1
|
Villa S, Aasvang EK, Attal N, Baron R, Bourinet E, Calvo M, Finnerup NB, Galosi E, Hockley JRF, Karlsson P, Kemp H, Körner J, Kutafina E, Lampert A, Mürk M, Nochi Z, Price TJ, Rice ASC, Sommer C, Taba P, Themistocleous AC, Treede RD, Truini A, Üçeyler N, Bennett DL, Schmid AB, Denk F. Harmonizing neuropathic pain research: outcomes of the London consensus meeting on peripheral tissue studies. Pain 2024:00006396-990000000-00743. [PMID: 39432804 DOI: 10.1097/j.pain.0000000000003445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/08/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT Neuropathic pain remains difficult to treat, with drug development hampered by an incomplete understanding of the pathogenesis of the condition, as well as a lack of biomarkers. The problem is compounded by the scarcity of relevant human peripheral tissues, including skin, nerves, and dorsal root ganglia. Efforts to obtain such samples are accelerating, increasing the need for standardisation across laboratories. In this white paper, we report on a consensus meeting attended by neuropathic pain experts, designed to accelerate protocol alignment and harmonization of studies involving relevant peripheral tissues. The meeting was held in London in March 2024 and attended by 28 networking partners, including industry and patient representatives. We achieved consensus on minimal recommended phenotyping, harmonised wet laboratory protocols, statistical design, reporting, and data sharing. Here, we also share a variety of relevant standard operating procedures as supplementary protocols. We envision that our recommendations will help unify human tissue research in the field and accelerate our understanding of how abnormal interactions between sensory neurons and their local peripheral environment contribute towards neuropathic pain.
Collapse
Affiliation(s)
- Sara Villa
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), King's College London, United Kingdom
| | - Eske K Aasvang
- Anesthesiological Department, Center for Cancer and Organ Dysfunction, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Nadine Attal
- INSERM U987, APHP, UVSQ Paris SACLAY University, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Emmanuel Bourinet
- Laboratories of Excellence, Ion Channel Science and Therapeutics, Institut de Génomique Fonctionnelle, Montpellier, France
- CNRS UMR5203, Montpellier, France
- INSERM, U661, Montpellier, France
- Université de Montpellier, Montpellier, France
| | - Margarita Calvo
- Biological Sciences Faculty and Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, CL, United States
| | - Nanna B Finnerup
- Department of Clinical Medicine, Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Eleonora Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | | | - Pall Karlsson
- Department of Clinical Medicine, Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Harriet Kemp
- Pain Research Group, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Jannis Körner
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Department of Anesthesiology, Uniklinik RWTH Aachen University
- Intensive and Intermediate Care, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Ekaterina Kutafina
- Institute for Biomedical Informatics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Research Aachen, SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Margarita Mürk
- Pathology Department, Tartu University Hospital, Tartu, Estonia
| | - Zahra Nochi
- Department of Clinical Medicine, Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Andrew S C Rice
- Pain Research Group, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | | | - Rolf-Detlef Treede
- Department of Psychiatry and Psychotherapy, Central Institute for Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andrea Truini
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Nurcan Üçeyler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Annina B Schmid
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Franziska Denk
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), King's College London, United Kingdom
| |
Collapse
|
2
|
Cooper AH, Barry AM, Chrysostomidou P, Lolignier R, Wang J, Redondo Canales M, Titterton HF, Bennett DL, Weir GA. Peripheral nerve injury results in a biased loss of sensory neuron subpopulations. Pain 2024:00006396-990000000-00668. [PMID: 39158319 DOI: 10.1097/j.pain.0000000000003321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/25/2024] [Indexed: 08/20/2024]
Abstract
ABSTRACT There is a rich literature describing the loss of dorsal root ganglion (DRG) neurons following peripheral axotomy, but the vulnerability of discrete subpopulations has not yet been characterised. Furthermore, the extent or even presence of neuron loss following injury has recently been challenged. In this study, we have used a range of transgenic recombinase driver mouse lines to genetically label molecularly defined subpopulations of DRG neurons and track their survival following traumatic nerve injury. We find that spared nerve injury leads to a marked loss of cells containing DRG volume and a concomitant loss of small-diameter DRG neurons. Neuron loss occurs unequally across subpopulations and is particularly prevalent in nonpeptidergic nociceptors, marked by expression of Mrgprd. We show that this subpopulation is almost entirely lost following spared nerve injury and severely depleted (by roughly 50%) following sciatic nerve crush. Finally, we used an in vitro model of DRG neuron survival to demonstrate that nonpeptidergic nociceptor loss is likely dependent on the absence of neurotrophic support. Together, these results profile the extent to which DRG neuron subpopulations can survive axotomy, with implications for our understanding of nerve injury-induced plasticity and pain.
Collapse
Affiliation(s)
- Andrew H Cooper
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Romane Lolignier
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - Jinyi Wang
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | | | - Heather F Titterton
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Greg A Weir
- School of Psychology and Neuroscience, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
3
|
Mogil JS, Parisien M, Esfahani SJ, Diatchenko L. Sex differences in mechanisms of pain hypersensitivity. Neurosci Biobehav Rev 2024; 163:105749. [PMID: 38838876 DOI: 10.1016/j.neubiorev.2024.105749] [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: 04/10/2024] [Revised: 05/23/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
The introduction of sex-as-a-biological-variable policies at funding agencies around the world has led to an explosion of very recent observations of sex differences in the biology underlying pain. This review considers evidence of sexually dimorphic mechanisms mediating pain hypersensitivity, derived from modern assays of persistent pain in rodent animal models. Three well-studied findings are described in detail: the male-specific role of spinal cord microglia, the female-specific role of calcitonin gene-related peptide (CGRP), and the female-specific role of prolactin and its receptor. Other findings of sex-specific molecular involvement in pain are subjected to pathway analyses and reveal at least one novel hypothesis: that females may preferentially use Th1 and males Th2 T cell activity to mediate chronic pain.
