1
|
Du J, Yang Y, Rao J, Ma X, Tang S, Liu J, Liu Y, Liu S, Li G, Liang S, Gao Y. Resident Macrophages in the Cervical Sympathetic Ganglia Participate in P2Y12 Receptor Mediated Diabetic Cardiac Autonomic Neuropathy. Mol Neurobiol 2025:10.1007/s12035-025-04883-9. [PMID: 40146499 DOI: 10.1007/s12035-025-04883-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Diabetic cardiac autonomic neuropathy (DCAN) represents a significant and prevalent complication of diabetes. Further research is required to ascertain the role of the P2Y12 receptor, which is expressed on macrophages and satellite glial cells (SGCs), in the pathophysiology of DCAN. The objective of this study was to ascertain whether resident macrophages in the superior cervical ganglion (SCG) are involved in the pathological changes associated with DCAN, which are mediated by the P2Y12 receptor in satellite glial cells (SGCs). The findings showed that DCAN rats had higher sympathetic nerve discharge activity than the control group. Furthermore, the expression of P2Y12 receptor, glial fibrillary acidic protein (GFAP), macrophage-like targets (colony-stimulating factor 1 receptor (CSF1R), colony-stimulating factor 1 (CSF1)), and interleukin-34 (IL-34) in SCG among DCAN rats was clearly elevated. Moreover, co-expression levels of NeuN and CSF1 in neurons, P2Y12 and GFAP as well as P2Y12 and IBA-1 in SCGs were increased. However, treatment with P2Y12 shRNA led to significant reductions in all above parameters. The action mechanism may involve reducing the expression of P2Y12 receptors in macrophages and SGCs, decreasing the expression of CSF1 in SCG neurons to weaken the CSF1-CSF1R signal, inhibiting the activation of macrophages and SGCs, and reducing the release of inflammatory factors. This ultimately alleviated abnormal neuronal excitation in SCG and maintaining balance in cardiac autonomic nervous activity. Therefore, targeting the P2Y12 receptor to disrupt the resident macrophages participate in pathological changes, may be an effective approach for improving DCAN.
Collapse
Affiliation(s)
- Junpei Du
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yuxin Yang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jingan Rao
- Second Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaoqian Ma
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Shanshan Tang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jian Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yeqing Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Shipan Liu
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Guodong Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Shangdong Liang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Yun Gao
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Shimizu K, Yasukawa T, Izumida T, Ozasa K, Takizawa K, Noma N, Hayashi M, Takeichi O. Mirogabalin as a Therapeutic Option for Neuropathic Pain Emerging Postendodontic Treatment: An Observational Study. J Endod 2025; 51:279-286. [PMID: 39736439 DOI: 10.1016/j.joen.2024.12.015] [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: 10/18/2024] [Revised: 12/18/2024] [Accepted: 12/22/2024] [Indexed: 01/01/2025]
Abstract
INTRODUCTION We have recently reported the clinical efficacy of mirogabalin for occlusal pain due to post-traumatic trigeminal neuropathic pain (PTTN-occlusal pain) after endodontic treatment according to the International Classification of Orofacial Pain criteria. This study aimed to determine the mirogabalin administration period and timing of dose reduction and suspension for treating this condition based on managing a certain number of cases. METHODS Patients diagnosed with PTTN-occlusal pain after or during endodontic treatment were included in the study. An endodontist retreated if necessary. Patients who initially experienced occlusal pain but gradually adapted to bite well without medication were classified into the provisional restoration group (n = 13). Patients who experienced severe pain during mastication and could not chew with a provisional restoration were classified into the mirogabalin medication group (n = 15). Mirogabalin (10 mg/day) was prescribed once a day before bedtime. The patients receiving visual analog scale (VAS) were instructed to record daily changes in pain intensity in a VAS diary. The practitioners also recorded scores during consultations. RESULTS In the provisional restoration group, VAS scores significantly decreased from week 6 and plateaued from week 11 (P < .05). This adaptation period was applied to reduction or discontinuation of mirogabalin treatment in the mirogabalin medication group. After initiating mirogabalin, VAS scores significantly decreased gradually from week 1 (P < .05). Although the dose was tapered around week 7 and discontinued by week 11, no recurrence of pain was observed following dose reduction or discontinuation. CONCLUSIONS Mirogabalin was effective in treating PTTN-occlusal pain.
Collapse
Affiliation(s)
- Kohei Shimizu
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan.
| | - Takuya Yasukawa
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Takuro Izumida
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Kana Ozasa
- Department of Oral Medicine, Nihon University School of Dentistry, Tokyo, Japan; Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Keita Takizawa
- Department of Oral Medicine, Nihon University School of Dentistry, Tokyo, Japan
| | - Noboru Noma
- Department of Oral Medicine, Nihon University School of Dentistry, Tokyo, Japan; Division of Clinical Research, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Makoto Hayashi
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Osamu Takeichi
- Department of Endodontics, Nihon University School of Dentistry, Tokyo, Japan; Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
3
|
Yi Z, Si H, Liang S, Li G, Dang Y, Zhou C. P2Y12-mediated HIV gp120 and ddC-induced neuropathic pain improved by esculin. Neuroreport 2025; 36:117-126. [PMID: 39976049 DOI: 10.1097/wnr.0000000000002125] [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] [Indexed: 02/21/2025]
Abstract
We studied whether esculin (ES) has the effect of alleviating peripheral neuropathic pain (NP) in rat models of HIV glycoprotein 120 (gp120) together with zalcitabine (2',3'-dideoxycytidine; ddC) treatment and explored the possible mechanism of it. The rats pain behaviors were evaluated by observing the paw withdrawal threshold (PWT) and the paw withdrawal latency (PWL). The rats were divided into a control group, sham group, gp120 combined with a ddC treatment group (gp120& ddC group), gp120&ddC combined with ES treatment group (gp120&ddC+ES group), which ES was administered intragastrically, and gp120&ddC combined with short hair RNA of P2Y12 receptor (rP2Y12) treatment group (gp120&ddC+shP2Y12 group), which shRNA of rP2Y12 was injected intrathecally with a dose of 25 µg/20 µl for every rat, and a negative control plasmid was administered to the gp120&ddC+nc group. Western blotting was used to measure the protein expression levels of the rP2Y12, the nuclear factor of activated T-cells type c1 (NFATc1), phospho-NFATc1 and the C-C motif chemokine ligand 3 (CCL3) in the L4-L6 dorsal root ganglia (DRG). Real-time quantitative PCR (RT-qPCR) was used to test the mRNA expression level of the CCL3. Double-labeling immunofluorescence was used to identify the co-localization of the rP2Y12 with glial fibrillary acidic protein (GFAP) in DRG. Fluorescence imaging with calcium indicator fluo-3 AM (7.5 μM) was performed to observe the change of intracellular calcium concentration ([Ca2+]i). Molecular docking was performed to identify the interaction between rP2Y12 and the ligand ES. We found that accompanied by the attenuation of mechanical allodynia and thermal hyperalgesia, rP2Y12 expression in the gp120+ddC+ES group of rats was downregulated compared with the gp120+ddC ones, as was the coexpression of the rP2Y12 and GFAP of satellite glial cells (SGCs) in DRG, and the CCL3 mRNA levels and protein expression were both decreased. In addition, mechanistic studies have found that there is a docking pocket between ES and the rP2Y12 protein, which causes ES to decrease the [Ca2+]i, thus increasing the phosphorylation level of NFATc1. Taken together, the results suggest that ES can combine with the rP2Y12, inhibit DRG SGCs activation caused by gp120&ddC, reduce [Ca2+]i, and prevent the NFATc1-mediated gene transcription of CCL3, finally relieving NP in rats treated with gp120&ddC.
Collapse
Affiliation(s)
- Zhihua Yi
- School of Nursing, Jiangxi Medical College, Nanchang University
- Jiangxi Provincial Key Laboratory of autonomic nervous function and disease
| | - Han Si
- School of Nursing, Jiangxi Medical College, Nanchang University
- Jiangxi Provincial Key Laboratory of autonomic nervous function and disease
| | - Shangdong Liang
- Jiangxi Provincial Key Laboratory of autonomic nervous function and disease
- Department of Physiology
| | - Guilin Li
- Jiangxi Provincial Key Laboratory of autonomic nervous function and disease
- Department of Physiology
| | | | - Congfa Zhou
- Department of Anatomy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
4
|
Birren SJ, Goodrich LV, Segal RA. Satellite Glial Cells: No Longer the Most Overlooked Glia. Cold Spring Harb Perspect Biol 2025; 17:a041367. [PMID: 38768970 PMCID: PMC11694750 DOI: 10.1101/cshperspect.a041367] [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] [Indexed: 05/22/2024]
Abstract
Many glial biologists consider glia the neglected cells of the nervous system. Among all the glia of the central and peripheral nervous system, satellite glia may be the most often overlooked. Satellite glial cells (SGCs) are located in ganglia of the cranial nerves and the peripheral nervous system. These small cells surround the cell bodies of neurons in the trigeminal ganglia (TG), spiral ganglia, nodose and petrosal ganglia, sympathetic ganglia, and dorsal root ganglia (DRG). Essential SGC features include their intimate connections with the associated neurons, their small size, and their derivation from neural crest cells. Yet SGCs also exhibit tissue-specific properties and can change rapidly, particularly in response to injury. To illustrate the range of SGC functions, we will focus on three types: those of the spiral, sympathetic, and DRG, and consider both their shared features and those that differ based on location.
Collapse
Affiliation(s)
- Susan J Birren
- Department of Biology, Brandeis University, Waltham, Massachusetts 02453, USA
| | - Lisa V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Rosalind A Segal
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
5
|
Scheuren PS, Calvo M. Exploring neuroinflammation: A key driver in neuropathic pain disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:311-338. [PMID: 39580216 DOI: 10.1016/bs.irn.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Inflammation is a fundamental part of the body's natural defense mechanism, involving immune cells and inflammatory mediators to promote healing and protect against harm. In the event of a lesion or disease of the somatosensory nervous system, inflammation, however, triggers a cascade of changes in both the peripheral and central nervous systems, ultimately contributing to chronic neuropathic pain. Substantial evidence links neuroinflammation to various conditions associated with neuropathic pain. This chapter will explore the role of neuroinflammation in the initiation, maintenance, and resolution of peripheral and central neuropathic pain. Additionally, biomarkers of neuroinflammation in humans will be examined, emphasizing their relevance in different neuropathic pain disorders.
Collapse
Affiliation(s)
- Paulina S Scheuren
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, BC, Canada; Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Margarita Calvo
- Physiology Department, Pontificia Universidad Católica de Chile, Santiago, Chile; Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| |
Collapse
|
6
|
Starobova H, Alshammari A, Winkler IG, Vetter I. The role of the neuronal microenvironment in sensory function and pain pathophysiology. J Neurochem 2024; 168:3620-3643. [PMID: 36394416 DOI: 10.1111/jnc.15724] [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: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022]
Abstract
The high prevalence of pain and the at times low efficacy of current treatments represent a significant challenge to healthcare systems worldwide. Effective treatment strategies require consideration of the diverse pathophysiologies that underlie various pain conditions. Indeed, our understanding of the mechanisms contributing to aberrant sensory neuron function has advanced considerably. However, sensory neurons operate in a complex dynamic microenvironment that is controlled by multidirectional interactions of neurons with non-neuronal cells, including immune cells, neuronal accessory cells, fibroblasts, adipocytes, and keratinocytes. Each of these cells constitute and control the microenvironment in which neurons operate, inevitably influencing sensory function and the pathology of pain. This review highlights the importance of the neuronal microenvironment for sensory function and pain, focusing on cellular interactions in the skin, nerves, dorsal root ganglia, and spinal cord. We discuss the current understanding of the mechanisms by which neurons and non-neuronal cells communicate to promote or resolve pain, and how this knowledge could be used for the development of mechanism-based treatments.
Collapse
Affiliation(s)
- Hana Starobova
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ammar Alshammari
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Ingrid G Winkler
- Mater Research Institute, The University of Queensland, Queensland, South Brisbane, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- The School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
7
|
Álvarez-Tosco K, González-Fernández R, González-Nicolás MÁ, Martín-Ramírez R, Morales M, Gutiérrez R, Díaz-Flores L, Arnau MR, Machín F, Ávila J, Lázaro A, Martín-Vasallo P. Dorsal root ganglion inflammation by oxaliplatin toxicity: DPEP1 as possible target for peripheral neuropathy prevention. BMC Neurosci 2024; 25:44. [PMID: 39278931 PMCID: PMC11403972 DOI: 10.1186/s12868-024-00891-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Peripheral neuropathy (PN) constitutes a dose-limiting side effect of oxaliplatin chemotherapy that often compromises the efficacy of antineoplastic treatments. Sensory neurons damage in dorsal root ganglia (DRG) are the cellular substrate of PN complex molecular origin. Dehydropeptidase-1 (DPEP1) inhibitors have shown to avoid platin-induced nephrotoxicity without compromising its anticancer efficiency. The objective of this study was to describe DPEP1 expression in rat DRG in health and in early stages of oxaliplatin toxicity. To this end, we produced and characterized anti-DPEP1 polyclonal antibodies and used them to define the expression, and cellular and subcellular localization of DPEP1 by immunohistochemical confocal microscopy studies in healthy controls and short term (six days) oxaliplatin treated rats. RESULTS DPEP1 is expressed mostly in neurons and in glia, and to a lesser extent in endothelial cells. Rats undergoing oxaliplatin treatment developed allodynia. TNF-𝛼 expression in DRG revealed a pattern of focal and at different intensity levels of neural cell inflammatory damage, accompanied by slight variations in DPEP1 expression in endothelial cells and in nuclei of neurons. CONCLUSIONS DPEP1 is expressed in neurons, glia and endothelial cells of DRG. Oxaliplatin caused allodynia in rats and increased TNF-α expression in DRG neurons. The expression of DPEP1 in neurons and other cells of DRG suggest this protein as a novel strategic molecular target in the prevention of oxaliplatin-induced acute neurotoxicity.
