-surround organization but disinhibition unmasks more excitatory input to excitatory neurons. Differences in intrinsic excitability also affect how chloride dysregulation affects spiking. These results deepen understanding of how excitation and inhibition are normally balanced in the spinal dorsal horn, and how their imbalance disrupts somatosensory processing.
Collapse
Affiliation(s)
- Kwan Yeop Lee
- Neurosciences and Mental HealthThe Hospital for Sick ChildrenTorontoCanada
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoCanada
| | - Stéphanie Ratté
- Neurosciences and Mental HealthThe Hospital for Sick ChildrenTorontoCanada
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoCanada
| | - Steven A Prescott
- Neurosciences and Mental HealthThe Hospital for Sick ChildrenTorontoCanada
- Department of PhysiologyUniversity of TorontoTorontoCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoCanada
| |
Collapse
|
48
|
Navratilova E, Porreca F. Substance P and Inflammatory Pain: Getting It Wrong and Right Simultaneously. Neuron 2019; 101:353-355. [PMID: 30731054 DOI: 10.1016/j.neuron.2019.01.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
How do neuropeptides participate in the classic neuroinflammatory sequelae of tissue injury that includes pain, immune cell infiltration, and swelling? In this issue of Neuron, Green et al. (2019) reveal that MrgprB2/MrgprX2 is a mast cell substance-P-specific receptor that critically links neural and immune systems and offers new approaches for neuroinflammatory therapeutics.
Collapse
Affiliation(s)
- Edita Navratilova
- Department of Pharmacology, University of Arizona, Tucson, AZ 85718, USA.
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85718, USA; Mayo Clinic, Scottsdale, AZ 85259, USA.
| |
Collapse
|
49
|
Mapplebeck JCS, Lorenzo LE, Lee KY, Gauthier C, Muley MM, De Koninck Y, Prescott SA, Salter MW. Chloride Dysregulation through Downregulation of KCC2 Mediates Neuropathic Pain in Both Sexes. Cell Rep 2019; 28:590-596.e4. [PMID: 31315039 DOI: 10.1016/j.celrep.2019.06.059] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 05/09/2019] [Accepted: 06/14/2019] [Indexed: 02/08/2023] Open
Abstract
The behavioral features of neuropathic pain are not sexually dimorphic despite sex differences in the underlying neuroimmune signaling. This raises questions about whether neural processing is comparably altered. Here, we test whether the K+-Cl- co-transporter KCC2, which regulates synaptic inhibition, plays an equally important role in development of neuropathic pain in male and female rodents. Past studies on KCC2 tested only males. We find that inhibiting KCC2 in uninjured animals reproduces behavioral and electrophysiological features of neuropathic pain in both sexes and, consistent with equivalent injury-induced downregulation of KCC2, that counteracting chloride dysregulation reverses injury-induced behavioral and electrophysiological changes in both sexes. These findings demonstrate that KCC2 downregulation contributes equally to pain hypersensitivity in males and females. Whereas diverse (and sexually dimorphic) mechanisms regulate KCC2, regulation of intracellular chloride relies almost exclusively on KCC2. Directly targeting KCC2 thus remains a promising strategy for treatment of neuropathic pain in both sexes.
Collapse
Affiliation(s)
- Josiane C S Mapplebeck
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada; University of Toronto Centre for the Study of Pain, Toronto, ON, Canada
| | | | - Kwan Yeop Lee
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Cédric Gauthier
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec, QC, Canada
| | - Milind M Muley
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada; University of Toronto Centre for the Study of Pain, Toronto, ON, Canada
| | - Yves De Koninck
- CERVO Brain Research Centre, Quebec Mental Health Institute, Quebec, QC, Canada; Department of Psychiatry and Neuroscience, Université Laval, Quebec, QC, Canada
| | - Steven A Prescott
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada; University of Toronto Centre for the Study of Pain, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada; University of Toronto Centre for the Study of Pain, Toronto, ON, Canada.
| |
Collapse
|
50
|
Gazerani P. Identification of novel analgesics through a drug repurposing strategy. Pain Manag 2019; 9:399-415. [DOI: 10.2217/pmt-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The identification of new indications for approved or failed drugs is a process called drug repositioning or drug repurposing. The motivation includes overcoming the productivity gap that exists in drug development, which is a high-cost–high-risk process. Repositioning also includes rescuing drugs that have safely entered the market but have failed to demonstrate sufficient efficiency for the initial clinical indication. Considering the high prevalence of chronic pain, the lack of sufficient efficacy and the safety issues of current analgesics, repositioning seems to be an attractive approach. This review presents example of drugs that already have been repositioned and highlights new technologies that are available for the identification of additional compounds to stimulate the curiosity of readers for further exploration.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine, Department of Health Science & Technology, Aalborg University, Frederik Bajers Vej 3 B, 9220 Aalborg East, Denmark
| |
Collapse
|