1
|
Zhang R, Lin J, Wang S, Yang C, Zhou C, Yang Y, Liu J, Jin X, Zhang L, Ma Y. Astragalin relieves inflammatory pain and negative mood in CFA mice by down-regulating mGluR5 signaling pathway. Sci Rep 2025; 15:5774. [PMID: 39962225 PMCID: PMC11832914 DOI: 10.1038/s41598-025-90279-7] [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: 06/10/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
As a flavonoid compound, astragalin (AST) is found in a variety of medicinal plants. In clinical studies, AST has anti-inflammatory and analgesia effects on rheumatoid arthritis, bronchopneumonia diseases and so on, but its specific role and mechanism is still not clear. This study aimed to investigate the effect and molecular mechanism of AST on inflammatory pain and pain-related emotions in complete Freund's adjuvant (CFA) mice. In this study, we observed that AST significantly alleviated CFA-induced inflammatory pain and associated emotional disturbances in mice. The mechanism may be related to down-regulating mGluR5-mediated PKCλ-ERK1/2-FOXO6 signaling pathway in CFA mice. Treatment with the mGluR5-specific inhibitor MTEP resulted in the downregulation of proteins associated with the PKCλ-ERK1/2-FOXO6 pathway, similar to the effects observed with AST administration. These results suggested that AST might play a crucial role in the management of inflammatory pain and related emotions, shedding light on its underlying mechanism for treating such conditions.
Collapse
Affiliation(s)
- Runheng Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiahong Lin
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shuhan Wang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Cuizhu Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chang Zhou
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yaqi Yang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing Liu
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiaobao Jin
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li Zhang
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yuxin Ma
- Department of Anatomy, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China.
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
2
|
Jiang X, Guo Y, Fang M, Wang X, Zhang B, Song Y, Qian J. Suppression of CGRP and TRPV1 Expression in Dorsal Root Ganglia of Knee Osteoarthritis Rats by Huojing Decoction via TrkA/MKK3/6/p38 Pathway. J Inflamm Res 2024; 17:5311-5326. [PMID: 39157588 PMCID: PMC11330260 DOI: 10.2147/jir.s472341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024] Open
Abstract
Objective Knee osteoarthritis (KOA) is a chronic condition characterized by persistent pain that can lead to severe disability. In this study, we primarily investigated the analgesic effect of Huojing decoction on MIA-induced knee arthritis. Methods The network pharmacology method was employed to acquire target information of Huojing decoction and KOA. MIA was intratibially injected to induce KOA pain in rats. Huojing decoction was then administered once daily via intragastric administration for 14 days. Pain level was assessed by paw withdrawal threshold (PWT) and paw withdrawal latency (PWL). The levels of inflammatory cytokines were determined by ELISA and PCR. TRPV1 and CGRP were detected through immunohistochemistry. The protein expression of TrkA, MKK3/6 and p38 was assessed by Western blot. Results Mechanical allodynia and thermal hyperalgesia were observed in KOA rats. The expression levels of inflammatory cytokines were significantly decreased after Huojing decoction infusion of KOA rats. TRPV1 and CGRP were reduced with treatment. Furthermore, the protein expressions of TrkA, MKK3/6 and p38 in the DRG of rats were significantly decreased. Conclusion Our data suggested that Huojing decoction can alleviate inflammation in KOA pain rats. Additionally, it can inhibit the expression of TrKA, MKK3/6 and p38 signaling pathways, indicating its analgesic effect on KOA pain rats.
Collapse
Affiliation(s)
- Xinchao Jiang
- Department of Pain Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Yinyin Guo
- Department of Oncology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Mei Fang
- Department of Pain Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Xin Wang
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Biao Zhang
- Department of Intensive Care, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Yi Song
- Department of Pain Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| | - Jianxue Qian
- Department of Pain Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
3
|
de Souza DM, Malange KF, Nishijima CM, de Melo Lima BH, Capetini VC, de Oliveira ALR, Anhê GF, Tambeli CH, Parada CA. Intraarticular monomethyl fumarate as a perspective therapy for osteoarthritis by macrophage polarization. Inflammopharmacology 2024; 32:1239-1252. [PMID: 38472616 DOI: 10.1007/s10787-024-01443-w] [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: 10/29/2023] [Accepted: 01/24/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic disease that may lead to joint structure degeneration, cartilage destruction, osteophyte formation, subchondral bone disruption, and pain. In this scenario, a higher proportion of the proinflammatory macrophage type 1 (M1) than the anti-inflammatory macrophage type 2 (M2) could be highlighted as a hallmark of OA progression. The balance between these two macrophage types emerges as a new therapeutic target in OA. This study aimed to evaluate the analgesia and macrophage profile in the treatment of experimental osteoarthritis (EOA) with systemic dimethyl fumarate (DMF) or local intra-articular monomethyl fumarate (MMF). RESULTS DMF via gavage or MMF via intra-articular in the right knee of EOA rats showed improvements in gait parameters and the nociceptive recovery of the mechanical threshold assessment by adapted electronic von Frey treatment on the twenty-first day (long-lasting phase). DMF treatment decreased proinflammatory TNF-α while increasing anti-inflammatory IL-10 cytokines from the macerated capsule on the fifth day (inflammatory phase). MMF treatment showed joint capsule mRNA extraction downregulating iNOS and TNF-α gene expression while upregulating IL-10 and MCP-1. However, CD206 was not significant but higher than untreated EOA rats' joints on the seventh day (inflammatory phase). CONCLUSIONS Our studies with EOA model induced by MIA suggest a new perspective for human treatment committed with OA based on macrophage polarization as a therapeutic target, switching the proinflammatory profile M1 to the anti-inflammatory profile M2 with DMF systematic or by MMF locally treatment according to the OA severity.
Collapse
Affiliation(s)
- Douglas Menezes de Souza
- Department of Pharmacology, School of Medical Sciences, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Tessália Vieira de Camargo, 126Campinas, Sao Paulo, 13083-887, Brazil.
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Carl Von Linnaeus, Campinas, Sao Paulo, 13083-864, Brazil.
| | - Kauê Franco Malange
- Department of Anesthesiology, University of California, La Jolla, San Diego, CA, 92037, USA
| | - Catarine Massucato Nishijima
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Carl Von Linnaeus, Campinas, Sao Paulo, 13083-864, Brazil
| | - Bruno Henrique de Melo Lima
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, Sao Paulo, 13083-862, Brazil
| | - Vinicius Cooper Capetini
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Cidade Universitária Zeferino Vaz, Rua Tessália Vieira de Camargo, 126, Campinas, Sao Paulo, 13083-887, Brazil
| | - Alexandre L R de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Monteiro Lobato, 255, Campinas, Sao Paulo, 13083-862, Brazil
| | - Gabriel Forato Anhê
- Department of Translational Medicine, School of Medical Sciences, University of Campinas, Cidade Universitária Zeferino Vaz, Rua Tessália Vieira de Camargo, 126, Campinas, Sao Paulo, 13083-887, Brazil
| | - Claudia Herrera Tambeli
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Carl Von Linnaeus, Campinas, Sao Paulo, 13083-864, Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Rua Carl Von Linnaeus, Campinas, Sao Paulo, 13083-864, Brazil.