Collapse
Affiliation(s)
- Karen Álvarez-Tosco
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Unidad de Investigación, Hospital Universitario Nuestra Señora de la Candelaria, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain
- Departamento de Farmacia Hospitalaria, Hospital Universitario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Rebeca González-Fernández
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - María Ángeles González-Nicolás
- Laboratorio de Fisiopatología Renal, Departamento de Nefrología, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Rita Martín-Ramírez
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Manuel Morales
- Departamento de Oncología Médica, Hospital Universitario Nuestra Señora de la Candelaria, Santa Cruz de Tenerife, Spain
| | - Ricardo Gutiérrez
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Lucio Díaz-Flores
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - María Rosa Arnau
- Servicio de Estabulario y Animalario del Servicio General de Apoyo a la Investigación (SEGAI), Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Félix Machín
- Unidad de Investigación, Hospital Universitario Nuestra Señora de la Candelaria, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Julio Ávila
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Alberto Lázaro
- Laboratorio de Fisiopatología Renal, Departamento de Nefrología, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain.
| | - Pablo Martín-Vasallo
- Laboratorio de Biología del Desarrollo, UD de Bioquímica y Biología Molecular, Universidad de La Laguna, San Cristóbal de La Laguna, Spain.
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, San Cristóbal de La Laguna, Spain.
| |
Collapse
|
8
|
Goto T, Kuramoto E, Iwai H, Yamanaka A. Cytoarchitecture and intercellular interactions in the trigeminal ganglion: Associations with neuropathic pain in the orofacial region. J Oral Biosci 2024; 66:485-490. [PMID: 39032827 DOI: 10.1016/j.job.2024.07.003] [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: 04/30/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Disorders of the trigeminal nerve, a sensory nerve of the orofacial region, often lead to complications in dental practice, including neuropathic pain, allodynia, and ectopic pain. Management of these complications requires an understanding of the cytoarchitecture of the trigeminal ganglion, where the cell bodies of the trigeminal nerve are located, and the mechanisms of cell-cell interactions. HIGHLIGHTS In the trigeminal ganglion, ganglion, satellite, Schwann, and immune cells coexist and interact. Cell-cell interactions are complex and occur through direct contact via gap junctions or through mediators such as adenosine triphosphate, nitric oxide, peptides, and cytokines. Interactions between the nervous and immune systems within the trigeminal ganglion may have neuroprotective effects during nerve injury or may exacerbate inflammation and produce chronic pain. Under pathological conditions of the trigeminal nerve, cell-cell interactions can cause allodynia and ectopic pain. Although cell-cell interactions that occur via mediators can act at some distance, they are more effective when the cells are close together. Therefore, information on the three-dimensional topography of trigeminal ganglion cells is essential for understanding the pathophysiology of ectopic pain. CONCLUSIONS A three-dimensional map of the somatotopic localization of trigeminal ganglion neurons revealed that ganglion cells innervating distant orofacial regions are often apposed to each other, interacting with and potentially contributing to ectopic pain. Elucidation of the complex network of mediators and their receptors responsible for intercellular communication within the trigeminal ganglion is essential for understanding ectopic pain.
Collapse
Affiliation(s)
- Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Eriko Kuramoto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | - Haruki Iwai
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Atsushi Yamanaka
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
9
|
Bom ADOP, Dias-Soares M, Corrêa RCD, Neves CL, Hosch NG, de Lucena GG, Oliveira CG, Pagano RL, Chacur M, Giorgi R. Molecular Aspects Involved in the Mechanisms of Bothrops jararaca Venom-Induced Hyperalgesia: Participation of NK1 Receptor and Glial Cells. Toxins (Basel) 2024; 16:187. [PMID: 38668612 PMCID: PMC11053884 DOI: 10.3390/toxins16040187] [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: 03/08/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/29/2024] Open
Abstract
Accidents caused by Bothrops jararaca (Bj) snakes result in several local and systemic manifestations, with pain being a fundamental characteristic. The inflammatory process responsible for hyperalgesia induced by Bj venom (Bjv) has been studied; however, the specific roles played by the peripheral and central nervous systems in this phenomenon remain unclear. To clarify this, we induced hyperalgesia in rats using Bjv and collected tissues from dorsal root ganglia (DRGs) and spinal cord (SC) at 2 and 4 h post-induction. Samples were labeled for Iba-1 (macrophage and microglia), GFAP (satellite cells and astrocytes), EGR1 (neurons), and NK1 receptors. Additionally, we investigated the impact of minocycline, an inhibitor of microglia, and GR82334 antagonist on Bjv-induced hyperalgesia. Our findings reveal an increase in Iba1 in DRG at 2 h and EGR1 at 4 h. In the SC, markers for microglia, astrocytes, neurons, and NK1 receptors exhibited increased expression after 2 h, with EGR1 continuing to rise at 4 h. Minocycline and GR82334 inhibited venom-induced hyperalgesia, highlighting the crucial roles of microglia and NK1 receptors in this phenomenon. Our results suggest that the hyperalgesic effects of Bjv involve the participation of microglial and astrocytic cells, in addition to the activation of NK1 receptors.
Collapse
Affiliation(s)
- Ariela de Oliveira Pedro Bom
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
- Postgraduate Program in Toxinology, Butantan Institute, São Paulo 05503-900, SP, Brazil
| | - Monique Dias-Soares
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
| | - Raíssa Cristina Darroz Corrêa
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
- Postgraduate Program in Toxinology, Butantan Institute, São Paulo 05503-900, SP, Brazil
| | - Camila Lima Neves
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
| | | | - Gabriela Gomes de Lucena
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
| | - Camilla Garcia Oliveira
- Laboratory of Functional Neuroanatomy of Pain, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-900, SP, Brazil; (C.G.O.); (M.C.)
| | - Rosana Lima Pagano
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo 01308-060, SP, Brazil;
| | - Marucia Chacur
- Laboratory of Functional Neuroanatomy of Pain, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo 05508-900, SP, Brazil; (C.G.O.); (M.C.)
| | - Renata Giorgi
- Laboratory of Pathophysiology, Butantan Institute, São Paulo 05503-900, SP, Brazil; (A.d.O.P.B.); (M.D.-S.); (R.C.D.C.); (C.L.N.); (G.G.d.L.)
| |
Collapse
|
10
|
Kim HS, Lee D, Shen S. Endoplasmic reticular stress as an emerging therapeutic target for chronic pain: a narrative review. Br J Anaesth 2024; 132:707-724. [PMID: 38378384 PMCID: PMC10925894 DOI: 10.1016/j.bja.2024.01.007] [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: 07/27/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 02/22/2024] Open
Abstract
Chronic pain is a severely debilitating condition with enormous socioeconomic costs. Current treatment regimens with nonsteroidal anti-inflammatory drugs (NSAIDs), steroids, or opioids have been largely unsatisfactory with uncertain benefits or severe long-term side effects. This is mainly because chronic pain has a multifactorial aetiology. Although conventional pain medications can alleviate pain by keeping several dysfunctional pathways under control, they can mask other underlying pathological causes, ultimately worsening nerve pathologies and pain outcome. Recent preclinical studies have shown that endoplasmic reticulum (ER) stress could be a central hub for triggering multiple molecular cascades involved in the development of chronic pain. Several ER stress inhibitors and unfolded protein response modulators, which have been tested in randomised clinical trials or apprpoved by the US Food and Drug Administration for other chronic diseases, significantly alleviated hyperalgesia in multiple preclinical pain models. Although the role of ER stress in neurodegenerative disorders, metabolic disorders, and cancer has been well established, research on ER stress and chronic pain is still in its infancy. Here, we critically analyse preclinical studies and explore how ER stress can mechanistically act as a central node to drive development and progression of chronic pain. We also discuss therapeutic prospects, benefits, and pitfalls of using ER stress inhibitors and unfolded protein response modulators for managing intractable chronic pain. In the future, targeting ER stress to impact multiple molecular networks might be an attractive therapeutic strategy against chronic pain refractory to steroids, NSAIDs, or opioids. This novel therapeutic strategy could provide solutions for the opioid crisis and public health challenge.
Collapse
Affiliation(s)
- Harper S Kim
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donghwan Lee
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shiqian Shen
- Department of Anesthesiology, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Delibaş B, Vianney JM, Kaplan S. The assessment of neuronal plasticity following sciatic nerve injuries in rats using electron microscopy and stereological methods. J Chem Neuroanat 2024; 136:102396. [PMID: 38331230 DOI: 10.1016/j.jchemneu.2024.102396] [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: 12/18/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024]
Abstract
The transmission of signals to the cell body from injured axons induces significant alterations in primary sensory neurons located in the ganglion tissue, the site of the perikaryon of the affected nerve fibers. Disruption of the continuity between the proximal and distal ends leads to substantial adaptability in ganglion cells and induces macrophage-like activity in the satellite cells. Research findings have demonstrated the plasticity of satellite cells following injury. Satellite cells work together with sensory neurons to extend the interconnected surface area in order to permit effective communication. The dynamic cellular environment within the ganglion undergoes several alterations that ultimately lead to differentiation, transformation, or cell death. In addition to necrotic and apoptotic cell morphology, phenomena such as histomorphometric alterations, including the development of autophagic vacuoles, chromatolysis, cytosolic degeneration, and other changes, are frequently observed in cells following injury. The use of electron microscopic and stereological techniques for assessing ganglia and nerve fibers is considered a gold standard in terms of investigating neuropathic pain models, regenerative therapies, some treatment methods, and quantifying the outcomes of pharmacological and bioengineering interventions. Stereological techniques provide observer-independent and reliable results, which are particularly useful in the quantitative assessment of three-dimensional structures from two-dimensional images. Employing the fractionator and disector techniques within stereological methodologies yields unbiased data when assessing parameters such as number. The fundamental concept underlying these methodologies involves ensuring that each part of the structure under evaluation has an equal opportunity of being sampled. This review describes the stereological and histomorphometric evaluation of dorsal root ganglion neurons and satellite cells following nerve injury models.
Collapse
Affiliation(s)
- Burcu Delibaş
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkiye
| | - John-Mary Vianney
- School of Life Science and Bioengineering, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania
| | - Süleyman Kaplan
- School of Life Science and Bioengineering, Nelson Mandela-African Institution of Science and Technology, Arusha, Tanzania; Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkiye.
| |
Collapse
|
12
|
Fan XX, Ye L, Yang YH, Huang WJ, Ko CY. Migraine Duration as a Potential Amplifier of Obesity. Diabetes Metab Syndr Obes 2024; 17:1025-1037. [PMID: 38476349 PMCID: PMC10928920 DOI: 10.2147/dmso.s447781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose Migraine is a complex neurovascular disorder with obesity as a notable risk factor. This study aimed to investigate an under-researched area of the association between migraine duration and body composition. Patients and Methods Patients with migraine from a neurology outpatient department were enrolled and were categorized into four groups based on illness duration: 1 year, 1-5 years, 5-10 years, and >10 years. Patient demographics, blood biochemistry, and body composition data were collected and analyzed statistically. Results Patients with migraine were predominantly female, with lower education levels, significant work stress, poor sleep, and limited exercise. Longer migraine duration corresponded to increased obesity metrics. Notably, those patients with under 1 year of illness showed elevated blood lipid and liver function levels, whereas those with >10 years showed increased weight, waist circumference, body mass index, and fat content, despite higher physical activity. Significant positive correlation between obesity metrics and migraine duration was seen in patients who had migraine for >1 year. Conclusion Our findings indicate that protracted episodes of migraine could amplify obesity tendencies, underscoring the imperative of weight regulation in migraine intervention to diminish ensuing adiposity-associated hazards.
Collapse
Affiliation(s)
- Xi-Xin Fan
- The School of Public Health, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People’s Republic of China
| | - Lichao Ye
- Department of Neurology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People’s Republic of China
| | - Ya-Hui Yang
- The School of Public Health, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People’s Republic of China
| | - Wen-Jian Huang
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People’s Republic of China
- Huidong Center for Chronic Disease Control, Huizhou, People’s Republic of China
| | - Chih-Yuan Ko
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, People’s Republic of China
| |
Collapse
|
13
|
Wood RM, Vasquez EL, Goyins KA, Gutierrez Kuri E, Connelly K, Humayun S, Macpherson LJ. Cyclophosphamide induces the loss of taste bud innervation in mice. Chem Senses 2024; 49:bjae010. [PMID: 38421250 PMCID: PMC10929424 DOI: 10.1093/chemse/bjae010] [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: 10/06/2023] [Indexed: 03/02/2024] Open
Abstract
Many common chemotherapeutics produce disruptions in the sense of taste which can lead to loss of appetite, nutritional imbalance, and reduced quality of life, especially if taste loss persists after treatment ends. Cyclophosphamide (CYP), an alkylating chemotherapeutic agent, affects taste sensitivity through its cytotoxic effects on mature taste receptor cells (TRCs) and on taste progenitor cell populations, retarding the capacity to replace TRCs. Mechanistic studies have focused primarily on taste cells, however, taste signaling requires communication between TRCs and the gustatory nerve fibers that innervate them. Here, we evaluate cyclophosphamide's effects on the peripheral gustatory nerve fibers that innervate the taste buds. Following histological analysis of tongue tissues, we find that CYP reduces innervation within the fungiform and circumvallates taste buds within 4 days after administration. To better understand the dynamics of the denervation process, we used 2-photon intravital imaging to visualize the peripheral gustatory nerve fibers within individual fungiform taste buds up to 20 days after CYP treatment. We find that gustatory fibers retract from the taste bud properly but are maintained within the central papilla core. These data indicate that in addition to TRCs, gustatory nerve fibers are also affected by CYP treatment. Because the connectivity between TRCs and gustatory neurons must be re-established for proper function, gustatory fibers should continue to be included in future studies to understand the mechanisms leading to chemotherapy-induced persistent taste loss.