Collapse
Affiliation(s)
- Jeffrey S Mogil
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3A 1B1, Canada.
| | - Marc Parisien
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3A 1B1, Canada
| | - Sahel J Esfahani
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3A 1B1, Canada
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, QC H3A 1B1, Canada
| |
Collapse
|
4
|
Testa L, Dotta S, Vercelli A, Marvaldi L. Communicating pain: emerging axonal signaling in peripheral neuropathic pain. Front Neuroanat 2024; 18:1398400. [PMID: 39045347 PMCID: PMC11265228 DOI: 10.3389/fnana.2024.1398400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Peripheral nerve damage often leads to the onset of neuropathic pain (NeuP). This condition afflicts millions of people, significantly burdening healthcare systems and putting strain on families' financial well-being. Here, we will focus on the role of peripheral sensory neurons, specifically the Dorsal Root Ganglia neurons (DRG neurons) in the development of NeuP. After axotomy, DRG neurons activate regenerative signals of axons-soma communication to promote a gene program that activates an axonal branching and elongation processes. The results of a neuronal morphological cytoskeleton change are not always associated with functional recovery. Moreover, any axonal miss-targeting may contribute to NeuP development. In this review, we will explore the epidemiology of NeuP and its molecular causes at the level of the peripheral nervous system and the target organs, with major focus on the neuronal cross-talk between intrinsic and extrinsic factors. Specifically, we will describe how failures in the neuronal regenerative program can exacerbate NeuP.
Collapse
Affiliation(s)
- Livia Testa
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Sofia Dotta
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Letizia Marvaldi
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| |
Collapse
|
5
|
Reynders A, Anissa Jhumka Z, Gaillard S, Mantilleri A, Malapert P, Magalon K, Etzerodt A, Salio C, Ugolini S, Castets F, Saurin AJ, Serino M, Hoeffel G, Moqrich A. Gut microbiota promotes pain chronicity in Myosin1A deficient male mice. Brain Behav Immun 2024; 119:750-766. [PMID: 38710336 DOI: 10.1016/j.bbi.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.
Collapse
Affiliation(s)
- Ana Reynders
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France.
| | - Z Anissa Jhumka
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | | | - Annabelle Mantilleri
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Pascale Malapert
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Karine Magalon
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Sophie Ugolini
- Aix-Marseille-Université, CNRS, INSER, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Francis Castets
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Andrew J Saurin
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France
| | - Matteo Serino
- Institut de Recherche en Santé Digestive, Université de Toulouse-Paul Sabatier, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Guillaume Hoeffel
- Aix-Marseille-Université, CNRS, INSER, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institut de Biologie du Développement de Marseille, Marseille, France.
| |
Collapse
|
6
|
Qu Y, Cai R, Li Q, Wang H, Lu L. Neuroinflammation signatures in dorsal root ganglia following chronic constriction injury. Heliyon 2024; 10:e31481. [PMID: 38813203 PMCID: PMC11133895 DOI: 10.1016/j.heliyon.2024.e31481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/17/2023] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Neuropathic pain (NP) is a common debilitating chronic pain condition with limited effective therapeutics. Further investigating mechanisms underlying NP is therefore of great importance for discovering more promising therapeutic targets. In the current study, we employed high-throughput RNA sequencing to explore transcriptome profiles of mRNAs and microRNAs in the dorsal root ganglia (DRG) following chronic constriction injury (CCI) and also integrated published datasets for comprehensive analysis. First, we established CCI rat model confirmed by behavioral testings, and excavated 467 differentially expressed mRNAs (DEGs) and 16 differentially expressed microRNAs (DEmiRNAs) in the ipsilateral lumbar 4-6 DRG of CCI rats 11 days after surgery. Functional enrichment analysis of 337 upregulated DEGs showed that most of the DEGs were enriched in inflammation- and immune-associated biological processes and signaling pathways. The protein-protein interaction networks were constructed and hub DEGs were screened. Besides hub DEGs, we also identified 113 overlapped DEGs by intersecting our dataset with dataset GSE100122. Subsequently, we predicted potential miRNA-mRNA regulatory pairs using DEmiRNAs and a given set of key DEGs (including hub and overlapped DEGs). By integrative analysis, we found commonly differentially expressed mRNAs and miRNAs following CCI of different time points and different nerve injury types. Highlighted mRNAs include Atf3, Vip, Gal, Npy, Adcyap1, Reg3b, Jun, Cd74, Gadd45a, Tgm1, Csrp3, Sprr1a, Serpina3n, Gap43, Serpinb2 and Vtcn1, while miRNAs include miR-21-5p, miR-34a-5p, miR-200a-3p, miR-130a-5p, miR-216b-5p, miR-217-5p, and miR-541-5p. Additionally, 15 DEGs, including macrophages-specific (Cx3cr1, Arg1, Cd68, Csf1r) and the ones related to macrophages' involvement in NP (Ccl2, Fcgr3a, Bdnf, Ctss, Tyrobp) were verified by qRT-PCR. By functional experiments in future studies, promising therapeutic targets for NP treatment may be identified among these mRNAs and miRNAs.
Collapse
Affiliation(s)
- Yao Qu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin, China
- Department of Pain Medicine, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Ruirui Cai
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, No.2555 Jingyue Street, Changchun, 130117, Jilin, China
| | - Qiao Li
- Department of Spinal Surgery, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, No.2555 Jingyue Street, Changchun, 130117, Jilin, China
| | - Laijin Lu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130021, Jilin, China
| |
Collapse
|
7
|
Barakat A, Munro G, Heegaard AM. Finding new analgesics: Computational pharmacology faces drug discovery challenges. Biochem Pharmacol 2024; 222:116091. [PMID: 38412924 DOI: 10.1016/j.bcp.2024.116091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/10/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024]
Abstract
Despite the worldwide prevalence and huge burden of pain, pain is an undertreated phenomenon. Currently used analgesics have several limitations regarding their efficacy and safety. The discovery of analgesics possessing a novel mechanism of action has faced multiple challenges, including a limited understanding of biological processes underpinning pain and analgesia and poor animal-to-human translation. Computational pharmacology is currently employed to face these challenges. In this review, we discuss the theory, methods, and applications of computational pharmacology in pain research. Computational pharmacology encompasses a wide variety of theoretical concepts and practical methodological approaches, with the overall aim of gaining biological insight through data acquisition and analysis. Data are acquired from patients or animal models with pain or analgesic treatment, at different levels of biological organization (molecular, cellular, physiological, and behavioral). Distinct methodological algorithms can then be used to analyze and integrate data. This helps to facilitate the identification of biological molecules and processes associated with pain phenotype, build quantitative models of pain signaling, and extract translatable features between humans and animals. However, computational pharmacology has several limitations, and its predictions can provide false positive and negative findings. Therefore, computational predictions are required to be validated experimentally before drawing solid conclusions. In this review, we discuss several case study examples of combining and integrating computational tools with experimental pain research tools to meet drug discovery challenges.