| |
Collapse
|
4
|
Liao Z, Umar M, Huang X, Qin L, Xiao G, Chen Y, Tong L, Chen D. Transient receptor potential vanilloid 1: A potential therapeutic target for the treatment of osteoarthritis and rheumatoid arthritis. Cell Prolif 2024; 57:e13569. [PMID: 37994506 PMCID: PMC10905355 DOI: 10.1111/cpr.13569] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 11/24/2023] Open
Abstract
This study aims to determine the molecular mechanisms and analgesic effects of transient receptor potential vanilloid 1 (TRPV1) in the treatments of osteoarthritis (OA) and rheumatoid arthritis (RA). We summarize and analyse current studies regarding the biological functions and mechanisms of TRPV1 in arthritis. We search and analyse the related literature in Google Scholar, Web of Science and PubMed databases from inception to September 2023 through the multi-combination of keywords like 'TRPV1', 'ion channel', 'osteoarthritis', 'rheumatoid arthritis' and 'pain'. TRPV1 plays a crucial role in regulating downstream gene expression and maintaining cellular function and homeostasis, especially in chondrocytes, synovial fibroblasts, macrophages and osteoclasts. In addition, TRPV1 is located in sensory nerve endings and plays an important role in nerve sensitization, defunctionalization or central sensitization. TRPV1 is a non-selective cation channel protein. Extensive evidence in recent years has established the significant involvement of TRPV1 in the development of arthritis pain and inflammation, positioning it as a promising therapeutic target for arthritis. TRPV1 likely represents a feasible therapeutic target for the treatment of OA and RA.
Collapse
Affiliation(s)
- Zhidong Liao
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and MinistryGuangxi Medical UniversityNanningGuangxiChina
| | - Muhammad Umar
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
| | - Xingyun Huang
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research LaboratoryLi Ka Shing Institute of Health Sciences, The Chinese University of Hong KongHong KongChina
| | - Guozhi Xiao
- School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Yan Chen
- Department of Bone and Joint Surgerythe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Liping Tong
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
| | - Di Chen
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- Faculty of Pharmaceutical SciencesShenzhen Institute of Advanced TechnologyShenzhenChina
| |
Collapse
|
5
|
Molot J, Sears M, Anisman H. Multiple Chemical Sensitivity: It's time to catch up to the science. Neurosci Biobehav Rev 2023; 151:105227. [PMID: 37172924 DOI: 10.1016/j.neubiorev.2023.105227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 05/06/2023] [Indexed: 05/15/2023]
Abstract
Multiple chemical sensitivity (MCS) is a complex medical condition associated with low dose chemical exposures. MCS is characterized by diverse features and common comorbidities, including fibromyalgia, cough hypersensitivity, asthma, and migraine, and stress/anxiety, with which the syndrome shares numerous neurobiological processes and altered functioning within diverse brain regions. Predictive factors linked to MCS comprise genetic influences, gene-environment interactions, oxidative stress, systemic inflammation, cell dysfunction, and psychosocial influences. The development of MCS may be attributed to the sensitization of transient receptor potential (TRP) receptors, notably TRPV1 and TRPA1. Capsaicin inhalation challenge studies demonstrated that TRPV1 sensitization is manifested in MCS, and functional brain imaging studies revealed that TRPV1 and TRPA1 agonists promote brain-region specific neuronal variations. Unfortunately, MCS has often been inappropriately viewed as stemming exclusively from psychological disturbances, which has fostered patients being stigmatized and ostracized, and often being denied accommodation for their disability. Evidence-based education is essential to provide appropriate support and advocacy. Greater recognition of receptor-mediated biological mechanisms should be incorporated in laws, and regulation of environmental exposures.
Collapse
Affiliation(s)
- John Molot
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| | - Margaret Sears
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| | - Hymie Anisman
- Family Medicine, University of Ottawa Faculty of Medicine, Ottawa ON Canada; Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa Canada.
| |
Collapse
|
6
|
Alcohol-Induced Headache with Neuroinflammation: Recent Progress. FERMENTATION-BASEL 2023. [DOI: 10.3390/fermentation9020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Ethanol and other congeners in alcoholic beverages and foods are known triggers of alcohol-induced headaches (AIHs). Recent studies implicate AIHs as an important underlying factor for neuroinflammation. Studies show the relationship between alcoholic beverages, AIH agents, neuroinflammation, and the pathway they elicit. However, studies elucidating specific AIH agents’ pathways are scarce. Works reviewing their pathways can give invaluable insights into specific substances’ patterns and how they can be controlled. Hence, we reviewed the current understanding of how AIH agents in alcoholic beverages affect neuroinflammation and their specific roles. Ethanol upregulates transient receptor potential cation channel subfamily V member 1 (TRPV1) and Toll-like receptor 4 (TLR4) expression levels; both receptors trigger a neuroinflammation response that promotes AIH manifestation—the most common cause of AIHs. Other congeners such as histamine, 5-HT, and condensed tannins also upregulate TRPV1 and TLR4, neuroinflammatory conditions, and AIHs. Data elucidating AIH agents, associating pathways, and fermentation parameters can help reduce or eliminate AIH inducers and create healthier beverages.
Collapse
|
7
|
Zhang Q, Zhou M, Huo M, Si Y, Zhang Y, Fang Y, Zhang D. Mechanisms of acupuncture-electroacupuncture on inflammatory pain. Mol Pain 2023; 19:17448069231202882. [PMID: 37678839 PMCID: PMC10515556 DOI: 10.1177/17448069231202882] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 09/06/2023] [Indexed: 09/09/2023] Open
Abstract
Acupuncture, as a traditional treatment, has been extensively used in China for thousands of years. According to the World Health Organization (WHO), acupuncture is recommended for the treatment of 77 diseases. And 16 of these diseases are related to inflammatory pain. As a combination of traditional acupuncture and modern electrotherapy, electroacupuncture (EA) has satisfactory analgesic effects on various acute and chronic pain. Because of its good analgesic effects and no side effects, acupuncture has been widely accepted all over the world. Despite the increase in the number of studies, the mechanisms via which acupuncture exerts its analgesic effects have not been conclusively established. A literature review of related research is of great significance to elaborate on its mechanisms and to inform on further research directions. We elucidated on its mechanisms of action on inflammatory pain from two levels: peripheral and central. It includes the mechanisms of acupuncture in the periphery (immune cells and neurons, purinergic pathway, nociceptive ion channel, cannabinoid receptor and endogenous opioid peptide system) and central nervous system (TPRV1, glutamate and its receptors, glial cells, GABAergic interneurons and signaling molecules). In this review, we collected relevant recent studies to systematically explain the mechanisms of acupuncture in treating inflammatory pain, with a view to providing direction for future applications of acupuncture in inflammatory pain and promoting clinical development.