Collapse
Affiliation(s)
- Ryan M Wood
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
- The Graduate Program in Neuroscience, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Erin L Vasquez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Krystal A Goyins
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
- The Graduate Program in Developmental and Regenerative Sciences, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Eduardo Gutierrez Kuri
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Kevin Connelly
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Saima Humayun
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Lindsey J Macpherson
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
- Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
14
|
Zamith Cunha R, Semprini A, Salamanca G, Gobbo F, Morini M, Pickles KJ, Roberts V, Chiocchetti R. Expression of Cannabinoid Receptors in the Trigeminal Ganglion of the Horse. Int J Mol Sci 2023; 24:15949. [PMID: 37958932 PMCID: PMC10648827 DOI: 10.3390/ijms242115949] [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: 09/10/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Cannabinoid receptors are expressed in human and animal trigeminal sensory neurons; however, the expression in the equine trigeminal ganglion is unknown. Ten trigeminal ganglia from five horses were collected post-mortem from an abattoir. The expression of cannabinoid receptors type 1 (CB1R) and type 2 (CB2R), and the cannabinoid-related receptors like transient receptor potential vanilloid type 1 (TRPV1), peroxisome proliferator-activated receptor gamma (PPARɣ), and G protein-related receptor 55 (GPR55) in the trigeminal ganglia (TG) of the horse were studied, using immunofluorescence on cryosections and formalin-fixed paraffin-embedded (FFPE) sections. Neurons and glial cells were identified using fluorescent Nissl staining NeuroTrace® and an antibody directed against the glial marker glial fibrillary acidic protein (GFAP), respectively. Macrophages were identified by means of an antibody directed against the macrophages/microglia marker ionized calcium-binding adapter molecule 1 (IBA1). The protein expression of CB1R, CB2R, TRPV1, and PPARɣ was found in the majority of TG neurons in both cryosections and FFPE sections. The expression of GPR55 immunoreactivity was mainly detectable in FFPE sections, with expression in the majority of sensory neurons. Some receptors were also observed in glial cells (CB2R, TRPV1, PPARγ, and GPR55) and inflammatory cells (PPARγ and GPR55). These results support further investigation of such receptors in disorders of equine trigeminal neuronal excitability.
Collapse
Affiliation(s)
- Rodrigo Zamith Cunha
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Alberto Semprini
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Giulia Salamanca
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Maria Morini
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| | - Kirstie J. Pickles
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham LE12 5RD, UK;
| | - Veronica Roberts
- Bristol Vet School, University of Bristol, Bristol BS40 5DU, UK;
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences, University of Bologna, 37200 Bologna, Italy; (R.Z.C.); (A.S.); (G.S.); (F.G.); (M.M.)
| |
Collapse
|
15
|
Kume M, Ahmad A, DeFea KA, Vagner J, Dussor G, Boitano S, Price TJ. Protease-Activated Receptor 2 (PAR2) Expressed in Sensory Neurons Contributes to Signs of Pain and Neuropathy in Paclitaxel Treated Mice. THE JOURNAL OF PAIN 2023; 24:1980-1993. [PMID: 37315729 PMCID: PMC10615692 DOI: 10.1016/j.jpain.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common, dose-limiting side effect of cancer therapy. Protease-activated receptor 2 (PAR2) is implicated in a variety of pathologies, including CIPN. In this study, we demonstrate the role of PAR2 expressed in sensory neurons in a paclitaxel (PTX)-induced model of CIPN in mice. PAR2 knockout/wildtype (WT) mice and mice with PAR2 ablated in sensory neurons were treated with PTX administered via intraperitoneal injection. In vivo behavioral studies were done in mice using von Frey filaments and the Mouse Grimace Scale. We then examined immunohistochemical staining of dorsal root ganglion (DRG) and hind paw skin samples from CIPN mice to measure satellite cell gliosis and intra-epidermal nerve fiber (IENF) density. The pharmacological reversal of CIPN pain was tested with the PAR2 antagonist C781. Mechanical allodynia caused by PTX treatment was alleviated in PAR2 knockout mice of both sexes. In the PAR2 sensory neuronal conditional knockout (cKO) mice, both mechanical allodynia and facial grimacing were attenuated in mice of both sexes. In the DRG of the PTX-treated PAR2 cKO mice, satellite glial cell activation was reduced compared to control mice. IENF density analysis of the skin showed that the PTX-treated control mice had a reduction in nerve fiber density while the PAR2 cKO mice had a comparable skin innervation as the vehicle-treated animals. Similar results were seen with satellite cell gliosis in the DRG, where gliosis induced by PTX was absent in PAR cKO mice. Finally, C781 was able to transiently reverse established PTX-evoked mechanical allodynia. PERSPECTIVE: Our work demonstrates that PAR2 expressed in sensory neurons plays a key role in PTX-induced mechanical allodynia, spontaneous pain, and signs of neuropathy, suggesting PAR2 as a possible therapeutic target in multiple aspects of PTX CIPN.
Collapse
Affiliation(s)
- Moeno Kume
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Ayesha Ahmad
- 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
| | - Scott Boitano
- University of Arizona Bio5 Research Institute
- University of Arizona Heath Sciences, Asthma and Airway Disease Research Center
- University of Arizona Heath Sciences, Department of Physiology
| | - Theodore J. Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| |
Collapse
|
16
|
Jurczak A, Sandor K, Bersellini Farinotti A, Krock E, Hunt MA, Agalave NM, Barbier J, Simon N, Wang Z, Rudjito R, Vazquez-Mora JA, Martinez-Martinez A, Raoof R, Eijkelkamp N, Grönwall C, Klareskog L, Jimenéz-Andrade JM, Marchand F, Svensson CI. Insights into FcγR involvement in pain-like behavior induced by an RA-derived anti-modified protein autoantibody. Brain Behav Immun 2023; 113:212-227. [PMID: 37437817 DOI: 10.1016/j.bbi.2023.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/21/2023] [Accepted: 07/05/2023] [Indexed: 07/14/2023] Open
Abstract
Joint pain is one of the most debilitating symptoms of rheumatoid arthritis (RA) and patients frequently rate improvements in pain management as their priority. RA is hallmarked by the presence of anti-modified protein autoantibodies (AMPA) against post-translationally modified citrullinated, carbamylated and acetylated proteins. It has been suggested that autoantibody-mediated processes represent distinct mechanisms contributing to pain in RA. In this study, we investigated the pronociceptive properties of monoclonal AMPA 1325:01B09 (B09 mAb) derived from the plasma cell of an RA patient. We found that B09 mAb induces pain-like behavior in mice that is not associated with any visual, histological or transcriptional signs of inflammation in the joints, and not alleviated by non-steroidal anti-inflammatory drugs (NSAIDs). Instead, we found that B09 mAb is retained in dorsal root ganglia (DRG) and alters the expression of several satellite glia cell (SGC), neuron and macrophage-related factors in DRGs. Using mice that lack activating FcγRs, we uncovered that FcγRs are critical for the development of B09-induced pain-like behavior, and partially drive the transcriptional changes in the DRGs. Finally, we observed that B09 mAb binds SGC in vitro and in combination with external stimuli like ATP enhances transcriptional changes and protein release of pronociceptive factors from SGCs. We propose that certain RA antibodies bind epitopes in the DRG, here on SGCs, form immune complexes and activate resident macrophages via FcγR cross-linking. Our work supports the growing notion that autoantibodies can alter nociceptor signaling via mechanisms that are at large independent of local inflammatory processes in the joint.
Collapse
Affiliation(s)
- Alexandra Jurczak
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Alex Bersellini Farinotti
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Emerson Krock
- The Alan Edwards Centre for Research on Pain, Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Matthew A Hunt
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Nilesh M Agalave
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Julie Barbier
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand 38-63001, France
| | - Nils Simon
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Zhenggang Wang
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Resti Rudjito
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden
| | - Juan Antonio Vazquez-Mora
- Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Arisai Martinez-Martinez
- Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Ramin Raoof
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Niels Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Caroline Grönwall
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Lars Klareskog
- Department of Medicine, Division of Rheumatology, Center for Molecular Medicine, Karolinska Institute, Karolinska University Hospital, Stockholm 171 76, Sweden
| | - Juan Miguel Jimenéz-Andrade
- Unidad Academica Multidisciplinaria Reynosa Aztlan, Universidad Autonoma de Tamaulipas, Reynosa, Tamaulipas, Mexico
| | - Fabien Marchand
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand 38-63001, France
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Solnavägen 9, 171 65, Sweden.
| |
Collapse
|
17
|
Zhao L, Liu S, Zhang X, Yang J, Mao M, Zhang S, Xu S, Feng S, Wang X. Satellite glial cell-secreted exosomes after in-vitro oxaliplatin treatment presents a pro-nociceptive effect for dorsal root ganglion neurons and induce mechanical hypersensitivity in naïve mice. Mol Cell Neurosci 2023; 126:103881. [PMID: 37467904 DOI: 10.1016/j.mcn.2023.103881] [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: 03/05/2023] [Revised: 06/29/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND The pathophysiological mechanism underlying chemotherapy-induced neuropathic pain (CINP) remains unclear. Sensory neuronal hypersensitivity in the dorsal root ganglion (DRG) is essential for the onset and maintenance of chronic pain. Satellite glial cells (SGCs) in the DRG potentially affect the function of sensory neurons, possibly by mediating extracellular or paracrine signaling. Exosomes play an essential role in cell-cell communication. However, the role of SGC-secreted exosomes in glia-neuron communication and CINP remains unclear. METHODS SGCs and sensory neurons were cultured from the DRG of mice. The SGCs were treated with 4 μM oxaliplatin for 24 h. Glial fibrillary acid protein (GFAP) and connexin-43 (Cx-43) expressions in the SGCs were examined with immunocytochemistry (ICC). Enzyme-linked immunosorbent assay (ELISA) detected cytokine release in the SGCs after oxaliplatin treatment. Subsequently, SGC-secreted exosomes were collected using ultracentrifugation and identified by nanoparticle tracking analysis, transmission electron microscopy, and western blotting. Subsequently, DRG neurons were incubated with SGC-secreted exosomes for 24 h. The percentage of reactive oxygen species (ROS)-positive neurons was detected using flow cytometry, and acid-sensing ion channel 3 (ASIC3) and transient receptor potential vanilloid 1 (TRPV1) expression were examined by western blotting. SGC-secreted exosomes were intrathecally injected into naïve mice. The mechanical withdrawal threshold was assessed 24, 48, and 72 h following the injection. TRPV1 expression in the DRG was examined 72 h after intrathecal injection. Furthermore, differentially expressed (DE) miRNAs within the SGC-secreted exosomes were detected using RNA sequencing and bioinformatics analysis. Gene Ontology (GO) enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway analyses were performed to predict the function of the target genes of DE miRNAs. Finally, the DE miRNAs with pain regulation potential were identified in silico. RESULTS After in-vitro oxaliplatin treatment, ICC showed an increase in the immunoreactivity of GFAP and Cx-43 in the SGCs. ELISA results suggested an increased release of tumor necrosis factor-α and interleukin (IL)-1β, but a decreased release of IL-10. Oxaliplatin treatment increased the secretion of exosomes in the SGCs from 4.34 to 5.99 × 1011 (particles/ml). The exosome-specific markers CD9 and TSG101 were positive, whereas calnexin was negative for the obtained exosomes. Additionally, the SGC-secreted exosomes were endocytosed by DRG neurons after co-incubation. Moreover, after incubation with conditioned SGC-secreted exosomes (after 4 μM oxaliplatin treatment), the percentage of ROS-positive DRG neurons increased and ASIC3 and TRPV1 expressions were upregulated. After the intrathecal injection of the conditioned SGC-secreted exosomes, the mice presented with mechanical hypersensitivity and TRPV1 expression upregulation in the DRG. Notably, 25 and 120 significantly upregulated and downregulated miRNAs, respectively, were identified in the conditioned SGC-secreted exosomes. When predicting the function of target genes of DE miRNAs, certain GO terms, such as synapse organization, neurogenesis regulation, histone modification, and pain-related KEGG or Reactome pathways, including vascular endothelial growth factor A-vascular endothelial growth factor receptor 2, mammalian target of rapamycin, and mitogen-activated protein kinase signaling pathways, related to nervous system function were predicted. Finally, 27 pain regulation-related miRNAs, including miR-324-3p, miR-181a-5p, and miR-122-5p, were identified in silico. CONCLUSION Our study demonstrates that SGC-secreted exosomes after in-vitro oxaliplatin treatment present a pro-nociceptive effect for DRG neurons and induce mechanical hypersensitivity in naïve mice, possibly via the contained miRNA cargo. Identifying the candidate miRNAs and verifying their functions in vivo are required to elucidate the exosomes mediating 'glia-neuron' communication under CINP condition.