Collapse
Affiliation(s)
- Ahmed Barakat
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Assiut, Egypt.
| | | | - Anne-Marie Heegaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
O'Brien JA, Karrasch JF, Huang Y, Vine EE, Cunningham AL, Harman AN, Austin PJ. Nerve-myeloid cell interactions in persistent human pain: a reappraisal using updated cell subset classifications. Pain 2024; 165:753-771. [PMID: 37975868 DOI: 10.1097/j.pain.0000000000003106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 09/04/2023] [Indexed: 11/19/2023]
Abstract
ABSTRACT The past 20 years have seen a dramatic shift in our understanding of the role of the immune system in initiating and maintaining pain. Myeloid cells, including macrophages, dendritic cells, Langerhans cells, and mast cells, are increasingly implicated in bidirectional interactions with nerve fibres in rodent pain models. However, our understanding of the human setting is still poor. High-dimensional functional analyses have substantially changed myeloid cell classifications, with recently described subsets such as epidermal dendritic cells and DC3s unveiling new insight into how myeloid cells interact with nerve fibres. However, it is unclear whether this new understanding has informed the study of human chronic pain. In this article, we perform a scoping review investigating neuroimmune interactions between myeloid cells and peripheral nerve fibres in human chronic pain conditions. We found 37 papers from multiple pain states addressing this aim in skin, cornea, peripheral nerve, endometrium, and tumour, with macrophages, Langerhans cells, and mast cells the most investigated. The directionality of results between studies was inconsistent, although the clearest pattern was an increase in macrophage frequency across conditions, phases, and tissues. Myeloid cell definitions were often outdated and lacked correspondence with the stated cell types of interest; overreliance on morphology and traditional structural markers gave limited insight into the functional characteristics of investigated cells. We therefore critically reappraise the existing literature considering contemporary myeloid cell biology and advocate for the application of established and emerging high-dimensional proteomic and transcriptomic single-cell technologies to clarify the role of specific neuroimmune interactions in chronic pain.
Collapse
Affiliation(s)
- Jayden A O'Brien
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Jackson F Karrasch
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Yun Huang
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erica E Vine
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Anthony L Cunningham
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Andrew N Harman
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| | - Paul J Austin
- Brain and Mind Centre, The University of Sydney, Sydney, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Virus Research, The Westmead Institute for Medical Research, Westmead, Australia
| |
Collapse
|
9
|
Dourson AJ, Jankowski MP. Developmental impact of peripheral injury on neuroimmune signaling. Brain Behav Immun 2023; 113:156-165. [PMID: 37442302 PMCID: PMC10530254 DOI: 10.1016/j.bbi.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/01/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
A peripheral injury drives neuroimmune interactions at the level of the injury and throughout the neuraxis. Understanding these systems will be beneficial in the pursuit to target persistent pain that involves both neural and immune components. In this review, we discuss the impact of injury on the development of neuroimmune signaling, along with data that suggest a possible cellular immune memory. We also discuss the parallel effects of injury in the nervous system and immune related areas including bone marrow, lymph node and central nervous system-related cells. Finally, we relate these findings to patient populations and current research that evaluates human tissue.
Collapse
Affiliation(s)
- Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Pediatric Pain Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
10
|
Wang L, Ishihara S, Li J, Miller RE, Malfait AM. Notch signaling is activated in knee-innervating dorsal root ganglia in experimental models of osteoarthritis joint pain. Arthritis Res Ther 2023; 25:63. [PMID: 37061736 PMCID: PMC10105425 DOI: 10.1186/s13075-023-03039-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/28/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND We aimed to explore activation of the Notch signaling pathway in knee-innervating lumbar dorsal root ganglia (DRG) in the course of experimental osteoarthritis (OA) in mice, and its role in knee hyperalgesia. METHODS Cultured DRG cells were stimulated with the TLR4 agonist, lipopolysaccharide (LPS). Notch signaling in the cells was either inhibited with the γ-secretase inhibitor, DAPT, or with soluble Jagged1, or activated through immobilized Jagged1. CCL2 production was analyzed at mRNA and protein levels. In in vivo experiments, knee hyperalgesia was induced in naïve mice through intra-articular (IA) injection of LPS. The effect of inhibiting Notch signaling was examined by pre-injecting DAPT one hour before LPS. OA was induced through surgical destabilization of the medial meniscus (DMM) in male C57BL/6 mice. Gene expression in DRG was analyzed by qRT-PCR and RNAscope in situ hybridization. Activated Notch protein (NICD) expression in DRG was evaluated by ELISA and immunofluorescence staining. DAPT was injected IA 12 weeks post DMM to inhibit Notch signaling, followed by assessing knee hyperalgesia and CCL2 expression in the DRG. RESULTS In DRG cell cultures, LPS increased NICD in neuronal cells. Inhibition of Notch signaling with either DAPT or soluble Jagged1 attenuated LPS-induced increases of Ccl2 mRNA and CCL2 protein. Conversely, activating Notch signaling with immobilized Jagged1 enhanced these LPS effects. In vivo, IA injection of LPS increased expression of Notch genes and NICD in the DRG. Pre-injection of DAPT prior to LPS alleviated LPS-induced knee hyperalgesia, and decreased LPS-induced CCL2 expression in the DRG. Notch signaling genes were differentially expressed in the DRG from late-stage experimental OA. Notch1, Hes1, and NICD were increased in the neuronal cell bodies in DRG after DMM surgery. IA administration of DAPT alleviated knee hyperalgesia post DMM, and decreased CCL2 expression in the DRG. CONCLUSIONS These findings suggest a synergistic effect of Notch signaling with TLR4 in promoting CCL2 production and mediating knee hyperalgesia. Notch signaling is activated in knee-innervating lumbar DRG in mice with experimental OA, and is involved in mediating knee hyperalgesia. The pathway may therefore be explored as a target for alleviating OA pain.