Collapse
Affiliation(s)
- Qingxiang Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengmeng Zhou
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mingzhu Huo
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuxin Si
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Youlin Zhang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuxin Fang
- Research Center of Experimental Acupuncture Science, College of Acumox and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| |
Collapse
|
8
|
Molot J, Sears M, Marshall LM, Bray RI. Neurological susceptibility to environmental exposures: pathophysiological mechanisms in neurodegeneration and multiple chemical sensitivity. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:509-530. [PMID: 34529912 DOI: 10.1515/reveh-2021-0043] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/13/2021] [Indexed: 05/23/2023]
Abstract
The World Health Organization lists air pollution as one of the top five risks for developing chronic non-communicable disease, joining tobacco use, harmful use of alcohol, unhealthy diets and physical inactivity. This review focuses on how host defense mechanisms against adverse airborne exposures relate to the probable interacting and overlapping pathophysiological features of neurodegeneration and multiple chemical sensitivity. Significant long-term airborne exposures can contribute to oxidative stress, systemic inflammation, transient receptor subfamily vanilloid 1 (TRPV1) and subfamily ankyrin 1 (TRPA1) upregulation and sensitization, with impacts on olfactory and trigeminal nerve function, and eventual loss of brain mass. The potential for neurologic dysfunction, including decreased cognition, chronic pain and central sensitization related to airborne contaminants, can be magnified by genetic polymorphisms that result in less effective detoxification. Onset of neurodegenerative disorders is subtle, with early loss of brain mass and loss of sense of smell. Onset of MCS may be gradual following long-term low dose airborne exposures, or acute following a recognizable exposure. Upregulation of chemosensitive TRPV1 and TRPA1 polymodal receptors has been observed in patients with neurodegeneration, and chemically sensitive individuals with asthma, migraine and MCS. In people with chemical sensitivity, these receptors are also sensitized, which is defined as a reduction in the threshold and an increase in the magnitude of a response to noxious stimulation. There is likely damage to the olfactory system in neurodegeneration and trigeminal nerve hypersensitivity in MCS, with different effects on olfactory processing. The associations of low vitamin D levels and protein kinase activity seen in neurodegeneration have not been studied in MCS. Table 2 presents a summary of neurodegeneration and MCS, comparing 16 distinctive genetic, pathophysiological and clinical features associated with air pollution exposures. There is significant overlap, suggesting potential comorbidity. Canadian Health Measures Survey data indicates an overlap between neurodegeneration and MCS (p < 0.05) that suggests comorbidity, but the extent of increased susceptibility to the other condition is not established. Nevertheless, the pathways to the development of these conditions likely involve TRPV1 and TRPA1 receptors, and so it is hypothesized that manifestation of neurodegeneration and/or MCS and possibly why there is divergence may be influenced by polymorphisms of these receptors, among other factors.
Collapse
Affiliation(s)
- John Molot
- Family Medicine, University of Ottawa Faculty of Medicine, North York, ON, Canada
| | | | | | - Riina I Bray
- Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Calceolarioside A, a Phenylpropanoid Glycoside from Calceolaria spp., Displays Antinociceptive and Anti-Inflammatory Properties. Molecules 2022; 27:molecules27072183. [PMID: 35408584 PMCID: PMC9000362 DOI: 10.3390/molecules27072183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Phenylpropanoid glycosides are a class of natural substances of plant origin with interesting biological activities and pharmacological properties. This study reports the antinociceptive and anti-inflammatory effects of calceolarioside A, a phenylpropanoid glycoside previously isolated from various Calceolaria species. In models of acute nociception induced by thermal stimuli, such as the hot plate and tail flick test, calceolarioside administered at doses of 1, 5, and 10 μg in the left cerebral ventricles did not modify the behavioral response of mice. In an inflammatory based persistent pain model as the formalin test, calceolarioside A at the high dose tested (100 μg/paw) reduced the licking activity induced by formalin by 35% in the first phase and by 75% in the second phase of the test. In carrageenan-induced thermal hyperalgesia, calceolarioside A (50 and 100 μg/paw) was able to significantly reverse thermal hyperalgesia induced by carrageenan. The anti-inflammatory activity of calceolarioside A was then assessed using the zymosan-induced paw edema model. Calceolarioside A (50 and 100 μg/paw) induced a significant reduction in the edema from 1 to 4 h after zymosan administration. Measuring IL-6, TNFα, and IL-1β pro-inflammatory cytokines released from LPS-stimulated THP-1 cells, calceolarioside A in a concentration-dependent manner reduced the release of these cytokines from THP-1 cells. Taken together, our results highlight, for the first time, the potential and selective anti-inflammatory properties of this natural-derived compound, prompting its rationale use for further investigations.
Collapse
|
10
|
Shen CL, Watkins BA, Kahathuduwa C, Chyu MC, Zabet-Moghaddam M, Elmassry MM, Luk HY, Brismée JM, Knox A, Lee J, Zumwalt M, Wang R, Wager TD, Neugebauer V. Tai Chi Improves Brain Functional Connectivity and Plasma Lysophosphatidylcholines in Postmenopausal Women With Knee Osteoarthritis: An Exploratory Pilot Study. Front Med (Lausanne) 2022; 8:775344. [PMID: 35047525 PMCID: PMC8761802 DOI: 10.3389/fmed.2021.775344] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/15/2021] [Indexed: 01/08/2023] Open
Abstract
Objective: A pre/post pilot study was designed to investigate neurobiological mechanisms and plasma metabolites in an 8-week Tai-Chi (TC) group intervention in subjects with knee osteoarthritis. Methods: Twelve postmenopausal women underwent Tai-Chi group exercise for 8 weeks (60 min/session, three times/week). Outcomes were measured before and after Tai Chi intervention including pain intensity (VAS), Brief Pain Inventory (BPI), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), plasma metabolites (amino acids and lipids), as well as resting-state functional magnetic resonance imaging (rs-fMRI, 10 min, eyes open), diffusion tensor imaging (DTI, 12 min), and structural MRI (4.5 min) in a subgroup. Clinical data was analyzed using paired t-tests; plasma metabolites were analyzed using Wilcoxon signed-rank tests; and rs-fMRI data were analyzed using seed-based correlations of the left and right amygdala in a two-level mixed-effects model (FSL software). Correlations between amygdala-medial prefrontal cortex (mPFC) connectivity and corresponding changes in clinical outcomes were examined. DTI connectivity of each amygdala was modeled using a Bayesian approach and probabilistic tractography. The associations between neurobiological effects and pain/physical function were examined. Results: Significant pre/post changes were observed with reduced knee pain (VAS with most pain: p = 0.018; WOMAC-pain: p = 0.021; BPI with worst level: p = 0.018) and stiffness (WOMAC-stiffness, p = 0.020), that likely contributed to improved physical function (WOMAC-physical function: p = 0.018) with TC. Moderate to large effect sizes pre/post increase in rs-fMRI connectivity were observed between bilateral mPFC and the amygdala seed regions (i.e., left: d = 0.988, p = 0.355; right: d = 0.600, p = 0.282). Increased DTI connectivity was observed between bilateral mPFC and left amygdala (d = 0.720, p = 0.156). There were moderate-high correlations (r = 0.28–0.60) between TC-associated pre-post changes in amygdala-mPFC functional connectivity and pain/physical function improvement. Significantly higher levels of lysophosphatidylcholines were observed after TC but lower levels of some essential amino acids. Amino acid levels (alanine, lysine, and methionine) were lower after 8 weeks of TC and many of the lipid metabolites were higher after TC. Further, plasma non-HDL cholesterol levels were lower after TC. Conclusion: This pilot study showed moderate to large effect sizes, suggesting an important role that cortico-amygdala interactions related to TC have on pain and physical function in subjects with knee osteoarthritis pain. Metabolite analyses revealed a metabolic shift of higher lyso-lipids and lower amino acids that might suggest greater fatty acid catabolism, protein turnover and changes in lipid redistribution in response to TC exercise. The results also support therapeutic strategies aimed at strengthening functional and structural connectivity between the mPFC and the amygdala. Controlled clinical trials are warranted to confirm these observed preliminary effects.