Collapse
Affiliation(s)
- Liping Zhao
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Shijiang Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaobao Zhang
- Department of Anesthesiology, The First People's Hospital of Lianyungang City, Lianyungang, Jiangsu Province, China
| | - Juan Yang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Mao Mao
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Susu Zhang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China
| | - Shiqin Xu
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China.
| | - Shanwu Feng
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China.
| | - Xian Wang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu Province, China.
| |
Collapse
|
18
|
Li L, Du J, Liu S, Yang R, Xu X, Yang Y, Ma X, Li G, Liu S, Li G, Liang S. The potential role of CpG oligodeoxynucleotides on diabetic cardiac autonomic neuropathy mediated by P2Y12 receptor in rat stellate ganglia. Int Immunopharmacol 2023; 119:110044. [PMID: 37264553 DOI: 10.1016/j.intimp.2023.110044] [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: 11/21/2022] [Revised: 02/11/2023] [Accepted: 03/13/2023] [Indexed: 06/03/2023]
Abstract
Cardiac autonomic neuropathy has a high prevalence in type 2 diabetes, which increases the risk of cardiovascular system disorders. CpG oligodeoxynucleotide (CpG-ODN), a Toll-like receptor 9 (TLR9) ligand, has been shown to have cardioprotection and cellular protection. Our previous work showed that P2Y12 in stellate ganglia (SG) is involved in the process of diabetic cardiac autonomic neuropathy (DCAN). Here, we aim to investigate whether CpG-ODN 1826 plays a protective role in DCAN and whether this beneficial protection involves regulation of the P2Y12-mediated cardiac sympathetic injury. Our results revealed that CpG-ODN 1826 activated TLR9 receptor, improved the abnormal blood pressure (BP), heart rate (HR), heart rate variability (HRV) and sympathetic nerve discharge (SND) activity in diabetic rats and reduced the up-regulated NF-κB, P2Y12 receptor, TNF-α and IL-1β in SG. Meanwhile, CpG-ODN 1826 significantly decreased the elevated ATP, nuclear receptor coactivator 4 (NCOA4), iron, ROS and MDA levels and increased GPX4 and GSH levels. In addition, CpG-ODN 1826 contributes to maintain normalization of mitochondrial structure in SG. Overall, CpG-ODN 1826 alleviates the sympathetic excitation and abnormal neuron-glial signal communication via activating TLR9 receptors to achieve a balance of autonomic activity and relieve the DCAN in rats. The mechanism may involve the regulation of P2Y12 receptor in SG by reducing ATP release and NF-κB expression, which counteract neuroinflammation and ferroptosis mediated by activated P2Y12 in SG.
Collapse
Affiliation(s)
- Lin Li
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Junpei Du
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Shipan Liu
- Undergraduate Student at Class 2103, First Clinical Medical College of Nanchang University, Nanchang 330006, PR China
| | - Runan Yang
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Xiumei Xu
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Yuxin Yang
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Xiaoqian Ma
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Guilin Li
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Shuangmei Liu
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Guodong Li
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China
| | - Shangdong Liang
- Neuropharmacology Laboratory of Physiology Department, Medical School of Nanchang University, Nanchang 330006, PR China; Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
19
|
Liu M, Li X, Wang J, Ji Y, Gu J, Wei Y, Peng L, Tian C, Lv P, Wang P, Liu X, Li W. Identification and validation of Rab11a in Rat orofacial inflammatory pain model induced by CFA. Neurochem Int 2023:105550. [PMID: 37268020 DOI: 10.1016/j.neuint.2023.105550] [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: 02/11/2023] [Revised: 04/29/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023]
Abstract
Orofacial pain (OFP) is a clinically very common and the most troubling condition; however, there is few effective way to relieve OFP. Rab11a, a small molecule guanosine triphosphate enzyme, is one of the Rab member family playing a vital role in intracellular endocytosis and the pain process. Therefore, we investigated the hub genes of rat OFP model induced by Complete Freund's Adjuvant (CFA) via re-analyzing microarray data (GSE111160). We found that Rab11a acted as a key hub gene in the process of OFP. During the validation of Rab11a, the OFP model was established by peripheral injection of CFA, which decreased the head withdrawal threshold (HWT) and head withdrawal lantency (HWL). Rab11a was observed in NeuN of Sp5C instead of GFAP/IBA-1, and double-IF of Rab11a and Fos positive cells were increased on the 7th day after CFA modeling statistically. Rab11a protein expression in TG and Sp5C of CFA group was also significantly increased. Interestingly, injection of Rab11a-targeted short hairpin RNA (Rab11a-shRNA) into Sp5C could reverse the decrease in HWT and HWL and reduce the expression level of Rab11a. Electrophysiological recording further demonstrated that the activity of Sp5C neuron was improved in CFA group, while Rab11a-shRNA considerably decreased the enhancement of Sp5C neuronal activity. Finally, we detected the expression level of p-PI3K, p-AKT, and p-mTOR in Sp5C of rats after injecting the Rab11a-shRNA virus. To our surprise, CFA upregulated the phosphorylation of PI3K, AKT and mTOR in Sp5C, and Rab11a-shRNA downregulated these molecules' expression. Our data suggest that CFA activates the PI3K/AKT signaling pathway through up-regulating Rab11a expression, which can induce OFP hyperalgesia development furtherly. Targeting Rab11a may be a novel treatment strategy for OFP.
Collapse
Affiliation(s)
- Miaomiao Liu
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xin Li
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, Shandong, China
| | - Jian Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuanyuan Ji
- Department of Anatomy, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Junxiang Gu
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi Wei
- Department of Anatomy, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Liwei Peng
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Tian
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peiyuan Lv
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peng Wang
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xin Liu
- Department of Stomatology, The 960th Hospital of People's Liberation Army, Jinan, Shandong, China.
| | - Weixin Li
- Department of Neurosurgery, Tangdu Hospital of the Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
20
|
McGinnis A, Ji RR. The Similar and Distinct Roles of Satellite Glial Cells and Spinal Astrocytes in Neuropathic Pain. Cells 2023; 12:965. [PMID: 36980304 PMCID: PMC10047571 DOI: 10.3390/cells12060965] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Preclinical studies have identified glial cells as pivotal players in the genesis and maintenance of neuropathic pain after nerve injury associated with diabetes, chemotherapy, major surgeries, and virus infections. Satellite glial cells (SGCs) in the dorsal root and trigeminal ganglia of the peripheral nervous system (PNS) and astrocytes in the central nervous system (CNS) express similar molecular markers and are protective under physiological conditions. They also serve similar functions in the genesis and maintenance of neuropathic pain, downregulating some of their homeostatic functions and driving pro-inflammatory neuro-glial interactions in the PNS and CNS, i.e., "gliopathy". However, the role of SGCs in neuropathic pain is not simply as "peripheral astrocytes". We delineate how these peripheral and central glia participate in neuropathic pain by producing different mediators, engaging different parts of neurons, and becoming active at different stages following nerve injury. Finally, we highlight the recent findings that SGCs are enriched with proteins related to fatty acid metabolism and signaling such as Apo-E, FABP7, and LPAR1. Targeting SGCs and astrocytes may lead to novel therapeutics for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
21
|
Biscetti L, Cresta E, Cupini LM, Calabresi P, Sarchielli P. The putative role of neuroinflammation in the complex pathophysiology of migraine: From bench to bedside. Neurobiol Dis 2023; 180:106072. [PMID: 36907522 DOI: 10.1016/j.nbd.2023.106072] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/18/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
The implications of neurogenic inflammation and neuroinflammation in the pathophysiology of migraine have been clearly demonstrated in preclinical migraine models involving several sites relevant in the trigemino-vascular system, including dural vessels and trigeminal endings, the trigeminal ganglion, the trigeminal nucleus caudalis as well as central trigeminal pain processing structures. In this context, a relevant role has been attributed over the years to some sensory and parasympathetic neuropeptides, in particular calcitonin gene neuropeptide, vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Several preclinical and clinical lines of evidence also support the implication of the potent vasodilator and messenger molecule nitric oxide in migraine pathophysiology. All these molecules are involved in vasodilation of the intracranial vasculature, as well as in the peripheral and central sensitization of the trigeminal system. At meningeal level, the engagement of some immune cells of innate immunity, including mast-cells and dendritic cells, and their mediators, has been observed in preclinical migraine models of neurogenic inflammation in response to sensory neuropeptides release due to trigemino-vascular system activation. In the context of neuroinflammatory events implicated in migraine pathogenesis, also activated glial cells in the peripheral and central structures processing trigeminal nociceptive signals seem to play a relevant role. Finally, cortical spreading depression, the pathophysiological substrate of migraine aura, has been reported to be associated with inflammatory mechanisms such as pro-inflammatory cytokine upregulation and intracellular signalling. Reactive astrocytosis consequent to cortical spreading depression is linked to an upregulation of these inflammatory markers. The present review summarizes current findings on the roles of immune cells and inflammatory responses in the pathophysiology of migraine and their possible exploitation in the view of innovative disease-modifying strategies.
Collapse
Affiliation(s)
- Leonardo Biscetti
- Istituto Nazionale di Ricovero e Cura dell'Anziano a carattere scientifico, IRCCS-INRCA, Ancona, Italy.
| | - Elena Cresta
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Paolo Calabresi
- Department of Neuroscience, Università Cattolica Sacro Cuore, Rome, Italy; Neurologia, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Paola Sarchielli
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
22
|
Abstract
Satellite glial cells (SGCs) that surround sensory neurons in the peripheral nervous system ganglia originate from neural crest cells. Although several studies have focused on SGCs, the origin and characteristics of SGCs are unknown, and their lineage remains unidentified. Traditionally, it has been considered that SGCs regulate the environment around neurons under pathological conditions, and perform functions of supporting, nourishing, and protecting neurons. However, recent studies demonstrated that SGCs may have the characteristics of stem cells. After nerve injury, SGCs up-regulate the expression of stem cell markers and can differentiate into functional sensory neurons. Moreover, SGCs express several markers of Schwann cell precursors and Schwann cells, such as CDH19, MPZ, PLP1, SOX10, ERBB3, and FABP7. Schwann cell precursors have also been proposed as a potential source of neurons in the peripheral nervous system. The similarity in function and markers suggests that SGCs may represent a subgroup of Schwann cell precursors. Herein, we discuss the roles and functions of SGCs, and the lineage relationship between SGCs and Schwann cell precursors. We also describe a new perspective on the roles and functions of SGCs. In the DRG located on the posterior root of spinal nerves, satellite glial cells wrap around each sensory neuron to form an anatomically and functionally distinct unit with the sensory neurons. Following nerve injury, satellite glial cells up-regulate the expression of progenitor markers, and can differentiate into neurons.
Collapse
|
23
|
de Sousa LM, de Figueiredo Costa AC, Pereira AF, da Silva Martins C, de Oliveira Filho OV, Goes P, Vale ML, Gondim DV. Temporomandibular joint arthritis increases canonical Wnt pathway expression in the articular cartilage and trigeminal ganglion in rats. Bone Rep 2023; 18:101649. [PMID: 36700243 PMCID: PMC9869417 DOI: 10.1016/j.bonr.2022.101649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/27/2022] [Accepted: 12/16/2022] [Indexed: 01/18/2023] Open
Abstract
The canonical Wnt pathway participates in inflammatory diseases and it is involved in neuropathic pain. This study evaluated the immunoexpression of the canonical Wnt signaling pathway in the articular cartilage of the temporomandibular joint (TMJ) and along the nociceptive trigeminal pathway in arthritic rats. For this, male Wistar rats were divided into Control (C) and Arthritic (RA) groups. Arthritis induction was performed through subcutaneous injection of methylated bovine serum albumin (mBSA) and complete Freund Adjuvant (CFA)/ Incomplete Freund Adjuvant (IFA) on the first 14 days (once a week), followed by 3 weekly intra-articular injections of mBSA (10 μl/joint; left TMJ). The following parameters were evaluated: nociceptive threshold, inflammatory infiltrate, type I and III collagen birefringence, immunohistochemistry for IL-1β, TNF-α, IL-6, Wnt10b, β-catenin, cyclin-D1 in articular cartilage, c-Myc in synovial membrane, and immunofluorescence analysis for c-Fos, Wnt-10b and β-catenin in the trigeminal ganglion and the trigeminal subnucleus caudalis. The RA group showed intense articular cartilage damage with proliferation of type III collagen, increased immunoexpression of proinflammatory cytokines and Wnt-10b, β-catenin and cyclin-D1 in the articular cartilage and c-Myc in the synovial membrane. In the RA group, a reduction in the nociceptive threshold was observed, followed by a significant increase in the expression of Wnt-10b in neurons and β-catenin in satellite cells of the trigeminal ganglion. c-Fos immunoexpression was observed in neurons, peripherally and centrally, in arthritic rats. Our data demonstrated that TMJ arthritis in rats causes articular cartilage damage and nociceptive behavior, with increased immunoexpression of canonical Wnt pathway in the articular cartilage and trigeminal ganglion.