Collapse
Affiliation(s)
- Lai Wang
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 West Harrison Street, Suite 510, Chicago, IL, 60612, USA.
| | - Shingo Ishihara
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 West Harrison Street, Suite 510, Chicago, IL, 60612, USA
| | - Jun Li
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 West Harrison Street, Suite 510, Chicago, IL, 60612, USA
| | - Rachel E Miller
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 West Harrison Street, Suite 510, Chicago, IL, 60612, USA
| | - Anne-Marie Malfait
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical Center, 1611 West Harrison Street, Suite 510, Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Abstract
Interactions between the immune and nervous systems are of central importance in neuropathic pain, a common and debilitating form of chronic pain caused by a lesion or disease affecting the somatosensory system. Our understanding of neuroimmune interactions in pain research has advanced considerably. Initially considered as passive bystanders, then as culprits in the pathogenesis of neuropathic pain, immune responses in the nervous system are now established to underpin not only the initiation and progression of pain but also its resolution. Indeed, immune cells and their mediators are well-established promoters of neuroinflammation at each level of the neural pain pathway that contributes to pain hypersensitivity. However, emerging evidence indicates that specific subtypes of immune cells (including antinociceptive macrophages, pain-resolving microglia and T regulatory cells) as well as immunoresolvent molecules and modulators of the gut microbiota-immune system axis can reduce the pain experience and contribute to the resolution of neuropathic pain. This Review provides an overview of the immune mechanisms responsible for the resolution of neuropathic pain, including those involved in innate, adaptive and meningeal immunity as well as interactions with the gut microbiome. Specialized pro-resolving mediators and therapeutic approaches that target these neuroimmune mechanisms are also discussed.
Collapse
|
12
|
Ray PR, Shiers S, Caruso JP, Tavares-Ferreira D, Sankaranarayanan I, Uhelski ML, Li Y, North RY, Tatsui C, Dussor G, Burton MD, Dougherty PM, Price TJ. RNA profiling of human dorsal root ganglia reveals sex differences in mechanisms promoting neuropathic pain. Brain 2023; 146:749-766. [PMID: 35867896 PMCID: PMC10169414 DOI: 10.1093/brain/awac266] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/16/2022] [Accepted: 06/22/2022] [Indexed: 11/12/2022] Open
Abstract
Neuropathic pain is a leading cause of high-impact pain, is often disabling and is poorly managed by current therapeutics. Here we focused on a unique group of neuropathic pain patients undergoing thoracic vertebrectomy where the dorsal root ganglia is removed as part of the surgery allowing for molecular characterization and identification of mechanistic drivers of neuropathic pain independently of preclinical models. Our goal was to quantify whole transcriptome RNA abundances using RNA-seq in pain-associated human dorsal root ganglia from these patients, allowing comprehensive identification of molecular changes in these samples by contrasting them with non-pain-associated dorsal root ganglia. We sequenced 70 human dorsal root ganglia, and among these 50 met inclusion criteria for sufficient neuronal mRNA signal for downstream analysis. Our expression analysis revealed profound sex differences in differentially expressed genes including increase of IL1B, TNF, CXCL14 and OSM in male and CCL1, CCL21, PENK and TLR3 in female dorsal root ganglia associated with neuropathic pain. Coexpression modules revealed enrichment in members of JUN-FOS signalling in males and centromere protein coding genes in females. Neuro-immune signalling pathways revealed distinct cytokine signalling pathways associated with neuropathic pain in males (OSM, LIF, SOCS1) and females (CCL1, CCL19, CCL21). We validated cellular expression profiles of a subset of these findings using RNAscope in situ hybridization. Our findings give direct support for sex differences in underlying mechanisms of neuropathic pain in patient populations.
Collapse
Affiliation(s)
- Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - James P Caruso
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA.,Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Megan L Uhelski
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Yan Li
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Y North
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio Tatsui
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Michael D Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| | - Patrick M Dougherty
- Department of Pain Medicine, Division of Anesthesiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
13
|
Article Type: Original Article Title: Linalyl Acetate Ameliorates Mechanical Hyperalgesia Through Suppressing Inflammation by TSLP/IL-33 Signaling. Neurochem Res 2022; 47:3805-3816. [DOI: 10.1007/s11064-022-03763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 10/31/2022]
Abstract
AbstractNeuropathic pain is a debilitating chronic disorder, significantly causing personal and social burdens, in which activated neuroinflammation is one major contributor. Thymic stromal lymphopoietin (TSLP) and interleukin (IL)-33 is important for chronic inflammation. Linalyl acetate (LA) is main component of lavender oil with an anti-inflammatory property through TSLP signaling. The aim of the study is to investigate how LA regulates mechanical hyperalgesia after sciatic nerve injury (SNI). Adult Sprague-Dawley male rats were separated into 3 groups: control group, SNI group and SNI with LA group. LA was administrated intraperitoneally one day before SNI. Pain behavior test was evaluated through calibration forceps testing. Ipsilateral sciatic nerves (SNs), dorsal root ganglions (DRGs) and spinal cord were collected for immunofluorescence staining and Western blotting analyses. SNI rats were more sensitive to hyperalgesia response to mechanical stimulus since operation, which was accompanied by spinal cord glial cells reactions and DRG neuro-glial interaction. LA could relieve the pain sensation, proinflammatory cytokines and decrease the expression of TSLP/TSLPR complex. Also, LA could reduce inflammation through reducing IL-33 signaling. This study is the first to indicate that LA can modulate pain through TSLP/TSLPR and IL-33 signaling after nerve injury.