Collapse
Affiliation(s)
- Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Bruce A Watkins
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Chanaka Kahathuduwa
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Laboratory Sciences and Primary Care, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Psychiatry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Ming-Chien Chyu
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Medical Engineering, Texas Tech University, Lubbock, TX, United States
| | - Masoud Zabet-Moghaddam
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, United States
| | - Moamen M Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, United States
| | - Hui-Ying Luk
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX, United States
| | - Jean-Michel Brismée
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Rehabilitation Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Ami Knox
- Clinical Research Institute, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Jaehoon Lee
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Educational Psychology and Leadership, Texas Tech University, Lubbock, TX, United States
| | - Mimi Zumwalt
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Orthopedic Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Tor D Wager
- Department of Psychological and Brain Sciences, Dartmouth College, Hanover, NH, United States
| | - Volker Neugebauer
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
11
|
Rotpenpian N, Arayapisit T, Roumwong A, Pakaprot N, Tantisira M, Wanasuntronwong A. A standardized extract of Centella asiatica (ECa 233) prevents temporomandibular joint osteoarthritis by modulating the expression of local inflammatory mediators in mice. J Appl Oral Sci 2021; 29:e20210329. [PMID: 34705985 PMCID: PMC8523094 DOI: 10.1590/1678-7757-2021-0329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/15/2021] [Indexed: 11/29/2022] Open
Abstract
Objectives To investigate the effect of a standardized extract of Centella asiatica (ECa 233), which has anti-inflammatory properties, on the local expression of the transient receptor potential vanilloid 1 (TRPV1), the acid-sensing ion channel subunit 3 (ASIC3), and the calcitonin gene-related peptide (CGRP) in the temporomandibular joint (TMJ) structure 21 days after injecting the TMJ with complete Freund’s adjuvant (CFA). Methodology A mouse model was induced by analyzing the CFA-injected TMJ on days 7, 14, and 21. We assessed TMJ histology by the osteoarthritis cartilage grade score. Then, we observed the effect of different ECa 233 concentrations (30, 100, and 300 mg/kg) and of 140 mg/kg ibuprofen doses on TRPV1, ASIC3, and CGRP local expression on day 21. Results Osteoarthritis cartilage scores were 1.17±0.37 and 3.83±0.68 on days 14 and 21, respectively, in the CFA group (n=5). On day 21, TRPV1, ASIC3, and CGRP expression significantly increased in the CFA group. In the ibuprofen-treated group, TRPV1 expression significantly decreased, but ASIC3 and CGRP showed no significant difference. All ECa 233 doses reduced TRPV1 expression, but the 100 mg/kg ECa 233 dose significantly decreased ASIC3 expression. Conclusions TRPV1, ASIC3, and CGRP expression increased in mice with TMJ-OA on day 21. All ECa 233 and ibuprofen doses inhibited pathogenesis by modulating the local expression of TRPV1 and ASIC3. Therefore, ECa 233 was more effective than ibuprofen.
Collapse
Affiliation(s)
| | | | - Atitaya Roumwong
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Narawut Pakaprot
- Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mayuree Tantisira
- Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | | |
Collapse
|
12
|
Soga M, Izumi T, Nanchi I, Horita N, Yamamoto M, Kawasaki S, Ogawa K, Fujita M, Morioka Y. Suppression of joint pain in transient receptor potential vanilloid 4 knockout rats with monoiodoacetate-induced osteoarthritis. Pain Rep 2021; 6:e951. [PMID: 34396019 PMCID: PMC8357256 DOI: 10.1097/pr9.0000000000000951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/16/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. Knee joint pain in osteoarthritis model rats is caused by the sensitization of transient receptor potential vanilloid 4 in the dorsal root ganglion neurons Introduction: Transient receptor potential vanilloid 4 (TRPV4) modulates osteoarthritic (OA) pain in animal models. However, the pathophysiological function of TRPV4 in regulating OA pain remains poorly understood. Methods: We developed TRPV4-knockout (TRPV4-KO) rats and assessed the effects of Trpv4 gene deficiency in a monoiodoacetate (MIA)-induced OA pain model (MIA rats) by examining pain-related behavior, pathological changes, and electrophysiological changes in dorsal root ganglion (DRG) neurons. The changes detected in TRPV4-KO rats were confirmed in wild-type rats using a TRPV4 antagonist. Results: Transient receptor potential vanilloid 4–KO rats showed the same pain threshold as wild-type rats for thermal or pressure stimuli under normal conditions. Trpv4 gene deletion did not suppress the development of osteoarthritis pathologically in MIA rats. However, the OA-related mechanical pain behaviors observed in MIA rats, including decreased grip strength, increased mechanical allodynia, and reduced weight-bearing on the ipsilateral side, were completely suppressed in TRPV4-KO rats. The DRG neurons in wild-type but not TRPV4-KO MIA rats were depolarized with increased action potentials. Transient receptor potential vanilloid 4 antagonist treatments recapitulated the effects of genetic Trpv4 deletion. Conclusion: Transient receptor potential vanilloid 4 was sensitized in the DRG neurons of MIA rats and played a critical role in the development of OA pain. These results suggest that the inhibition of TRPV4 might be a novel potent analgesic strategy for treating OA pain.