Collapse
Affiliation(s)
- Luane Macêdo de Sousa
- Postgraduate Program in Morphofunctional Sciences, Faculty of Medicine, Federal University of Ceará, Brazil
| | | | - Anamaria Falcão Pereira
- Postgraduate Program in Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil
| | - Conceição da Silva Martins
- Postgraduate Program in Morphofunctional Sciences, Faculty of Medicine, Federal University of Ceará, Brazil
| | | | - Paula Goes
- Postgraduate Program in Morphofunctional Sciences, Faculty of Medicine, Federal University of Ceará, Brazil
- Postgraduate Program in Dentistry, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Brazil
| | - Mariana Lima Vale
- Postgraduate Program in Morphofunctional Sciences, Faculty of Medicine, Federal University of Ceará, Brazil
- Postgraduate Program in Pharmacology, Faculty of Medicine, Federal University of Ceará, Brazil
| | - Delane Viana Gondim
- Postgraduate Program in Morphofunctional Sciences, Faculty of Medicine, Federal University of Ceará, Brazil
- Postgraduate Program in Dentistry, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Brazil
- Corresponding author at: Department of Morphology, Faculty of Medicine, Federal University of Ceará, Rua Delmiro de Farias, S/N, Rodolfo Teófilo, CEP: 60430-170 Fortaleza, CE, Brazil.
| |
Collapse
|
24
|
Reducha PV, Bömers JP, Edvinsson L, Haanes KA. Rodent behavior following a dural inflammation model with anti-CGRP migraine medication treatment. Front Neurol 2023; 14:1082176. [PMID: 36908624 PMCID: PMC9995475 DOI: 10.3389/fneur.2023.1082176] [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: 10/27/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Background Migraine is a widespread and prevalent disease with a complex pathophysiology, of which neuroinflammation and increased pain sensitivity have been suggested to be involved. Various studies have investigated the presence of different inflammatory markers in migraineurs and investigated the role of inflammation in inflammatory models with complete Freund's adjuvant (CFA) or inflammatory soup added to the dura mater. Objective The aim of the current study was to examine whether application of CFA to the dura mater would cause behavioral alterations that are migraine relevant. In addition, we investigated the potential mitigating effects of fremanezumab, a CGRP (calcitonin gene-related peptide) specific antibody, following CFA application. Methods Male Sprague-Dawley rats were randomly divided into six groups: fresh (n = 7), fresh + carprofen (n = 6), fresh + anti-CGRP (n = 6), sham (n = 7), CFA (n = 16), CFA + anti-CGRP (n = 8). CFA was applied for 15 min on a 3 × 3 mm clearing of the skull exposing the dura mater of male Sprague-Dawley rats. We applied the Light/Dark box and Open Field test, combined with the electronic von Frey test to evaluate outcomes. Finally, we observed CGRP immunoreactivity in the trigeminal ganglion. Results No differences were observed in the Light/Dark box test. The Open Field test detected behavior differences, notably that sham rats spend less time in the central zone, reared less and groomed more than fresh + carprofen rats. The other groups were not significantly different compared to sham rats, indicating that activation of the TGVS is present in sham surgery and cannot be exacerbated by CFA. However, for the allodynia, we observed specific periorbital sensitization, not observed in the sham animals. This could not be mitigated by fremanezumab, although it clearly reduced the amount of CGRP positive fibers. Conclusion CFA surgically administered to the dura causes periorbital allodynia and increases CGRP positive fibers in the trigeminal ganglion. Fremanezumab does not reduce periorbital allodynia even though it reduces CGRP positive fibers in the TG. Further work is needed to investigate whether CFA administered to the dura could be used as a non-CGRP inflammatory migraine model.
Collapse
Affiliation(s)
- Philip V Reducha
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Glostrup, Denmark.,Section of Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jesper P Bömers
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Glostrup, Denmark.,Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Glostrup, Denmark.,Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Kristian A Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Glostrup, Denmark.,Section of Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Yamakita S, Fujita D, Sudo K, Ishikawa D, Kushimoto K, Horii Y, Amaya F. Activation of neurons and satellite glial cells in the DRG produces morphine-induced hyperalgesia. Mol Pain 2023; 19:17448069231181973. [PMID: 37254240 PMCID: PMC10291868 DOI: 10.1177/17448069231181973] [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: 02/09/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 06/01/2023] Open
Abstract
Activation of neurons and glial cells in the dorsal root ganglion is one of the key mechanisms for the development of hyperalgesia. The aim of the present study was to examine the role of neuroglial activity in the development of opioid-induced hyperalgesia. Male rats were treated with morphine daily for 3 days. The resultant phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 in the dorsal root ganglion was analyzed by immunohistochemistry and Western blotting. Pain hypersensitivity was analyzed using behavioral studies. The amount of cytokine expression in the dorsal root ganglion was also analyzed. Repeated morphine treatment induced hyperalgesia and marked induction of phosphorylated ERK1/2 in the neurons and satellite glial cells on day 3. An opioid receptor antagonist, toll like receptor-4 inhibitor, MAP/ERK kinase (MEK) inhibitor and gap junction inhibitor inhibited morphine-induced hyperalgesia and ERK1/2 phosphorylation. Morphine treatment induced alteration of cytokine expression, which was inhibited by the opioid receptor antagonist, toll like receptor-4 inhibitor, MEK inhibitor and gap junction inhibitor. Dexamethasone inhibited morphine-induced hyperalgesia and ERK1/2 phosphorylation after morphine treatment. The peripherally restricted opioid receptor antagonist, methylnaltrexone, inhibited hyperalgesia and ERK1/2 phosphorylation. Morphine activates ERK1/2 in neurons and satellite glial cells in the dorsal root ganglion via the opioid receptor and toll like receptor-4. ERK1/2 phosphorylation is gap junction-dependent and is associated with the alteration of cytokine expression. Inhibition of neuroinflammation by activation of neurons and glia might be a promising target to prevent opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Shunsuke Yamakita
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Fujita
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daiki Ishikawa
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohsuke Kushimoto
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiko Horii
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumimasa Amaya
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pain Management and Palliative Care Medicine, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
26
|
Antonopoulos SR, Durham PL. Grape seed extract suppresses calcitonin gene-related peptide secretion and upregulates expression of GAD 65/67 and GABAB receptor in primary trigeminal ganglion cultures. IBRO Neurosci Rep 2022; 13:187-197. [PMID: 36093283 PMCID: PMC9449751 DOI: 10.1016/j.ibneur.2022.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/20/2022] [Indexed: 12/02/2022] Open
Abstract
The trigeminal ganglion is implicated in the underlying pathology of migraine and temporomandibular joint disorders (TMD), which are orofacial pain conditions involving peripheral and central sensitization. The neuropeptide calcitonin gene-related peptide (CGRP) is synthesized in some trigeminal ganglion neurons, and its release promotes inflammation, peripheral and central sensitization, and pain signaling. Recent studies in preclinical migraine and TMD models provide evidence that dietary supplementation with grape seed extract (GSE) inhibits trigeminal pain signaling. The goal of this study was to investigate the cellular mechanisms by which GSE modulates primary trigeminal ganglion cultures. The effect of GSE on CGRP secretion was determined by radioimmunoassay. To determine if GSE effects involved modulation of CGRP or the GABAergic system, expression of CGRP, GAD 65 and 67, GABAA receptor, and GABAB1 and GABAB2 receptor subunits were investigated by immunocytochemistry. GSE significantly inhibited basal CGRP secretion but did not alter neuronal CGRP expression. GAD 65 and 67 expression levels in neurons were significantly increased in response to GSE. While GSE did not cause a change in the neuronal expression of GABAA, GSE significantly increased GABAB1 expression in neurons, satellite glial cells, and Schwann cells. GABAB2 expression was significantly elevated in satellite glia and Schwann cells. These findings support the notion that GSE inhibition of basal CGRP secretion involves increased neuronal GAD 65 and 67 and GABAB receptor expression. GSE repression of CGRP release coupled with increased GABAB1 and GABAB2 glial cell expression would be neuroprotective by suppressing neuronal and glial excitability in the trigeminal ganglion. Grape seed extract inhibited basal CGRP release from cultured trigeminal neurons Neuronal expression of GAD 65/67 and GABAB1 was stimulated by grape seed extract Grape seed extract also increased GABAB1 in satellite glial cells and Schwann cells Glial expression of G protein-coupled GABAB2 was enhanced by grape seed extract Grape seed extract promotes neuroprotective cellular changes in trigeminal ganglion
Collapse
|
27
|
Staal RGW, Gandhi A, Zhou H, Cajina M, Jacobsen AM, Hestehave S, Hopper A, Poda S, Chandresana G, Zorn SH, Campbell B, Segerdahl M, Mӧller T, Munro G. Inhibition of P2X7 receptors by Lu AF27139 diminishes colonic hypersensitivity and CNS prostanoid levels in a rat model of visceral pain. Purinergic Signal 2022; 18:499-514. [PMID: 36001278 PMCID: PMC9832206 DOI: 10.1007/s11302-022-09892-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/27/2022] [Indexed: 01/14/2023] Open
Abstract
Visceral pain is a prominent feature of various gastrointestinal diseases. The P2X7 receptor is expressed by multiple cell types including dorsal root ganglion satellite glial cells, macrophages, and spinal microglia, all of which have been implicated in nociceptive sensitization. We have used the selective and CNS penetrant P2X7 receptor antagonist Lu AF27139 to explore this receptor's role in distinct rat models of inflammatory and visceral hypersensitivity. Rats injected with CFA in the hindpaw displayed a marked reduction in hindpaw mechanical threshold, which was dose-dependently reversed by Lu AF27139 (3-30 mg/kg, p.o.). In rats injected with TNBS in the proximal colon, the colorectal distension threshold measured distally was significantly lower than sham treated rats at 7 days post-injection (P < 0.001), indicative of a marked central sensitization. Colonic hypersensitivity was also reversed by Lu AF27139 (10-100 mg/kg) and by the κ-opioid receptor agonist U-50,488H (3 mg/kg, s.c.). Moreover, both Lu AF27139 and U-50,488H prevented a TNBS-induced increase in spinal and brain levels of PGE2 and LTB4, as well as an increase in brain levels of PGF2α and TXB2. Lu AF27139 was well tolerated as revealed by a lack of significant effect on rotarod motor function and coordination at all doses tested up to 300 mg/kg. Thus, P2X7 receptor antagonism is efficacious in a rat model of visceral pain, via a mechanism which potentially involves attenuation of microglial function within spinal and/or supraspinal pain circuits, albeit a peripheral site of action cannot be excluded.
Collapse
Affiliation(s)
- Roland G W Staal
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Adarsh Gandhi
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Hua Zhou
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Manuel Cajina
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | | | - Sara Hestehave
- Neurodegeneration In Vivo Lundbeck Research, Valby, Denmark
| | - Allen Hopper
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Suresh Poda
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Gamini Chandresana
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Stevin H Zorn
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Brian Campbell
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Marta Segerdahl
- Clinical Research Neurology Lundbeck Research, Valby, Denmark
| | - Thomas Mӧller
- Neuroinflammation Disease Biology Unit Lundbeck Research USA, Paramus, NJ, USA
| | - Gordon Munro
- Neurodegeneration In Vivo Lundbeck Research, Valby, Denmark.
- Hoba Therapeutics, Ole Maaløes Vej 3, 2200, Copenhagen N, Denmark.
| |
Collapse
|
28
|
Pathophysiology of Post-Traumatic Trigeminal Neuropathic Pain. Biomolecules 2022; 12:biom12121753. [PMID: 36551181 PMCID: PMC9775491 DOI: 10.3390/biom12121753] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trigeminal nerve injury is one of the causes of chronic orofacial pain. Patients suffering from this condition have a significantly reduced quality of life. The currently available management modalities are associated with limited success. This article reviews some of the common causes and clinical features associated with post-traumatic trigeminal neuropathic pain (PTNP). A cascade of events in the peripheral and central nervous system function is involved in the pathophysiology of pain following nerve injuries. Central and peripheral processes occur in tandem and may often be co-dependent. Due to the complexity of central mechanisms, only peripheral events contributing to the pathophysiology have been reviewed in this article. Future investigations will hopefully help gain insight into trigeminal-specific events in the pathophysiology of the development and maintenance of neuropathic pain secondary to nerve injury and enable the development of new therapeutic modalities.
Collapse
|
29
|
Nie L, Sun K, Gong Z, Li H, Quinn JP, Wang M. Src Family Kinases Facilitate the Crosstalk between CGRP and Cytokines in Sensitizing Trigeminal Ganglion via Transmitting CGRP Receptor/PKA Pathway. Cells 2022; 11:cells11213498. [PMID: 36359895 PMCID: PMC9655983 DOI: 10.3390/cells11213498] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
The communication between calcitonin gene-related peptide (CGRP) and cytokines plays a prominent role in maintaining trigeminal ganglion (TG) and trigeminovascular sensitization. However, the underlying regulatory mechanism is elusive. In this study, we explored the hypothesis that Src family kinases (SFKs) activity facilitates the crosstalk between CGRP and cytokines in sensitizing TG. Mouse TG tissue culture was performed to study CGRP release by enzyme-linked immunosorbent assay, cytokine release by multiplex assay, cytokine gene expression by quantitative polymerase chain reaction, and phosphorylated SFKs level by western blot. The results demonstrated that a SFKs activator, pYEEI (YGRKKRRQRRREPQY(PO3H2)EEIPIYL) alone, did not alter CGRP release or the inflammatory cytokine interleukin-1β (IL-1β) gene expression in the mouse TG. In contrast, a SFKs inhibitor, saracatinib, restored CGRP release, the inflammatory cytokines IL-1β, C-X-C motif ligand 1, C-C motif ligand 2 (CCL2) release, and IL-1β, CCL2 gene expression when the mouse TG was pre-sensitized with hydrogen peroxide and CGRP respectively. Consistently with this, the phosphorylated SFKs level was increased by both hydrogen peroxide and CGRP in the mouse TG, which was reduced by a CGRP receptor inhibitor BIBN4096 and a protein kinase A (PKA) inhibitor PKI (14–22) Amide. The present study demonstrates that SFKs activity plays a pivotal role in facilitating the crosstalk between CGRP and cytokines by transmitting CGRP receptor/PKA signaling to potentiate TG sensitization and ultimately trigeminovascular sensitization.