Collapse
|
14
|
Gheorghe RO, Grosu AV, Bica-Popi M, Ristoiu V. The Yin/Yang Balance of Communication between Sensory Neurons and Macrophages in Traumatic Peripheral Neuropathic Pain. Int J Mol Sci 2022; 23:ijms232012389. [PMID: 36293246 PMCID: PMC9603877 DOI: 10.3390/ijms232012389] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Traumatic peripheral neuropathic pain is a complex syndrome caused by a primary lesion or dysfunction of the peripheral nervous system. Secondary to the lesion, resident or infiltrating macrophages proliferate and initiate a cross-talk with the sensory neurons, at the level of peripheral nerves and sensory ganglia. The neuron–macrophage interaction, which starts very early after the lesion, is very important for promoting pain development and for initiating changes that will facilitate the chronicization of pain, but it also has the potential to facilitate the resolution of injury-induced changes and, consequently, promote the reduction of pain. This review is an overview of the unique characteristics of nerve-associated macrophages in the peripheral nerves and sensory ganglia and of the molecules and signaling pathways involved in the neuro-immune cross-talk after a traumatic lesion, with the final aim of better understanding how the balance between pro- and anti-nociceptive dialogue between neurons and macrophages may be modulated for new therapeutic approaches.
Collapse
|
15
|
Bogdan DM, Studholme K, DiBua A, Gordon C, Kanjiya MP, Yu M, Puopolo M, Kaczocha M. FABP5 deletion in nociceptors augments endocannabinoid signaling and suppresses TRPV1 sensitization and inflammatory pain. Sci Rep 2022; 12:9241. [PMID: 35655086 PMCID: PMC9163147 DOI: 10.1038/s41598-022-13284-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
The endocannabinoid anandamide (AEA) produces antinociceptive effects by activating cannabinoid receptor 1 (CB1). However, AEA also serves as an agonist at transient receptor potential vanilloid receptor 1 (TRPV1) in nociceptive sensory neurons, which may exacerbate pain. This potential functional duality is highlighted by the failure of an inhibitor of the AEA catabolic enzyme fatty acid amide hydrolase (FAAH) to afford pain relief in a clinical trial. Consequently, it remains to be determined whether elevating AEA levels in nociceptors leads to antinociceptive or pro-nociceptive effects. Fatty acid binding protein 5 (FABP5) is an intracellular carrier that mediates AEA transport to FAAH for inactivation. Leveraging the abundant expression of FABP5 in TRPV1+ nociceptors, we employed a conditional knockout strategy to demonstrate that FABP5 deletion in nociceptors augments AEA levels, resulting in the emergence of antinociceptive effects mediated by CB1. Mechanistically, FABP5 deletion suppresses inflammation- and nerve growth factor-mediated TRPV1 sensitization via CB1, an effect mediated by calcineurin. Unexpectedly, inhibition of FAAH failed to blunt TRPV1 sensitization, uncovering functionally distinct outputs resulting from FABP5 and FAAH inhibition. Collectively, our results demonstrate that FABP5 serves a key role in governing endocannabinoid signaling in nociceptors to disrupt TRPV1 sensitization and pain, and position FABP5 as a therapeutic target for the development of analgesics.
Collapse
Affiliation(s)
- Diane M Bogdan
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Keith Studholme
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Adriana DiBua
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Chris Gordon
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Martha P Kanjiya
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Mei Yu
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michelino Puopolo
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Martin Kaczocha
- Department of Anesthesiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
- Stony Brook University Pain and Analgesia Research Center (SPARC), Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
16
|
Wang F, Tao R, Zhao L, Hao XH, Zou Y, Lin Q, Liu MM, Goldman G, Luo D, Chen S. Differential lncRNA/mRNA Expression Profiling and Functional Network Analyses in Bmp2 Deletion of Mouse Dental Papilla Cells. Front Genet 2022; 12:702540. [PMID: 35003201 PMCID: PMC8727545 DOI: 10.3389/fgene.2021.702540] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022] Open
Abstract
Bmp2 is essential for dentin development and formation. Bmp2 conditional knock-out (KO) mice display a similar tooth phenotype of dentinogenesis imperfecta (DGI). To elucidate a foundation for subsequent functional studies of cross talk between mRNAs and lncRNAs in Bmp2-mediated dentinogenesis, we investigated the profiling of lncRNAs and mRNAs using immortalized mouse dental Bmp2 flox/flox (iBmp2fx/fx) and Bmp2 knock-out (iBmp2ko/ko) papilla cells. RNA sequencing was implemented to study the expression of the lncRNAs and mRNAs. Quantitative real-time PCR (RT-qPCR) was used to validate expressions of lncRNAs and mRNAs. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were used to predict functions of differentially expressed genes (DEGs). Protein-protein interaction (PPI) and lncRNA-mRNA co-expression network were analyzed by using bioinformatics methods. As a result, a total of 22 differentially expressed lncRNAs (16 downregulated vs 6 upregulated) and 227 differentially expressed mRNAs (133 downregulated vs. 94 upregulated) were identified in the iBmp2ko/ko cells compared with those of the iBmp2fx/fx cells. RT-qPCR results showed significantly differential expressions of several lncRNAs and mRNAs which were consistent with the RNA-seq data. GO and KEGG analyses showed differentially expressed genes were closely related to cell differentiation, transcriptional regulation, and developmentally relevant signaling pathways. Moreover, network-based bioinformatics analysis depicted the co-expression network between lncRNAs and mRNAs regulated by Bmp2 in mouse dental papilla cells and symmetrically analyzed the effect of Bmp2 during dentinogenesis via coding and non-coding RNA signaling.