Collapse
Affiliation(s)
- Masahiko Soga
- Department of Pharmacological Efficacy Evaluation, Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Japan
| | - Takaya Izumi
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Isamu Nanchi
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Narumi Horita
- Department of Pharmacological Efficacy Evaluation, Shionogi TechnoAdvance Research Co. Ltd., Toyonaka, Japan
| | - Miyuki Yamamoto
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Shiori Kawasaki
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Koichi Ogawa
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Masahide Fujita
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| | - Yasuhide Morioka
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co. Ltd., Toyonaka, Japan
| |
Collapse
|
13
|
Hwang HS, Park IY, Hong JI, Kim JR, Kim HA. Comparison of joint degeneration and pain in male and female mice in DMM model of osteoarthritis. Osteoarthritis Cartilage 2021; 29:728-738. [PMID: 33609695 DOI: 10.1016/j.joca.2021.02.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 01/13/2021] [Accepted: 02/08/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE While the prevalence of radiographic and symptomatic osteoarthritis (OA) is higher in women, male mice are more frequently used in animal experiments to explore its pathogenesis or drug efficacy. In this study, we examined whether sexual dimorphism affects pain and joint degeneration in destabilization of the medial meniscus (DMM) mouse model. METHODS DMM or sham surgery was performed on the knee of male and female C57BL/6 mice. Joint damage was assessed by safranin O staining and scored using the Osteoarthritis Research Society International (OARSI) scoring system. Von Frey hair, incapacitance, and rotarod tests were conducted to measure joint pain. The analgesic effect of capsazepine (CPZ), a TRPV1 antagonist, was compared between male and female mice. RESULTS Histology and OARSI scoring analysis showed that cartilage degeneration developed, and progressed in both male and female DMM groups, however, damage was less severe in females at the late stage of OA. Pain behavior, as measured by mechanical allodynia, was displayed for longer in male DMM mice compared to females. Incapacitance data showed that CPZ significantly reduced DMM-induced pain in male mice but not in female mice. Immunofluorescence microscopy analysis demonstrated that DMM surgery increased the expression of TRPV1 in both female and male dorsal root ganglion (DRG). Injection of CPZ significantly suppressed TRPV1 expression in the DRG of male mice only. CONCLUSION Joint damage develops comparably in both female and male mice after DMM although it progresses less in females. There was a subtle sex difference in pain behaviors and analgesic efficacy of a TRPV1 antagonist, which was accompanied by a differential regulation of TPRV1.
Collapse
Affiliation(s)
- H S Hwang
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Kyunggi, 14068, South Korea; Institute for Skeletal Aging, Hallym University, Chunchon, 24251, South Korea
| | - I Y Park
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Kyunggi, 14068, South Korea; Institute for Skeletal Aging, Hallym University, Chunchon, 24251, South Korea
| | - J I Hong
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Kyunggi, 14068, South Korea; Institute for Skeletal Aging, Hallym University, Chunchon, 24251, South Korea
| | - J R Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Kyunggi, 14068, South Korea; Institute for Skeletal Aging, Hallym University, Chunchon, 24251, South Korea
| | - H A Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Kyunggi, 14068, South Korea; Institute for Skeletal Aging, Hallym University, Chunchon, 24251, South Korea.
| |
Collapse
|
14
|
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
|
15
|
Li M, Zhang X, Li C, Liu Y, Yang S, Xu S. Galanin Receptor 2 Is Involved in Galanin-Induced Analgesic Effect by Activating PKC and CaMKII in the Nucleus Accumbens of Inflammatory Pain Rats. Front Neurosci 2021; 14:593331. [PMID: 33551722 PMCID: PMC7859109 DOI: 10.3389/fnins.2020.593331] [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: 08/10/2020] [Accepted: 12/28/2020] [Indexed: 11/13/2022] Open
Abstract
It has been reported that galanin has an analgesic effect via activating galanin receptors (GALRs). This study focused on the involvement of GALR2 in the galanin-induced analgesic effect and its signaling mechanism in the nucleus accumbens (NAc) of inflammatory rats. Animal models were established through injecting carrageenan into the plantar of rats’ left hind paw. The results showed that GALR2 antagonist M871 weakened partially the galanin-induced increases in hind paw withdrawal latency (HWL) to thermal stimulation and hind paw withdrawal threshold (HWT) to mechanical stimulation in NAc of inflammatory rats. Moreover, the GALR2 agonist M1145 prolonged the HWL and HWT, while M871 blocked the M1145-induced increases in HWL and HWT. Western blotting showed that the phosphorylation of calcium/calmodulin-dependent protein kinase II (p-CaMKII) and protein kinase C (p-PKC) in NAc were upregulated after carrageenan injection, while p-PKC and p-CaMKII were downregulated after intra-NAc administration of M871. Furthermore, the CaMKII inhibitor KN93 and PKC inhibitor GO6983 attenuated M1145-induced increases in HWL and HWT in NAc of rats with inflammatory pain. These results prove that GALR2 is involved in the galanin-induced analgesic effect by activating CaMKII and PKC in NAc of inflammatory pain rats, implying that GALR2 agonists probably are potent therapeutic options for inflammatory pain.
Collapse
Affiliation(s)
- Mengnan Li
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Xiaomin Zhang
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Chongyang Li
- Department of Oncology, Affiliated Hospital, Yunnan University, Kunming, China
| | - Yanan Liu
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Shuang Yang
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| | - Shilian Xu
- Department of Physiology, School of Basic Medicine, Kunming Medical University, Kunming, China
| |
Collapse
|
16
|
Jendzjowsky NG, Roy A, Iftinca M, Barioni NO, Kelly MM, Herrington BA, Visser F, Altier C, Wilson RJA. PKCε stimulation of TRPV1 orchestrates carotid body responses to asthmakines. J Physiol 2020; 599:1335-1354. [PMID: 33180962 PMCID: PMC7898719 DOI: 10.1113/jp280749] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
Abstract
Key points We have previously shown that carotid body stimulation by lysophosphatidic acid elicits a reflex stimulation of vagal efferent activity sufficient to cause bronchoconstriction in asthmatic rats. Here, we show that pathophysiological concentrations of asthma‐associated prototypical Th2 cytokines also stimulate the carotid bodies. Stimulation of the carotid bodies by these asthmakines involves a PKCε–transient receptor potential vanilloid 1 (TRPV1) signalling mechanism likely dependent on TRPV1 S502 and T704 phosphorylation sites. As the carotid bodies’ oxygen sensitivity is independent of PKCε–TRPV1 signalling, systemic blockade of PKCε may provide a novel therapeutic target to reduce allergen‐induced asthmatic bronchoconstriction. Consistent with the therapeutic potential of blocking the PKCε–TRPV1 pathway, systemic delivery of a PKCε‐blocking peptide suppresses asthmatic respiratory distress in response to allergen and reduces airway hyperresponsiveness to bradykinin.