Collapse
Affiliation(s)
- Lingdi Nie
- Centre for Neuroscience, Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool L69 7ZB, UK
| | - Kai Sun
- Centre for Neuroscience, Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China
| | - Ziyang Gong
- Centre for Neuroscience, Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China
| | - Haoyang Li
- Centre for Neuroscience, Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China
| | - John P. Quinn
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool L69 7ZB, UK
| | - Minyan Wang
- Centre for Neuroscience, Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, Liverpool L69 7ZB, UK
- Correspondence:
| |
Collapse
|
30
|
Reducha PV, Edvinsson L, Haanes KA. Could Experimental Inflammation Provide Better Understanding of Migraines? Cells 2022; 11:cells11152444. [PMID: 35954288 PMCID: PMC9368653 DOI: 10.3390/cells11152444] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Migraines constitute a common neurological and headache disorder affecting around 15% of the world’s population. In addition to other mechanisms, neurogenic neuroinflammation has been proposed to play a part in migraine chronification, which includes peripheral and central sensitization. There is therefore considerable evidence suggesting that inflammation in the intracranial meninges could be a key element in addition to calcitonin gene-related peptide (CGRP), leading to sensitization of trigeminal meningeal nociceptors in migraines. There are several studies that have utilized this approach, with a strong focus on using inflammatory animal models. Data from these studies show that the inflammatory process involves sensitization of trigeminovascular afferent nerve terminals. Further, by applying a wide range of different pharmacological interventions, insight has been gained on the pathways involved. Importantly, we discuss how animal models should be used with care and that it is important to evaluate outcomes in the light of migraine pathology.
Collapse
Affiliation(s)
- Philip Victor Reducha
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet Glostrup, 2600 Glostrup, Denmark
- Department of Biology, Section of Cell Biology and Physiology, University of Copenhagen, 1017 Copenhagen, Denmark
| | - Lars Edvinsson
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet Glostrup, 2600 Glostrup, Denmark
- Division of Experimental Vascular Research, Department of Clinical Sciences, Lund University Hospital, 221 00 Lund, Sweden
| | - Kristian Agmund Haanes
- Department of Clinical Experimental Research, Glostrup Research Institute, Copenhagen University Hospital, Rigshospitalet Glostrup, 2600 Glostrup, Denmark
- Department of Biology, Section of Cell Biology and Physiology, University of Copenhagen, 1017 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
31
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [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: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
32
|
Glutamine Maintains Satellite Glial Cells Growth and Survival in Culture. Neurochem Res 2022; 47:3635-3646. [PMID: 35522367 DOI: 10.1007/s11064-022-03614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/10/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Satellite glial cells (SGCs) tightly surround neurons and modulate sensory transmission in dorsal root ganglion (DRG). At present, the biological property of primary SGCs in culture deserves further investigation. To reveal the key factor for SGCs growth and survival, we examined the effects of different culture supplementations containing Dulbecco's Modified Eagle Medium (DMEM)/F12, DMEM high glucose (HG) or Neurobasal-A (NB). CCK-8 proliferation assay showed an increased proliferation of SGCs in DMEM/F12 and DMEM/HG, but not in NB medium. Bax, AnnexinV, and propidium iodide (PI) staining results showed that NB medium caused cell death and apoptosis. We showed that glutamine was over 2.5 mM in DMEM/F12 and DMEM/HG, whereas it was absence in NB medium. Interestingly, exogenous glutamine application significantly reversed the poor proliferation and cell death of SGCs in NB medium. These findings demonstrated that DMEM/F12 medium was optimal to get high-purity SGCs. Glutamine was the key molecule to maintain SGCs growth and survival in culture. Here, we provided a novel approach to get high-purity SGCs by changing the key component of culture medium. Our study shed a new light on understanding the biological property and modulation of glial cells of primary sensory ganglia.
Collapse
|
33
|
Hoshino Y, Okuno T, Saigusa D, Kano K, Yamamoto S, Shindou H, Aoki J, Uchida K, Yokomizo T, Ito N. Lysophosphatidic acid receptor 1/3 antagonist inhibits the activation of satellite glial cells and reduces acute nociceptive responses. FASEB J 2022; 36:e22236. [PMID: 35218596 DOI: 10.1096/fj.202101678r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) exerts various biological activities through six characterized G protein-coupled receptors (LPA1-6 ). While LPA-LPA1 signaling contributes toward the demyelination and retraction of C-fiber and induces neuropathic pain, the effects of LPA-LPA1 signaling on acute nociceptive pain is uncertain. This study investigated the role of LPA-LPA1 signaling in acute nociceptive pain using the formalin test. The pharmacological inhibition of the LPA-LPA1 axis significantly attenuated formalin-induced nociceptive behavior. The LPA1 mRNA was expressed in satellite glial cells (SGCs) in dorsal root ganglion (DRG) and was particularly abundant in SGCs surrounding large DRG neurons, which express neurofilament 200. Treatment with LPA1/3 receptor (LPA1/3 ) antagonist inhibited the upregulation of glial markers and inflammatory cytokines in DRG following formalin injection. The LPA1/3 antagonist also attenuated phosphorylation of extracellular signal-regulated kinase, especially in SGCs and cyclic AMP response element-binding protein in the dorsal horn following formalin injection. LPA amounts after formalin injection to the footpad were quantified by liquid chromatography/tandem mass spectrometry, and LPA levels were found to be increased in the innervated DRGs. Our results indicate that LPA produced in the innervated DRGs promotes the activation of SGCs through LPA1 , increases the sensitivity of primary neurons, and modulates pain behavior. These results facilitate our understanding of the pathology of acute nociceptive pain and demonstrate the possibility of the LPA1 on SGCs as a novel target for acute pain control.
Collapse
Affiliation(s)
- Yoko Hoshino
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daisuke Saigusa
- Laboratory of Biomedical and Analytical Sciences, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan.,Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shota Yamamoto
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Tokyo, Japan.,Department of Lipid Medical Science, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
34
|
Carozzi VA, Salio C, Rodriguez-Menendez V, Ciglieri E, Ferrini F. 2D <em>vs</em> 3D morphological analysis of dorsal root ganglia in health and painful neuropathy. Eur J Histochem 2021; 65. [PMID: 34664808 PMCID: PMC8547168 DOI: 10.4081/ejh.2021.3276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
Dorsal root ganglia (DRGs) are clusters of sensory neurons that transmit the sensory information from the periphery to the central nervous system, and satellite glial cells (SGCs), their supporting trophic cells. Sensory neurons are pseudounipolar neurons with a heterogeneous neurochemistry reflecting their functional features. DRGs, not protected by the blood brain barrier, are vulnerable to stress and damage of different origin (i.e., toxic, mechanical, metabolic, genetic) that can involve sensory neurons, SGCs or, considering their intimate intercommunication, both cell populations. DRG damage, primary or secondary to nerve damage, produces a sensory peripheral neuropathy, characterized by neurophysiological abnormalities, numbness, paraesthesia and dysesthesia, tingling and burning sensations and neuropathic pain. DRG stress can be morphologically detected by light and electron microscope analysis with alterations in cell size (swelling/atrophy) and in different subcellular compartments (i.e., mitochondria, endoplasmic reticulum, and nucleus) of neurons and/or SGCs. In addition, neurochemical changes can be used to portray abnormalities of neurons and SGC. Conventional immunostaining, i.e., immunohistochemical detection of specific molecules in tissue slices, can be employed to detect, localize and quantify particular markers of damage in neurons (i.e., nuclear expression of ATF3) or SGCs (i.e., increased expression of GFAP), markers of apoptosis (i.e., caspases), markers of mitochondrial suffering and oxidative stress (i.e., 8-OHdG), markers of tissue inflammation (i.e., CD68 for macrophage infiltration) etc. However classical (2D) methods of immunostaining disrupt the overall organization of the DRG, thus resulting in the loss of some crucial information. Whole-mount (3D) methods have been recently developed to investigate DRG morphology and neurochemistry without tissue slicing, giving the opportunity to study the intimate relationship between SGCs and sensory neurons in health and disease. Here, we aim to compare classical (2D) vs whole-mount (3D) approaches to highlight “pros” and “cons” of the two methodologies when analysing neuropathy-induced alterations in DRGs.
Collapse
Affiliation(s)
- Valentina Alda Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza (MB).
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| | | | | | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Grugliasco (TO).
| |
Collapse
|
35
|
Westgate CSJ, Israelsen IME, Jensen RH, Eftekhari S. Understanding the link between obesity and headache- with focus on migraine and idiopathic intracranial hypertension. J Headache Pain 2021; 22:123. [PMID: 34629054 PMCID: PMC8504002 DOI: 10.1186/s10194-021-01337-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/26/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Obesity confers adverse effects to every system in the body including the central nervous system. Obesity is associated with both migraine and idiopathic intracranial hypertension (IIH). The mechanisms underlying the association between obesity and these headache diseases remain unclear. METHODS We conducted a narrative review of the evidence in both humans and rodents, for the putative mechanisms underlying the link between obesity, migraine and IIH. RESULTS Truncal adiposity, a key feature of obesity, is associated with increased migraine morbidity and disability through increased headache severity, frequency and more severe cutaneous allodynia. Obesity may also increase intracranial pressure and could contribute to headache morbidity in migraine and be causative in IIH headache. Weight loss can improve both migraine and IIH headache. Preclinical research highlights that obesity increases the sensitivity of the trigeminovascular system to noxious stimuli including inflammatory stimuli, but the underlying molecular mechanisms remain unelucidated. CONCLUSIONS This review highlights that at the epidemiological and clinical level, obesity increases morbidity in migraine and IIH headache, where weight loss can improve headache morbidity. However, further research is required to understand the molecular underpinnings of obesity related headache in order to generate novel treatments.
Collapse
Affiliation(s)
- Connar Stanley James Westgate
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Glostrup Research Institute, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Ida Marchen Egerod Israelsen
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Glostrup Research Institute, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Rigmor Højland Jensen
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Glostrup Research Institute, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Sajedeh Eftekhari
- Danish Headache Center, Department of Neurology, Rigshospitalet- Glostrup, Glostrup Research Institute, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark.
| |
Collapse
|
36
|
Discrepancy in the Usage of GFAP as a Marker of Satellite Glial Cell Reactivity. Biomedicines 2021; 9:biomedicines9081022. [PMID: 34440226 PMCID: PMC8391720 DOI: 10.3390/biomedicines9081022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Satellite glial cells (SGCs) surrounding the neuronal somas in peripheral sensory ganglia are sensitive to neuronal stressors, which induce their reactive state. It is believed that such induced gliosis affects the signaling properties of the primary sensory neurons and is an important component of the neuropathic phenotype leading to pain and other sensory disturbances. Efforts to understand and manipulate such gliosis relies on reliable markers to confirm induced SGC reactivity and ultimately the efficacy of targeted intervention. Glial fibrillary acidic protein (GFAP) is currently the only widely used marker for such analyses. However, we have previously described the lack of SGC upregulation of GFAP in a mouse model of sciatic nerve injury, suggesting that GFAP may not be a universally suitable marker of SGC gliosis across species and experimental models. To further explore this, we here investigate the regulation of GFAP in two different experimental models in both rats and mice. We found that whereas GFAP was upregulated in both rodent species in the applied inflammation model, only the rat demonstrated increased GFAP in SGCs following sciatic nerve injury; we did not observe any such GFAP upregulation in the mouse model at either protein or mRNA levels. Our results demonstrate an important discrepancy between species and experimental models that prevents the usage of GFAP as a universal marker for SGC reactivity.