Collapse
Affiliation(s)
- Feng Wang
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Developmental Dentistry, School of Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Ran Tao
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Li Zhao
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xin-Hui Hao
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yi Zou
- Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Qing Lin
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Meng Meng Liu
- Department of Developmental Dentistry, School of Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Graham Goldman
- Department of Developmental Dentistry, School of Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Daoshu Luo
- Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shuo Chen
- Department of Developmental Dentistry, School of Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
17
|
Macrophage Activation in the Dorsal Root Ganglion in Rats Developing Autotomy after Peripheral Nerve Injury. Int J Mol Sci 2021; 22:ijms222312801. [PMID: 34884605 PMCID: PMC8657625 DOI: 10.3390/ijms222312801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
Autotomy, self-mutilation of a denervated limb, is common in animals after peripheral nerve injury (PNI) and is a reliable proxy for neuropathic pain in humans. Understanding the occurrence and treatment of autotomy remains challenging. The objective of this study was to investigate the occurrence of autotomy in nude and Wistar rats and evaluate the differences in macrophage activation and fiber sensitization contributing to the understanding of autotomy behavior. Autotomy in nude and Wistar rats was observed and evaluated 6 and 12 weeks after sciatic nerve repair surgery. The numbers of macrophages and the types of neurons in the dorsal root ganglion (DRG) between the two groups were compared by immunofluorescence studies. Immunostaining of T cells in the DRG was also assessed. Nude rats engaged in autotomy with less frequency than Wistar rats. Autotomy symptoms were also relatively less severe in nude rats. Immunofluorescence studies revealed increased macrophage accumulation and activation in the DRG of Wistar rats. The percentage of NF200+ neurons was higher at 6 and 12 weeks in Wistar rats compared to nude rats, but the percentage of CGRP+ neurons did not differ between two groups. Additionally, macrophages were concentrated around NF200-labeled A fibers. At 6 and 12 weeks following PNI, CD4+ T cells were not found in the DRG of the two groups. The accumulation and activation of macrophages in the DRG may account for the increased frequency and severity of autotomy in Wistar rats. Our results also suggest that A fiber neurons in the DRG play an important role in autotomy.
Collapse
|
18
|
Westlund K, Montera M, Goins A, Alles S, Afaghpour-Becklund M, Bartel R, Durvasula R, Kunamneni A. Single-chain Fragment variable antibody targeting cholecystokinin-B receptor for pain reduction. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100067. [PMID: 34458647 PMCID: PMC8378781 DOI: 10.1016/j.ynpai.2021.100067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 05/13/2023]
Abstract
The cholecystokinin B receptor and its neuropeptide ligand are upregulated in chronic neuropathic pain models. Single-chain Fragment variable antibodies were generated as preferred non-opioid targeting therapy blocking the cholecystokinin B receptor to inhibit chronic neuropathic pain models in vivo and in vitro. Engineered antibodies of this type feature binding activity similar to monoclonal antibodies but with stronger affinity and increased tissue penetrability due to their smaller size. More importantly, single-chain Fragment variable antibodies have promising biotherapeutic applications for both nervous and immune systems, now recognized as interactive in chronic pain. A mouse single-chain Fragment variable antibody library recognizing a fifteen amino acid extracellular peptide fragment of the cholecystokinin B receptor was generated from immunized spleens. Ribosome display, a powerful cell-free technology, was applied for recombinant antibody selection. Antibodies with higher affinity, stability, solubility, and binding specificity for cholecystokinin B not A receptor were selected and optimized for in vivo and in vitro efficacy. A single dose of the lead candidate reduced mechanical and cold hypersensitivity in two rodent models of neuropathic pain for at least seven weeks. Continuing efficacy was evident with either intraperitoneal or intranasal dosing. Likewise, the lead single-chain Fragment variable antibody totally prevented development of anxiety- and depression-like behaviors and cognitive deficits typical in the models. Reduction of neuronal firing frequency was evident in trigeminal ganglia primary neuronal cultures treated in vitro with the cholecystokinin B receptor antibody. Immunofluorescent staining intensity in the trigeminal neuron primary cultures was significantly reduced incrementally after overnight binding with increasingly higher dilutions of the single-chain Fragment variable antibody. While it is reported that single-chain Fragment variable antibodies are removed systemically within 2-6 h, Western blot evidence indicates the His-tag marker remained after 7 weeks in the trigeminal ganglia and in the dorsolateral medulla, providing evidence of brain and ganglia penetrance known to be compromised in overactivated states. This project showcases the in vivo efficacy of our lead single-chain Fragment variable antibody indicating its potential for development as a non-opioid, non-addictive therapeutic intervention for chronic pain. Importantly, studies by others have indicated treatments with cholecystokinin B receptor antagonists suppress maintenance and reactivation of morphine dependence in place preference tests while lowering tolerance and dose requirements. Our future studies remain to address these potential benefits that may accompany the cholecystokinin B receptor biological therapy. Both chronic sciatic and orofacial pain can be unrelenting and excruciating, reducing quality of life as well as diminishing physical and mental function. An effective non-opiate, non-addictive therapy with potential to significantly reduce chronic neuropathic pain long term is greatly needed.