Abstract The autonomic nervous system orchestrates organ‐specific, systemic and behavioural responses to inflammation. Recently, we demonstrated a vital role for lysophosphatidic acid in stimulating the primary autonomic oxygen chemoreceptors, the carotid bodies, in parasympathetic‐mediated asthmatic airway hyperresponsiveness. However, the cacophony of stimulatory factors and cellular mechanisms of carotid body activation are unknown. Therefore, we set out to determine the intracellular signalling involved in carotid body‐mediated sensing of asthmatic blood‐borne inflammatory mediators. We employed a range of in vitro and rat in situ preparations, site‐directed mutagenesis, patch‐clamp, nerve recordings and pharmacological inhibition to assess cellular signalling. We show that the carotid bodies are also sensitive to asthma‐associated prototypical Th2 cytokines which elicit sensory nerve excitation. This provides additional asthmatic ligands contributing to the previously established reflex arc resulting in efferent vagal activity and asthmatic bronchoconstriction. This novel sensing role for the carotid body is mediated by a PKCε‐dependent stimulation of transient receptor potential vanilloid 1 (TRPV1), likely via TRPV1 phosphorylation at sites T704 and S502. Importantly, carotid body oxygen sensing was unaffected by blocking either PKCε or TRPV1. Further, we demonstrate that systemic PKCε blockade reduces asthmatic respiratory distress in response to allergen and airway hyperresponsiveness. These discoveries support an inflammation‐dependent, oxygen‐independent function for the carotid body and suggest that targeting PKCε provides a novel therapeutic option to abate allergic airway disease without altering life‐saving autonomic hypoxic reflexes. We have previously shown that carotid body stimulation by lysophosphatidic acid elicits a reflex stimulation of vagal efferent activity sufficient to cause bronchoconstriction in asthmatic rats. Here, we show that pathophysiological concentrations of asthma‐associated prototypical Th2 cytokines also stimulate the carotid bodies. Stimulation of the carotid bodies by these asthmakines involves a PKCε–transient receptor potential vanilloid 1 (TRPV1) signalling mechanism likely dependent on TRPV1 S502 and T704 phosphorylation sites. As the carotid bodies’ oxygen sensitivity is independent of PKCε–TRPV1 signalling, systemic blockade of PKCε may provide a novel therapeutic target to reduce allergen‐induced asthmatic bronchoconstriction. Consistent with the therapeutic potential of blocking the PKCε–TRPV1 pathway, systemic delivery of a PKCε‐blocking peptide suppresses asthmatic respiratory distress in response to allergen and reduces airway hyperresponsiveness to bradykinin.
Collapse
Affiliation(s)
- Nicholas G Jendzjowsky
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Arijit Roy
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nicole O Barioni
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Margaret M Kelly
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Brittney A Herrington
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Frank Visser
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Richard J A Wilson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
17
|
Braga Ferreira LG, Faria JV, dos Santos JPS, Faria RX. Capsaicin: TRPV1-independent mechanisms and novel therapeutic possibilities. Eur J Pharmacol 2020; 887:173356. [DOI: 10.1016/j.ejphar.2020.173356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 02/08/2023]
|
18
|
Relationship between Pain Behavior and Changes in KCNA2 Expression in the Dorsal Root Ganglia of Rats with Osteoarthritis. Pain Res Manag 2020. [DOI: 10.1155/2020/4636838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. To investigate the relationship between pain behavior and potassium voltage-gated channel subfamily A member 2 (KCNA2) expression in dorsal root ganglia (DRGs) of rats with osteoarthritis (OA). Methods. Male Sprague-Dawley rats were randomly divided into three groups: blank control group (group C), normal saline group (group S), and group OA. Paw withdrawal mechanical threshold (PWMT) and paw withdrawal thermal latency (PWTL) were measured one day before injection and one, two, four, and six weeks after injection. At one, two, four, and six weeks after injection, pathological knee joint changes and activated transcription factor-3 (ATF-3) and KCNA2 expressions in DRGs were analyzed. Results. Compared with preinjection, PWMT and PWTL at two, four, and six weeks after injection were significantly decreased in the group OA (P<0.05 or 0.01). Compared with group C, PWMT and PWTL at two, four, and six weeks after injection were significantly decreased in the group OA (P<0.05 or 0.01). In the group OA, slight local articular cartilage surface destruction was found at week one. The cartilage surface destruction gradually developed, and the exacerbation of cartilage matrix reduction and bone hyperplasia were increasingly aggravated and eventually evolved into advanced OA in the second to sixth weeks. Compared with group C, ATF-3 expression was significantly increased, and KCNA2 expression was significantly decreased in the group OA at two, four, and six weeks after injection (P<0.05 or 0.01). Compared to baseline, ATF-3 expression was significantly increased, and KCNA2 expression was significantly decreased in the group OA (P<0.05 or 0.01). Conclusion. Pain behavior in OA rats was associated with decreased KCNA2 expression in DRGs.
Collapse
|
19
|
Mehrabadi S, Karimiyan SM, Ashabi G, Moradbeygi K, Hoseini M. Repeated Administration of Baclofen Modulates TRPV-1 Channel Expression by PKC Pathway in Dorsal Root Ganglia of Spinal Cord in a Morphine Tolerance Model of Rats. IRANIAN BIOMEDICAL JOURNAL 2020; 24:379-85. [PMID: 32660223 PMCID: PMC7601548 DOI: 10.29252/ibj.24.6.374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background: Tolerance and dependence to anti-nociceptive effect of morphine restricted its use. Nowadays co-administration of morphine and other drugs suggests diminishing this tolerance. Baclofen is one of the drugs that may be beneficial in the attenuation of tolerance to morphine. Studies have shown that changes in TRPV-1 expression during administration of morphine have a pivotal role in developing morphine tolerance. Therefore, the effect of baclofen on TRPV-1 expression during chronic administration of morphine was investigated in this study. Methods: A total of 48 rats were divided into four groups of control, morphine single injection, morphine tolerance, and morphine tolerance + baclofen. To induce morphine tolerance in rats, animals received 10 mg/kg of i.p. morphine sulfate once a day for 10 days. In the treatment group, baclofen (0.5 mg/kg) was injected for 10 days, before morphine injection. Finally, to evaluate baclofen treatment on morphine analgesia and hyperalgesia, thermal hyperalgesia and formalin test were used. TRPV-1 and PKC expression and protein production in DRG of spinal cord were then evaluated by real-time PCR and Western blot. Results: In baclofen treatment group, thermal hyperalgesia and formalin test improved in comparison with morphine tolerance group. In morphine tolerance group, both TRPV-1/PKC gene expression and protein levels increased in comparison with the control group. However, following the baclofen treatment, the TRPV-1 and PKC levels decreased. Conclusion: Baclofen can enhance anti-nociceptive effect of morphine by modulating TRPV-1 channel and PKC activity.
Collapse
Affiliation(s)
- Shima Mehrabadi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Morteza Karimiyan
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghorbangol Ashabi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Moradbeygi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Nursing, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Marjan Hoseini
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Hu W, Ding Y, Li Q, shi R, He Y. Transient receptor potential vanilloid 4 channels as therapeutic targets in diabetes and diabetes-related complications. J Diabetes Investig 2020; 11:757-769. [PMID: 32129549 PMCID: PMC7378409 DOI: 10.1111/jdi.13244] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/21/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
With an estimated 425 million diabetes patients worldwide in 2019, type 2 diabetes has reached a pandemic proportion and represents a major unmet medical need. A key determinant of the development and progression of type 2 diabetes is pancreatic -cell dysfunction, including the loss of cell mass, the impairment of insulin biosynthesis and inadequate exocytosis. Recent studies have shown that transient receptor potential vanilloid 4 (TRPV4), a Ca2+ -permeable non-selective cation channel, is involved in -cell replication, insulin production and secretion. TRPV4 agonists have insulinotropic activity in pancreatic -cell lines, but the prolonged activation of TRPV4 leads to -cell dysfunction and death. In addition, TRPV4 is involved in a wide variety of pathophysiological activities, and has been reported to play an important role in diabetes-related complications, such as obesity, cardiovascular diseases, diabetic retinopathy, nephropathy and neuropathy. In a rodent type 2 diabetes model, Trpv4 agonists promote vasodilation and improve cardiovascular function, whereas Trpv4 antagonists reduce high-fat diet-induced obesity, insulin resistance, diabetic nephropathy, retinopathy and neuropathy. These findings raise interest in using TRPV4 as a therapeutic target for type 2 diabetes. In this review, we intend to summarize the latest findings regarding the role of TRPV4 in diabetes as well as diabetes-related conditions, and to evaluate its potential as a therapeutic target for diabetes and diabetes-related diseases.