Collapse
|
37
|
Bolay H, Karadas Ö, Oztürk B, Sonkaya R, Tasdelen B, Bulut TDS, Gülbahar Ö, Özge A, Baykan B. HMGB1, NLRP3, IL-6 and ACE2 levels are elevated in COVID-19 with headache: a window to the infection-related headache mechanism. J Headache Pain 2021; 22:94. [PMID: 34384355 PMCID: PMC8358545 DOI: 10.1186/s10194-021-01306-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/30/2021] [Indexed: 12/15/2022] Open
Abstract
Background and aim Pathogenesis of COVID-19 -related headache is unknown, though the induction of the trigeminal neurons through inflammation is proposed. We aimed to investigate key systemic circulating inflammatory molecules and their clinical relations in COVID-19 patients with headache. Methods This cross-sectional study enrolled 88 COVID-19 patients, hospitalized on a regular ward during the second wave of the pandemic. Clinical characteristics of COVID-19 patients were recorded, and laboratory tests were studied. Results The mean ages of 48 COVID-19 patients with headache (47.71 ± 10.8) and 40 COVID-19 patients without headache (45.70 ± 12.72) were comparable. COVID-19 patients suffered from headache had significantly higher serum levels of HMGB1, NLRP3, ACE2, and IL-6 than COVID-19 patients without headache, whereas CGRP and IL-10 levels were similar in the groups. Angiotensin II level was significantly decreased in the headache group. COVID-19 patients with headache showed an increased frequency of pulmonary involvement and increased D- dimer levels. Furthermore, COVID-19 was more frequently associated with weight loss, nausea, and diarrhea in patients with headache. Serum NLRP3 levels were correlated with headache duration and hospital stay, while headache response to paracetamol was negatively correlated with HMGB1 and positively associated with IL-10 levels. Conclusion Stronger inflammatory response is associated with headache in hospitalized COVID-19 patients with moderate disease severity. Increased levels of the circulating inflammatory and/or nociceptive molecules like HMGB1, NLRP3, and IL-6 may play a role in the potential induction of the trigeminal system and manifestation of headache secondary to SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hayrunnisa Bolay
- Department of Neurology and Algology, Neuropsychiatry Center, Neuroscience and Neurotechnology Center (NÖROM), Gazi University Hospital, Medical Faculty, Besevler, 06510, Ankara, Turkey.
| | - Ömer Karadas
- Neurology Department, University of Health Science, Gülhane School of Medicine, Ankara, Turkey
| | - Bilgin Oztürk
- Neurology Department, University of Health Science, Gülhane School of Medicine, Ankara, Turkey
| | - Riza Sonkaya
- Neurology Department, University of Health Science, Gülhane School of Medicine, Ankara, Turkey
| | - Bahar Tasdelen
- Department of Biostatistics and Medical Informatics, Mersin University, Medical Faculty, Mersin, Turkey
| | - Tuba D S Bulut
- Department of Medical Biochemistry, Gazi University, Medical Faculty, Ankara, Turkey
| | - Özlem Gülbahar
- Department of Medical Biochemistry, Gazi University, Medical Faculty, Ankara, Turkey
| | - Aynur Özge
- Department of Neurology and Algology, Mersin University, Medical Faculty, Mersin, Turkey
| | - Betül Baykan
- Istanbul Faculty of Medicine, Department of Neurology, Headache Center, Istanbul University, Istanbul, Turkey
| |
Collapse
|
38
|
Ahmed F, Rahman M, Thompson R, Bereiter DA. Role of Connexin 43 in an Inflammatory Model for TMJ Hyperalgesia. FRONTIERS IN PAIN RESEARCH 2021; 2:715871. [PMID: 35295418 PMCID: PMC8915650 DOI: 10.3389/fpain.2021.715871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022] Open
Abstract
Temporomandibular joint disorders (TMD) consist of a heterogeneous group of conditions that present with pain in the temporomandibular joint (TMJ) region and muscles of mastication. This project assessed the role of connexin 43 (Cx43), a gap junction protein, in the trigeminal ganglion (TG) in an animal model for persistent inflammatory TMJ hyperalgesia. Experiments were performed in male and female rats to determine if sex differences influence the expression and/or function of Cx43 in persistent TMJ hyperalgesia. Intra-TMJ injection of Complete Freund's Adjuvant (CFA) caused a significant increase in Cx43 expression in the TG at 4 days and 10 days post-injection in ovariectomized (OvX) female rats and OvX females treated with estradiol (OvXE), while TG samples in males revealed only marginal increases. Intra-TG injection of interference RNA for Cx43 (siRNA Cx43) 3 days prior to recording, markedly reduced TMJ-evoked masseter muscle electromyographic (MMemg) activity in all CFA-inflamed rats, while activity in sham animals was not affected. Western blot analysis revealed that at 3 days after intra-TG injection of siRNA Cx43 protein levels for Cx43 were significantly reduced in TG samples of all CFA-inflamed rats. Intra-TG injection of the mimetic peptide GAP19, which inhibits Cx43 hemichannel formation, greatly reduced TMJ-evoked MMemg activity in all CFA-inflamed groups, while activity in sham groups was not affected. These results revealed that TMJ inflammation caused a persistent increase in Cx43 protein in the TG in a sex-dependent manner. However, intra-TG blockade of Cx43 by siRNA or by GAP19 significantly reduced TMJ-evoked MMemg activity in both males and females following TMJ inflammation. These results indicated that Cx43 was necessary for enhanced jaw muscle activity after TMJ inflammation in males and females, a result that could not be predicted on the basis of TG expression of Cx43 alone.
Collapse
|
39
|
Zhang Y, Standifer KM. Exacerbated Headache-Related Pain in the Single Prolonged Stress Preclinical Model of Post-traumatic Stress Disorder. Cell Mol Neurobiol 2021; 41:1009-1018. [PMID: 32930941 PMCID: PMC8159770 DOI: 10.1007/s10571-020-00962-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 09/03/2020] [Indexed: 11/29/2022]
Abstract
Chronic headache pain is one of the most commonly reported comorbid pain conditions with post-traumatic stress disorder (PTSD) patients and resistant to effective treatment, yet no combined preclinical model of the two disorders has been reported. Here, we used a modified chronic headache pain model to investigate the contribution of single prolonged stress (SPS) model of PTSD with sodium nitroprusside (SNP)-induced hyperalgesia. Injection of SNP (2 mg/kg, i.p.) occurred every other day from day 7 to day 15 after initiation of SPS in rats. Paw withdrawal threshold (PWT) to von Frey stimuli and tail flick latencies (TFL) dramatically decreased as early as 7 days after SPS and lasted until at least day 21. Basal PWT and TFL also significantly decreased during the SNP treatment period. The lower nociceptive thresholds recovered in 6 days following the final SNP injection in SNP group, but not in SPS + SNP group. Elevated nociceptin/OFQ (N/OFQ) levels observed in cerebrospinal fluid of SPS rats were even higher in SPS + SNP group. Glial fibrillary acidic protein (GFAP) and N/OFQ peptide (NOP) receptor mRNA expression increased in dorsal root ganglia (DRG) 21 days after SPS exposure; mRNA increases in the SPS/SNP group was more pronounced than SPS or SNP alone. GFAP protein expression was upregulated in trigeminal ganglia by SPS. Our results indicate that traumatic stress exaggerated chronic SNP-induced nociceptive hypersensitivity, and that N/OFQ and activated satellite glia cells may play an important role in the interaction between both conditions.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
40
|
Uhelski ML, Li Y, Fonseca MM, Romero-Snadoval EA, Dougherty PM. Role of innate immunity in chemotherapy-induced peripheral neuropathy. Neurosci Lett 2021; 755:135941. [PMID: 33961945 DOI: 10.1016/j.neulet.2021.135941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 01/18/2023]
Abstract
It has become increasingly clear that the innate immune system plays an essential role in the generation of many types of neuropathic pain including that which accompanies cancer treatment. In this article we review current findings of the role of the innate immune system in contributing to cancer treatment pain at the distal endings of peripheral nerve, in the nerve trunk, in the dorsal root ganglion and in the spinal dorsal horn.
Collapse
Affiliation(s)
- Megan L Uhelski
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Yan Li
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States
| | - Miriam M Fonseca
- The Department of Anesthesiology, Wake Forest School of Medicine, United States
| | | | - Patrick M Dougherty
- The Department of Pain Medicine Research, The Division of Anesthesiology, Critical Care and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, United States.
| |
Collapse
|
41
|
Leo M, Schmitt LI, Kutritz A, Kleinschnitz C, Hagenacker T. Cisplatin-induced activation and functional modulation of satellite glial cells lead to cytokine-mediated modulation of sensory neuron excitability. Exp Neurol 2021; 341:113695. [PMID: 33727094 DOI: 10.1016/j.expneurol.2021.113695] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/01/2021] [Accepted: 03/11/2021] [Indexed: 12/23/2022]
Abstract
Cisplatin plays an essential role in the treatment of various cancers. Cisplatin exhibits high efficacy, but it often leads to severe neurotoxic side effects, such as chemotherapy-induced polyneuropathy (CIPN). The pathophysiology of CIPN is not fully understood. There is increasing evidence for damage to satellite glial cells (SGC) and dorsal root ganglion (DRG) neurons. We investigated the influence of cisplatin on the function of SGCs and the direct influence on DRGs. Satellite glial cells were isolated from DRG and exposed to 0.1, 1, 10, or 100 μM cisplatin for 2 h, 4 h, and 24 h. Using immunocytochemical staining and Western blot analysis, the expression of the glial fibrillary acid protein (GFAP), reactive oxygen species (ROS), and inward rectifier potassium channel 4.1 (Kir4.1) was determined. An increase in the immune reactivity (IR) and protein levels of GFAP and ROS was measured, and a reduction of IR and protein level of Kir4.1 was detected. A decrease in these channels' current density was observed using the whole-cell patch-clamp recording. The interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) release of SGCs increased after cisplatin exposure as measured using ELISA, and interleukin-1β (IL-1β) decreased. The SGC-secreted factors in the supernatant after cisplatin treatment led to a modulation of cultured DRG neurons' excitability. Taken together, the modulation and function of different SGC proteins could be linked to a direct impact of cisplatin. Further, SGC-secreted factors influenced the excitability of sensory neurons. Overall, SGCs could be a potential target in preventing and treating chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Markus Leo
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany.
| | - Linda-Isabell Schmitt
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Andrea Kutritz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| | - Tim Hagenacker
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
42
|
Wang J, Lou Z, Xi H, Li Z, Li L, Li Z, Zhang K, Asakawa T. Verification of neuroprotective effects of alpha-lipoic acid on chronic neuropathic pain in a chronic constriction injury rat model. Open Life Sci 2021; 16:222-228. [PMID: 33817313 PMCID: PMC7968532 DOI: 10.1515/biol-2021-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/09/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022] Open
Abstract
Treatment of neuropathic pain is far from satisfactory. This study sought evidence of a neuroprotective effect of alpha-lipoic acid (ALA) to treat neuropathic pain in a chronic constriction injury (CCI) rat model. A total of 48 rats were randomly divided into sham, CCI, or CCI + ALA groups. Mechanical and thermal nociceptive thresholds were evaluated as behavioral assessments. Dorsal root ganglia cells were assessed morphologically with hematoxylin and eosin staining and for apoptosis with P53 immunohistochemical staining. Compared with the sham group, the CCI group had a shorter paw withdrawal threshold and paw withdrawal latency, abnormal morphologic manifestations, and increased numbers of satellite glial cells and P53+ cells. These changes were significantly reversed by treatment with ALA. Our study indicates neuroprotective effects of ALA on chronic neuropathic pain in a CCI rat model. ALA is potentially considered to be developed as a treatment for neuropathic pain caused by peripheral nerve injury, which requires further verification.
Collapse
Affiliation(s)
- Junhao Wang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhaohui Lou
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Haiyang Xi
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhi Li
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lepeng Li
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhenzhen Li
- Institute of Clinical Medicine, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kai Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Handayama, 1-20-1, Higashi-ku, Hamamatsu City, Shizuoka 431-3192, Japan
- Research Base of Traditional Chinese Medicine Syndrome, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| |
Collapse
|
43
|
Mailhot B, Christin M, Tessandier N, Sotoudeh C, Bretheau F, Turmel R, Pellerin È, Wang F, Bories C, Joly-Beauparlant C, De Koninck Y, Droit A, Cicchetti F, Scherrer G, Boilard E, Sharif-Naeini R, Lacroix S. Neuronal interleukin-1 receptors mediate pain in chronic inflammatory diseases. J Exp Med 2021; 217:151879. [PMID: 32573694 PMCID: PMC7478735 DOI: 10.1084/jem.20191430] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 03/03/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic pain is a major comorbidity of chronic inflammatory diseases. Here, we report that the cytokine IL-1β, which is abundantly produced during multiple sclerosis (MS), arthritis (RA), and osteoarthritis (OA) both in humans and in animal models, drives pain associated with these diseases. We found that the type 1 IL-1 receptor (IL-1R1) is highly expressed in the mouse and human by a subpopulation of TRPV1+ dorsal root ganglion neurons specialized in detecting painful stimuli, termed nociceptors. Strikingly, deletion of the Il1r1 gene specifically in TRPV1+ nociceptors prevented the development of mechanical allodynia without affecting clinical signs and disease progression in mice with experimental autoimmune encephalomyelitis and K/BxN serum transfer–induced RA. Conditional restoration of IL-1R1 expression in nociceptors of IL-1R1–knockout mice induced pain behavior but did not affect joint damage in monosodium iodoacetate–induced OA. Collectively, these data reveal that neuronal IL-1R1 signaling mediates pain, uncovering the potential benefit of anti–IL-1 therapies for pain management in patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Benoit Mailhot
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Marine Christin
- Department of Physiology and Cell Information Systems Group, McGill University, Montreal, Canada
| | - Nicolas Tessandier
- Axe Maladies infectieuses et immunitaires du Centre de recherche du CHU de Québec-Université Laval et Département de microbiologie-infectiologie et d'immunologie de l'Université Laval, Québec, Canada
| | - Chaudy Sotoudeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Palo Alto, CA
| | - Floriane Bretheau
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Roxanne Turmel
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Ève Pellerin
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Feng Wang
- Centre de recherche CERVO, Québec, Canada
| | | | - Charles Joly-Beauparlant
- Axe Endocrinologie-néphrologie du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | | | - Arnaud Droit
- Axe Endocrinologie-néphrologie du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| | - Francesca Cicchetti
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de psychiatrie et de neurosciences de l'Université Laval, Québec, Canada
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, University of North Carolina Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC.,New York Stem Cell Foundation - Robertson Investigator, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Eric Boilard
- Axe Maladies infectieuses et immunitaires du Centre de recherche du CHU de Québec-Université Laval et Département de microbiologie-infectiologie et d'immunologie de l'Université Laval, Québec, Canada
| | - Reza Sharif-Naeini
- Department of Physiology and Cell Information Systems Group, McGill University, Montreal, Canada
| | - Steve Lacroix
- Axe Neurosciences du Centre de recherche du CHU de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, Canada
| |
Collapse
|
44
|
Borghi SM, Fattori V, Carvalho TT, Tatakihara VLH, Zaninelli TH, Pinho-Ribeiro FA, Ferraz CR, Staurengo-Ferrari L, Casagrande R, Pavanelli WR, Cunha FQ, Cunha TM, Pinge-Filho P, Verri WA. Experimental Trypanosoma cruzi Infection Induces Pain in Mice Dependent on Early Spinal Cord Glial Cells and NFκB Activation and Cytokine Production. Front Immunol 2021; 11:539086. [PMID: 33574810 PMCID: PMC7870690 DOI: 10.3389/fimmu.2020.539086] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
The neglected tropical infirmity Chagas disease (CD) presents high mortality. Its etiological agent T. cruzi is transmitted by infected hematophagous insects. Symptoms of the acute phase of the infection include fever, fatigue, body aches, and headache, making diagnosis difficult as they are present in other illnesses as well. Thus, in endemic areas, individuals with undetermined pain may be considered for CD. Although pain is a characteristic symptom of CD, its cellular and molecular mechanisms are unknown except for demonstration of a role for peripheral TNF-α in CD pain. In this study, we evaluate the role of spinal cord glial cells in experimental T. cruzi infection in the context of pain using C57BL/6 mice. Pain, parasitemia, survival, and glial and neuronal function as well as NFκB activation and cytokine/chemokine production were assessed. T. cruzi infection induced chronic mechanical and thermal hyperalgesia. Systemic TNF-α and IL-1β peaked 14 days postinfection (p.i.). Infected mice presented increased spinal gliosis and NFκB activation compared to uninfected mice at 7 days p.i. Glial and NFκB inhibitors limited T. cruzi–induced pain. Nuclear phosphorylated NFκB was detected surrounded by glia markers, and glial inhibitors reduced its detection. T. cruzi–induced spinal cord production of cytokines/chemokines was also diminished by glial inhibitors. Dorsal root ganglia (DRG) neurons presented increased activity in infected mice, and the production of inflammatory mediators was counteracted by glial/NFκB inhibitors. The present study unveils the contribution of DRG and spinal cord cellular and molecular events leading to pain in T. cruzi infection, contributing to a better understanding of CD pathology.