Collapse
Key Words
- ANOVA, analysis of variance
- ARM, antibody ribosome mRNA
- Anxiety
- BBB, blood–brain barrier
- CCK-8, cholecystokinin octapeptide
- CCK-BR, cholecystokinin B receptor
- CPP, conditioned place preference
- Chronic pain
- DRG, dorsal root ganglia
- Depression
- Eukaryotic ribosome display
- FRICT-ION, foramen rotundum inflammatory compression trigeminal infraorbital nerve model
- GPCR, G-protein-coupled receptor
- IACUC, Institutional Animal Care and Use Committee
- ION, infraorbital nerve
- MΩ, megaOhms
- PBS, phosphate buffered saline
- SEM, standard error of the mean
- TG, trigeminal ganglia
- ms, milliseconds
- pA, picoAmps
- scFv
- scFv, single-chain Fragment variable antibody
Collapse
Affiliation(s)
- K.N. Westlund
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
- Biomedical Laboratory Research & Development (121F), New Mexico VA
Health Care System, Albuquerque, NM, USA
| | - M.A. Montera
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - A.E. Goins
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - S.R.A. Alles
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - M. Afaghpour-Becklund
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - R. Bartel
- Department of Anesthesiology & Critical Care Medicine, University of
New Mexico Health Sciences Center, Albuquerque, NM 87106-0001, USA
| | - R. Durvasula
- Division of Infectious Diseases, Department of Internal Medicine, Mayo
Clinic, Jacksonville, FL, USA
- Department of Medicine, Loyola University Medical Center, Maywood, IL
60153-3328, USA
| | - A. Kunamneni
- Division of Infectious Diseases, Department of Internal Medicine, Mayo
Clinic, Jacksonville, FL, USA
- Department of Medicine, Loyola University Medical Center, Maywood, IL
60153-3328, USA
| |
Collapse
|
19
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
20
|
Joshi HP, Jo HJ, Kim YH, An SB, Park CK, Han I. Stem Cell Therapy for Modulating Neuroinflammation in Neuropathic Pain. Int J Mol Sci 2021; 22:ijms22094853. [PMID: 34063721 PMCID: PMC8124149 DOI: 10.3390/ijms22094853] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain (NP) is a complex, debilitating, chronic pain state, heterogeneous in nature and caused by a lesion or disease affecting the somatosensory system. Its pathogenesis involves a wide range of molecular pathways. NP treatment is extremely challenging, due to its complex underlying disease mechanisms. Current pharmacological and nonpharmacological approaches can provide long-lasting pain relief to a limited percentage of patients and lack safe and effective treatment options. Therefore, scientists are focusing on the introduction of novel treatment approaches, such as stem cell therapy. A growing number of reports have highlighted the potential of stem cells for treating NP. In this review, we briefly introduce NP, current pharmacological and nonpharmacological treatments, and preclinical studies of stem cells to treat NP. In addition, we summarize stem cell mechanisms—including neuromodulation in treating NP. Literature searches were conducted using PubMed to provide an overview of the neuroprotective effects of stem cells with particular emphasis on recent translational research regarding stem cell-based treatment of NP, highlighting its potential as a novel therapeutic approach.
Collapse
Affiliation(s)
- Hari Prasad Joshi
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
- Spinal Cord Research Centre, Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Hyun-Jung Jo
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
| | - Yong-Ho Kim
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
| | - Seong-Bae An
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
| | - Chul-Kyu Park
- Gachon Pain Center, Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Gyeonggi-do, Korea; (H.-J.J.); (Y.-H.K.)
- Correspondence: (C.-K.P.); (I.H.)
| | - Inbo Han
- Department of Neurosurgery, School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (H.P.J.); (S.-B.A.)
- Correspondence: (C.-K.P.); (I.H.)
| |
Collapse
|
21
|
Hore ZL, Villa-Hernandez S, Denk F. Probing the peripheral immune response in mouse models of oxaliplatin-induced peripheral neuropathy highlights their limited translatability. Wellcome Open Res 2021; 6:68. [PMID: 34250264 PMCID: PMC8243229 DOI: 10.12688/wellcomeopenres.16635.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 04/03/2024] Open
Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a disabling side effect of various chemotherapeutic agents, including oxaliplatin. It is highly prevalent amongst cancer patients, causing sensory abnormalities and pain. Unfortunately, as the underlying mechanisms remain poorly understood, effective therapeutics are lacking. Neuro-immune interactions have been highlighted as potential contributors to the development and maintenance of CIPN, however, whether this is the case in oxaliplatin-induced peripheral neuropathy (OIPN) is yet to be fully established. Methods: In this study we used flow cytometry to examine the peripheral immune response of male C57BL/6 mice following both single and repeated oxaliplatin administration. In animals exposed to repeated dosing, we also undertook mechanical and thermal behavioural assays to investigate how oxaliplatin alters phenotype, and conducted RT-qPCR experiments on bone marrow derived macrophages in order to further inspect the effects of oxaliplatin on immune cells. Results: In contrast to other reports, we failed to observe substantial changes in overall leukocyte, lymphocyte or myeloid cell numbers in dorsal root ganglia, sciatic nerves or inguinal lymph nodes. We did however note subtle, tissue-dependant alterations in several myeloid subpopulations following repeated dosing. These included a significant reduction in MHCII antigen presenting cells in the sciatic nerve and an increase in infiltrating cell types into the inguinal lymph nodes. Though repeated oxaliplatin administration had a systemic effect, we were unable to detect a pain-like behavioural phenotype in response to either cold or mechanical stimuli. Consequently, we cannot comment on whether the observed myeloid changes are associated with OIPN. Conclusions: Our discussion puts these results into the wider context of the field, advocating for greater transparency in reporting, alignment in experimental design and the introduction of more clinically relevant models. Only through joint concerted effort can we hope to increase our understanding of the underlying mechanisms of CIPN, including any immune contributions.
Collapse
Affiliation(s)
- Zoe Lee Hore
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Sara Villa-Hernandez
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK
| |
Collapse
|
22
|
Lacagnina MJ, Heijnen CJ, Watkins LR, Grace PM. Autoimmune regulation of chronic pain. Pain Rep 2021; 6:e905. [PMID: 33981931 PMCID: PMC8108590 DOI: 10.1097/pr9.0000000000000905] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/19/2020] [Accepted: 01/19/2021] [Indexed: 01/24/2023] Open
Abstract
Chronic pain is an unpleasant and debilitating condition that is often poorly managed by existing therapeutics. Reciprocal interactions between the nervous system and the immune system have been recognized as playing an essential role in the initiation and maintenance of pain. In this review, we discuss how neuroimmune signaling can contribute to peripheral and central sensitization and promote chronic pain through various autoimmune mechanisms. These pathogenic autoimmune mechanisms involve the production and release of autoreactive antibodies from B cells. Autoantibodies-ie, antibodies that recognize self-antigens-have been identified as potential molecules that can modulate the function of nociceptive neurons and thereby induce persistent pain. Autoantibodies can influence neuronal excitability by activating the complement pathway; by directly signaling at sensory neurons expressing Fc gamma receptors, the receptors for the Fc fragment of immunoglobulin G immune complexes; or by binding and disrupting ion channels expressed by nociceptors. Using examples primarily from rheumatoid arthritis, complex regional pain syndrome, and channelopathies from potassium channel complex autoimmunity, we suggest that autoantibody signaling at the central nervous system has therapeutic implications for designing novel disease-modifying treatments for chronic pain.