Collapse
Affiliation(s)
- Wei Hu
- Department of Epidemiology and Medical StatisticsInstitute of Medical Systems BiologyGuangdong Medical UniversityDongguanChina
| | - Yuanlin Ding
- Department of Epidemiology and Medical StatisticsInstitute of Medical Systems BiologyGuangdong Medical UniversityDongguanChina
| | - Qingqing Li
- Department of Epidemiology and Medical StatisticsInstitute of Medical Systems BiologyGuangdong Medical UniversityDongguanChina
| | - Rou shi
- Department of Epidemiology and Medical StatisticsInstitute of Medical Systems BiologyGuangdong Medical UniversityDongguanChina
| | - Yuqing He
- Department of Epidemiology and Medical StatisticsInstitute of Medical Systems BiologyGuangdong Medical UniversityDongguanChina
- Liaobu HospitalGuangdong Medical UniversityDongguanChina
| |
Collapse
|
21
|
Sun L, Wang G, He M, Mei Z, Zhang F, Liu P. Effect and mechanism of the CACNA2D1-CGRP pathway in osteoarthritis-induced ongoing pain. Biomed Pharmacother 2020; 129:110374. [PMID: 32570114 DOI: 10.1016/j.biopha.2020.110374] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 12/27/2022] Open
Abstract
This study built an OA model in rats by monosodium iodoacetate (MIA) injection to determine the effects and mechanism of the voltage-dependent calcium channel subunit alpha-2/delta-1 (CACNA2D1)-calcitonin gene-related protein (CGRP) pathway in osteoarthritis (OA)-induced ongoing pain. CACNA2D1 expression was measured by qPCR assay, western blotting assay, and immunofluorescence. Pain behaviors in rats were assessed with the measurement of thermal paw withdrawal latency (PWL) and mechanical paw withdrawal threshold (PWT). The expression of CACNA2D1, neuropeptide Y (NPY), activating transcription factor 3 (ATF3), CGRP, protein kinase A (PKA), phosphorylated (p)-PKA, adenylyl cyclase (AC), protein kinase C (PKC), p-PKC, phospholipase C (PLC), and mitogen-activated protein kinase (MAPK) signaling pathway proteins were measured, OA rats had higher CACNA2D1 expression than normal rats. Knockdown of CACNA2D1 led to the elevation of the pain threshold of OA rats, and CACNA2D1 over-expression decreased the pain threshold of normal rats. Moreover, CACNA2D1 over-expression inhibited the expression of CGRP, up-regulated the expressions of NPY, ATF3, p-PKA, AC, p-PKC, PLC, p-Jun N-terminal kinase (JNK), and p-p38, and had no significant effect on phosphorylated extracellular signal-regulated kinase (p-ERK) expression in vivo and in vitro. Using this model of MIA-induced OA, we demonstrated that CACNA2D1 might be involved in the process of pain by modulating the CGRP and AC-PKA/PKC/MAPK signaling pathways in the dorsal root ganglion.
Collapse
Affiliation(s)
- Liang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Guodong Wang
- Departments of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Meifang He
- Laboratory of General Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhigang Mei
- Guangzhou Sihe Biotechnology Co., Ltd., Guangzhou, China
| | - Fazhou Zhang
- Guangzhou Sihe Biotechnology Co., Ltd., Guangzhou, China
| | - Ping Liu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
22
|
Yoneda S, Kasai E, Matsuo M, Tamano R, Sakurai Y, Asaki T, Fujita M. Duloxetine ameliorates the impairment of diffuse noxious inhibitory control in rat models of peripheral neuropathic pain and knee osteoarthritis pain. Neurosci Lett 2020; 729:134990. [PMID: 32315711 DOI: 10.1016/j.neulet.2020.134990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Diffuse noxious inhibitory control (DNIC) is a phenomenon to reflect descending pain modulation in animals. Conditioned pain modulation (CPM) is the human counterpart of DNIC and is reduced in patients with several chronic pain conditions. Duloxetine is a serotonin and noradrenaline reuptake inhibitor that ameliorates CPM impairment in patients with diabetic neuropathy. Although some studies have reported the effects of different pharmacological agents on CPM, few studies have compared the effects of some analgesics in both humans and rodents. Therefore, we established a stable evaluation method for DNIC in rats and determined whether duloxetine and other specific analgesics affect DNIC impairment in rat models of peripheral neuropathic pain and osteoarthritis pain, two types of chronic pain. As a conditioning stimulus, capsaicin was injected into the forepaw of rats. The paw withdrawal threshold (PWT) in response to mechanical pressure was measured for the hindpaw. Peripheral neuropathic pain and osteoarthritis pain models were developed by partial sciatic nerve ligation (PSNL) and the intra-articular injection of 2 mg monoiodoacetate (MIA), respectively. Capsaicin (30-100 μg/site) increased the PWT, in a dose-dependent manner, in naive rats. The threshold significantly increased at 30 μg and reached its maximal level at 100 μg. The change in PWT following capsaicin injection was significantly reduced in PSNL-treated rats, but the threshold was increased by the subcutaneous administration of duloxetine (10 mg/kg). The oral administrations of pregabalin (10 mg/kg) and celecoxib (3 mg/kg) did not affect the PWT in PSNL-treated rats. Similarly, MIA-injected rats also showed a reduced change in PWT following capsaicin injection. Duloxetine, but not pregabalin and celecoxib, significantly increased the PWT in MIA-injected rats. These results suggested that duloxetine can directly ameliorate DNIC impairment in rat models of chronic pain. Duloxetine may be useful for modulating chronic pain by restoring function to the endogenous, descending, inhibitory pathway.