Collapse
Affiliation(s)
- Sergio M Borghi
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil.,Center for Research in Health Science, University of Northern Paraná-Unopar, Londrina, Brazil
| | - Victor Fattori
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Thacyana T Carvalho
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Vera L H Tatakihara
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Tiago H Zaninelli
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Felipe A Pinho-Ribeiro
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Camila R Ferraz
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Larissa Staurengo-Ferrari
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Rubia Casagrande
- Departament of Pharmaceutical Sciences, Health Sciences Center, University Hospital, Londrina State University, Londrina, Brazil
| | - Wander R Pavanelli
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Phileno Pinge-Filho
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| | - Waldiceu A Verri
- Department of Pathology, Center of Biological Science, State University of Londrina, Londrina, Brazil
| |
Collapse
|
45
|
Tripathi GM, Tripathi S. Immunogenetics in Migraine. THE MOLECULAR IMMUNOLOGY OF NEUROLOGICAL DISEASES 2021:135-147. [DOI: 10.1016/b978-0-12-821974-4.00006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
46
|
Kanno K, Shimizu K, Shinoda M, Hayashi M, Takeichi O, Iwata K. Role of macrophage-mediated Toll-like receptor 4-interleukin-1R signaling in ectopic tongue pain associated with tooth pulp inflammation. J Neuroinflammation 2020; 17:312. [PMID: 33081813 PMCID: PMC7576725 DOI: 10.1186/s12974-020-01995-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 10/08/2020] [Indexed: 01/16/2023] Open
Abstract
Background The existence of referred pain and ectopic paresthesia caused by tooth pulp inflammation may make definitive diagnosis difficult and cause misdiagnosis or mistreatment; thus, elucidation of that molecular mechanism is urgent. In the present study, we investigated the mechanisms underlying ectopic pain, especially tongue hyperalgesia, after tooth pulp inflammation. Methods A rat model with mandibular first molar tooth pulp exposure was employed. Tooth pulp exposure-induced heat and mechanical-evoked tongue hypersensitivity was measured, and immunohistochemical staining for Iba1, a marker of active macrophages, IL-1β, IL-1 type I receptor (IL-1RΙ), and toll-like receptor 4 in the trigeminal ganglion was performed. In addition, we investigated the effects of injections of liposomal clodronate Clophosome-A (LCCA), a selective macrophage depletion agent, lipopolysaccharide from Rhodobacter sphaeroides (LPS-RS, a toll-like receptor 4 antagonist), IL-1β, or heat shock protein 70 (Hsp70, a selective agonist of toll-like receptor 4), to examine changes in tongue hypersensitivity and in the regulation of IL-1RΙ, toll-like receptor 4, and transient receptor potential vanilloid 1 (TRPV1) biosynthesis. Results At day 1 after tooth pulp exposure, obvious tooth pulp inflammation was observed. Tooth pulp exposure-induced heat and mechanical tongue hypersensitivity was observed from days 1 to 3 after tooth pulp exposure. The production of IL-1β in activated macrophages and toll-like receptor 4 and IL-1RΙ expression were significantly increased in trigeminal ganglion neurons innervating the tongue following tooth pulp exposure. Intra-trigeminal ganglion injection of LCCA significantly suppressed tongue hypersensitivity; however, toll-like receptor 4 and IL-1RΙ expression in trigeminal ganglion neurons innervating the tongue was not significantly altered. Intra-trigeminal ganglion injection of LPS-RS significantly suppressed tongue hypersensitivity and reduced IL-1RΙ expression in the trigeminal ganglion neurons innervating the tongue following tooth pulp exposure. Intra-trigeminal ganglion injection of recombinant Hsp70 significantly promoted tongue hypersensitivity and increased IL-1RI expression in trigeminal ganglion neurons innervating the tongue in naive rats. Furthermore, intra-trigeminal ganglion injection of recombinant IL-1β led to tongue hypersensitivity and enhanced TRPV1 expression in trigeminal ganglion neurons innervating the tongue in naive rats. Conclusions The present findings suggest that the neuron-macrophage interaction mediated by toll-like receptor 4 and IL-1RI activation in trigeminal ganglion neurons affects the pathogenesis of abnormal tongue pain following tooth pulp inflammation via IL-1RI and TRPV1 signaling in the trigeminal ganglion. Further research may contribute to the establishment of new therapeutic and diagnostic methods.
Collapse
Affiliation(s)
- Kohei Kanno
- Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kohei Shimizu
- Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan. .,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan.
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Makoto Hayashi
- Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Osamu Takeichi
- Department of Endodontics, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.,Division of Advanced Dental Treatment, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.,Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
47
|
Neferine alleviates P2X3 receptor in rat dorsal root ganglia mediated neuropathic pain. Neurosci Res 2020; 170:265-272. [PMID: 32882253 DOI: 10.1016/j.neures.2020.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 11/21/2022]
Abstract
Chronic neuropathic pain is caused by tissue damage or nervous system inflammation and is characterized by sensitivity to painful stimuli. P2X3 receptors play an important role in facilitating pain transmission. Neferine is a bisbenzylisoquinline alkaloid isolated from seed embryos of lotus, which has anti-inflammatory and anti-oxidation pharmacological functions. The present research investigated whether neferine relieves neuropathic pain related to the P2X3 receptor in rat dorsal root ganglia (DRGs). Chronic contraction injury (CCI) in rats was used as a model for neuropathic pain. The results indicated that the expression of P2X3 receptor was significantly increased in the DRGs of CCI rats and that mechanical allodynia and thermal hyperalgesia were also enhanced in CCI rats. Neferine markedly lowered the upregulated P2X3 receptor and interleukin-1beta, inhibited the phosphorylation and activation of ERK1/2 in the DRGs of CCI rats, and relieved neuropathic pain. Therefore, neferine alleviates neuropathic pain by downregulating the expression of P2X3 receptor.
Collapse
|
48
|
NLRP2 inflammasome in dorsal root ganglion as a novel molecular platform that produces inflammatory pain hypersensitivity. Pain 2020; 160:2149-2160. [PMID: 31162334 DOI: 10.1097/j.pain.0000000000001611] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Inflammatory pain hypersensitivity is associated with activation of primary afferent neurons. This study investigated the existence of the inflammasome in dorsal root ganglion (DRG) and the functional significance in the development of inflammatory pain hypersensitivity. Tissue inflammation was induced in male C57BL/6 mice with complete Freund's adjuvant (CFA) or ceramide injection into the hind paw. Behavioral testing was performed to investigate inflammation-induced pain hypersensitivity. Ipsilateral L5 DRGs were obtained for analysis. Expression of nucleotide oligomerization domain-like receptors (NLRs) was analyzed with real-time PCR. Cleaved interleukin (IL)-1β and NLRP2 expression was investigated with immunohistochemistry and western blotting. Caspase 1 activity was also measured. A caspase 1 inhibitor and NLRP2 siRNA were intrathecally administered to inhibit NLRP2 inflammasome signaling in DRG. Cleaved IL-1β expression was significantly increased after CFA injection in small-sized DRG neurons. The amount of cleaved IL-1β and caspase 1 activity were also increased. Among several NLRs, NLRP2 mRNA was significantly increased in DRG after CFA injection. NLRP2 was expressed in small-sized DRG neurons. Intrathecal injection of a caspase 1 inhibitor or NLRP2 siRNA reduced CFA-induced pain hypersensitivity and cleaved IL-1β expression in DRG. Induction of cleaved IL-1β and NLRP2 in DRG neurons was similarly observed after ceramide injection. NLRP2 siRNA inhibited ceramide-induced pain hypersensitivity. These results confirmed the existence of NLRP2 inflammasome in DRG neurons. Activation of the NLRP2 inflammasome leads to activation of DRG neurons and subsequent development of pain hypersensitivity in various types of tissue inflammation.
Collapse
|
49
|
Yomogida S, Sekiguchi M, Konno SI. Involvement between social defeat stress and pain-related behavior in a rat lumbar disk herniation model. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2020; 29:2431-2440. [PMID: 32700124 DOI: 10.1007/s00586-020-06533-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 06/07/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Psychological and social factors are involved in the disability and chronicity of pain. Our study aim was to investigate whether social defeat stress (SDS) as a psychophysical stress affected mechanical withdrawal thresholds in the lumbar disk herniation (LDH) rat model. Changes in microglia and astrocytes, which play important roles in neuropathic pain states, were also investigated. MATERIALS AND METHODS For the LDH model, nucleus pulposus (NP) was applied to the L5 dorsal root ganglion (DRG) in adult female Sprague-Dawley rats. SDS was performed 15 min daily for 8 days. Mechanical withdrawal thresholds were measured, and immunoreactive cells of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule-1 (Iba-1), which were used as markers of microglia, satellite glial cells, and astrocytes, were assessed in the DRG, spinal cord (SC), and ventrolateral periaqueductal gray matter (VLPAG). RESULTS Mechanical withdrawal thresholds decreased in the NP group for 21 days and for 35 days in the NP + SDS group. Expression of GFAP and Iba-1 in the DRG and SC increased up to day 21 in the NP and NP + SDS groups. In the sham + SDS and NP + SDS groups, expression of GFAP in the VLPAG decreased until day 35. CONCLUSION SDS prolongs mechanical allodynia induced by NP. Changes of GFAP expression in the VLPAG were associated with mechanical allodynia of the NP + SDS group during the late phase. These results suggest that psychological chronic stress might delay recovery from mechanical allodynia induced by the LDH model.
Collapse
Affiliation(s)
- Shota Yomogida
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Miho Sekiguchi
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan.
| | - Shin-Ichi Konno
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
50
|
Korczeniewska OA, Khan J, Eliav E, Benoliel R. Molecular mechanisms of painful traumatic trigeminal neuropathy-Evidence from animal research and clinical correlates. J Oral Pathol Med 2020; 49:580-589. [PMID: 32557871 DOI: 10.1111/jop.13078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 01/02/2023]
Abstract
Painful traumatic trigeminal neuropathy (PTTN) may occur following major craniofacial or oral trauma, or may be subsequent to relatively minor dental interventions. Following injury, pain may originate from a peripheral nerve, a ganglion, or from the central nervous system. In this review, we focus on molecular mechanisms of pain resulting from injury to the peripheral branch of the trigeminal nerve. This syndrome has been termed painful traumatic trigeminal neuropathy (PTTN) by the International Headache Society and replaces previous terms including atypical odontalgia, deafferentation pain, traumatic neuropathy and phantom toothache. We emphasize the scientific evidence supporting the events purported to lead to PTTN by reviewing the pathophysiology of PTTN based on relevant animal models. Additionally, we briefly overview clinical correlates and pathophysiological manifestations of PTTN.
Collapse
Affiliation(s)
- Olga A Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Junad Khan
- Eastman Institute of Oral Health, University of Rochester Medical Center, Rochester, New Jersey, USA
| | - Eli Eliav
- Eastman Institute of Oral Health, University of Rochester Medical Center, Rochester, New Jersey, USA
| | - Rafael Benoliel
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| |
Collapse
|