Collapse
Affiliation(s)
- Michael J. Lacagnina
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linda R. Watkins
- Department of Psychology and Neuroscience, Center for Neuroscience, University of Colorado, Boulder, CO, USA
| | - Peter M. Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
23
|
Sivanesan E, Goebel A. Complex regional pain syndrome: developing diagnostic tools and treatments from sympathetic nervous system, neuroimmune and neuromodulation discoveries in neuropathic pain. Reg Anesth Pain Med 2021; 46:193-195. [PMID: 33419876 DOI: 10.1136/rapm-2020-102327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/17/2020] [Accepted: 12/22/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Eellan Sivanesan
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andreas Goebel
- Pain Research Institute, University of Liverpool and Department of Pain Medicine, Walton Centre NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
24
|
Renthal W, Tochitsky I, Yang L, Cheng YC, Li E, Kawaguchi R, Geschwind DH, Woolf CJ. Transcriptional Reprogramming of Distinct Peripheral Sensory Neuron Subtypes after Axonal Injury. Neuron 2020; 108:128-144.e9. [PMID: 32810432 PMCID: PMC7590250 DOI: 10.1016/j.neuron.2020.07.026] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/27/2020] [Accepted: 07/22/2020] [Indexed: 12/27/2022]
Abstract
Primary somatosensory neurons are specialized to transmit specific types of sensory information through differences in cell size, myelination, and the expression of distinct receptors and ion channels, which together define their transcriptional and functional identity. By profiling sensory ganglia at single-cell resolution, we find that all somatosensory neuronal subtypes undergo a similar transcriptional response to peripheral nerve injury that both promotes axonal regeneration and suppresses cell identity. This transcriptional reprogramming, which is not observed in non-neuronal cells, resolves over a similar time course as target reinnervation and is associated with the restoration of original cell identity. Injury-induced transcriptional reprogramming requires ATF3, a transcription factor that is induced rapidly after injury and necessary for axonal regeneration and functional recovery. Our findings suggest that transcription factors induced early after peripheral nerve injury confer the cellular plasticity required for sensory neurons to transform into a regenerative state.
Collapse
Affiliation(s)
- William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd., Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA.
| | - Ivan Tochitsky
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Lite Yang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Rd., Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Yung-Chih Cheng
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Emmy Li
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Cir., Boston, MA 02115, USA.
| |
Collapse
|
25
|
Tavares-Ferreira D, Ray PR, Sankaranarayanan I, Mejia GL, Wangzhou A, Shiers S, Uttarkar R, Megat S, Barragan-Iglesias P, Dussor G, Akopian AN, Price TJ. Sex Differences in Nociceptor Translatomes Contribute to Divergent Prostaglandin Signaling in Male and Female Mice. Biol Psychiatry 2020; 91:129-140. [PMID: 33309016 PMCID: PMC8019688 DOI: 10.1016/j.biopsych.2020.09.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND There are clinically relevant sex differences in acute and chronic pain mechanisms, but we are only beginning to understand their mechanistic basis. Transcriptome analyses of rodent whole dorsal root ganglion (DRG) have revealed sex differences, mostly in immune cells. We examined the transcriptome and translatome of the mouse DRG with the goal of identifying sex differences. METHODS We used translating ribosome affinity purification sequencing and behavioral pharmacology to test the hypothesis that in Nav1.8-positive neurons, most of which are nociceptors, translatomes would differ by sex. RESULTS We found 80 genes with sex differential expression in the whole DRG transcriptome and 66 genes whose messenger RNAs were sex differentially actively translated (translatome). We also identified different motifs in the 3' untranslated region of messenger RNAs that were sex differentially translated. In further validation studies, we focused on Ptgds, which was increased in the translatome of female mice. The messenger RNA encodes the prostaglandin PGD2 synthesizing enzyme. We observed increased PTGDS protein and PGD2 in female mouse DRG. The PTGDS inhibitor AT-56 caused intense pain behaviors in male mice but was only effective at high doses in female mice. Conversely, female mice responded more robustly to another major prostaglandin, PGE2, than did male mice. PTGDS protein expression was also higher in female cortical neurons, suggesting that DRG findings may be generalizable to other nervous system structures. CONCLUSIONS Our results demonstrate sex differences in nociceptor-enriched translatomes and reveal unexpected sex differences in one of the oldest known nociceptive signaling molecule families, the prostaglandins.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Pradipta R. Ray
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | | | - Galo L. Mejia
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Ruta Uttarkar
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Salim Megat
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | | | - Gregory Dussor
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Armen N. Akopian
- University of Texas Health San Antonio, Department of Endodontics
| | - Theodore J. Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies,correspondence to Theodore J Price – , 800 W Campbell Rd, Richardson TX 75080, USA, 972-883-4311
| |
Collapse
|
26
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. Brain Behav Immun 2020; 89:559-568. [PMID: 32497778 PMCID: PMC7263237 DOI: 10.1016/j.bbi.2020.05.078] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We hypothesized that SARS-CoV-2 infection drives changes in immune cell-derived factors that then interact with receptors expressed by the sensory neuronal innervation of the lung to further promote important aspects of disease severity, including ARDS. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 pulmonary disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Muhammad S Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Juliet M Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Amelia J McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Sanjay V Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Michael D Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Neuroimmunology and Behavior Research Group, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| |
Collapse
|
27
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. SSRN 2020:3581446. [PMID: 32714114 PMCID: PMC7366818 DOI: 10.2139/ssrn.3581446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/04/2020] [Indexed: 11/15/2022]
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Muhammad S. Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Juliet M. Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Amelia J. McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Sanjay V. Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Michael D. Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|