Collapse
Affiliation(s)
- Sosuke Yoneda
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Erika Kasai
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Midori Matsuo
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Ryuta Tamano
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Yusuke Sakurai
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Toshiyuki Asaki
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Masahide Fujita
- Neuroscience, Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., 1-1 3chome, Futaba-cho, Toyonaka, Osaka, 561-0825, Japan.
| |
Collapse
|
23
|
Bai Q, Shao J, Cao J, Ren X, Cai W, Su S, George S, Tan Z, Zang W, Dong T. Protein kinase C-α upregulates sodium channel Nav1.9 in nociceptive dorsal root ganglion neurons in an inflammatory arthritis pain model of rat. J Cell Biochem 2019; 121:768-778. [PMID: 31385361 DOI: 10.1002/jcb.29322] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/15/2019] [Indexed: 12/31/2022]
Abstract
Previous studies have found that increased expression of Nav1.9 and protein kinase C (PKC) contributes to pain hypersensitivity in a couple of inflammatory pain models. Here we want to observe if PKC can regulate the expression of Nav1.9 in dorsal root ganglion (DRG) in rheumatoid arthritis (RA) pain model. A chronic knee joint inflammation model was produced by intra-articular injection of the complete Freund's adjuvant (CFA) in rats. Nociceptive behaviors including mechanical, cold, and heat hyperalgesia were examined. The expression of Nav1.9 and PKCα in DRG was detected by a quantitative polymerase chain reaction, Western blot, and immunofluorescence. The in vitro and in vivo effects of a PKC activator (phorbol 12-myristate 13-acetate [PMA]) and a PKC inhibitor (GF-109203X) on the expression of Nav1.9 were examined. Moreover, the effects of PKC modulators on nociceptive behaviors were studied. Increased mechanical, heat, and cold sensitivity was observed 3 to 14 days after CFA injection. Parallel increases in messenger RNA and protein expression of Nav1.9 and PKCα were found. Immunofluorescence experiments found that Nav1.9 was preferentially colocalized with IB4+DRG neurons in RA rats. In cultured DRG neurons, PMA increased Nav1.9 expression while GF-109203X prevented the effect of PMA. PMA increased Nav1.9 expression in naïve rats while GF-109203X decreased Nav1.9 expression in RA rats. In naïve rats, PMA caused mechanical and cold hyperalgesia. On the other hand, GF-109203X attenuated mechanical and cold hyperalgesia in RA-pain model. Nav1.9 might be upregulated by PKCα in DRG, which contributes to pain hypersensitivity in CFA-induced chronic knee joint inflammation model of RA pain.
Collapse
Affiliation(s)
- Qian Bai
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Henan, China
| | - Jinping Shao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Jing Cao
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Xiuhua Ren
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Weihua Cai
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Songxue Su
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Sanjeeth George
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas
| | - Zhiyong Tan
- Department of Pharmacology and Toxicology and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana
| | - Weidong Zang
- Department of Human Anatomy, School of Basic Medicine, Zhengzhou University, Henan, China
| | - Tieli Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Henan, China
| |
Collapse
|
24
|
Contribution of synovial macrophages to rat advanced osteoarthritis pain resistant to cyclooxygenase inhibitors. Pain 2019; 160:895-907. [PMID: 30585984 DOI: 10.1097/j.pain.0000000000001466] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Most advanced knee osteoarthritis (OA) patients experience chronic pain resistant to cyclooxygenase (COX) inhibitors. However, the cells and molecules involved in this advanced OA pain remain poorly understood. In this study, we developed a rat model of advanced knee OA by modification of the monoiodoacetate-induced OA pain model and examined involvement of synovial macrophages in advanced OA pain. Cyclooxygenase inhibitors, such as celecoxib and naproxen, and a steroid were ineffective, but an opioid and anti-nerve growth factor (NGF) antibody was effective for pain management in the advanced OA model. Similar to advanced OA patients, histological analysis indicated severe bone marrow damages, synovitis, and cartilage damage and an increase of macrophages with high expression of interleukin-1β, NGF, nitric oxide synthase (NOS) 1, NOS2, and COX-2 in the knee joint of the advanced OA model. Intravenous injection of clodronate liposomes depleted synovial macrophages, which decreased the level of not only proinflammatory mediator interleukin-1β but also NGF in the knee joint, leading to pain suppression in the advanced OA model. These data suggest the involvement of synovial macrophages in advanced knee OA pain resistant to COX inhibitors by increasing proinflammatory mediators, and that drugs targeting synovial macrophages might have potent analgesic effects.
Collapse
|
25
|
Sensitization of transient receptor potential vanilloid 4 and increasing its endogenous ligand 5,6-epoxyeicosatrienoic acid in rats with monoiodoacetate-induced osteoarthritis. Pain 2019; 159:939-947. [PMID: 29438227 DOI: 10.1097/j.pain.0000000000001169] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) receptor modulates pain, and this has been noted in several animal models. However, the involvement of TRPV4 in osteoarthritic (OA) pain remains poorly understood. This study assessed the functional changes in TRPV4 and the expression of its endogenous ligand 5,6-epoxyeicosatrienoic acid (5,6-EET) in a rat monoiodoacetate (MIA)-induced OA pain model (MIA rats). Monoiodoacetate-treated rats showed reduced grip strength as compared to sham-treated rats, and this loss in function could be recovered by the intraarticular administration of a TRPV4 antagonist (HC067047 or GSK2193874). By contrast, the intraarticular administration of the TRPV4 agonist, GSK1016790A, increased the pain-related behaviors in MIA rats but not in sham rats. TRPV4 expression was not increased in knee joints of MIA rats; however, the levels of phosphorylated TRPV4 at Ser824 were increased in dorsal root ganglion neurons. In addition, 5,6-EET was increased in lavage fluids from the knee joints of MIA rats and in meniscectomy-induced OA pain model rats. 5,6-EET and its metabolite were also detected in synovial fluids from patients with OA. In conclusion, TRPV4 was sensitized in the knee joints of MIA rats through phosphorylation in dorsal root ganglion neurons, along with an increase in the levels of its endogenous ligand 5,6-EET. The analgesic effects of the TRPV4 antagonist in the OA pain model rats suggest that TRPV4 may be a potent target for OA pain relief.
Collapse
|
26
|
[Expression of KCNA2 in the dorsal root ganglia of rats with osteoarthritis pain induced by monoiodoacetate]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:579-585. [PMID: 31140423 PMCID: PMC6743928 DOI: 10.12122/j.issn.1673-4254.2019.05.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To investigate the changes in the expression of voltage-gated potassium channel subunit KCNA2 in the dorsal root ganglion (DRG) neurons of rats with osteoarthritis (OA) pain induced by sodium monoiodoacetate and explore the mechanism. METHODS A total of 156 adult male Sprague-Dawley rats were randomly divided into blank control group, saline group and intra-articular monoiodoacetate injection-induced OA group. The paw withdrawal mechanical threshold (PWMT) was measured before and at 1, 2, 4, and 6 weeks after monoiodoacetate injection. At 4 weeks after the injection, the pathological changes in the knee joints were analyzed using HE staining and Safranin O-Fast Green staining, and the expression of activating transcription factor 3 (ATF-3) and inducible nitric oxide synthase (iNOS) in the DRG neurons were detected by immunofluorescence staining. The expression of Kcna2 mRNA in the DRG neurons was detected by RT-qPCR at 1, 2, 4 and 6 weeks after the injection. The expression of KCNA2 in the DRG was measured by Western blotting, and the methylation level of Kcna2 promoter region was measured by MSPCR at 4 weeks after the injection. RESULTS The PWMT of the rats in OA group was significantly decreased at 2, 4, and 6 weeks after the injection as compared with the baseline (P < 0.05 or P < 0.001) as well as the control group (P < 0.05 or P < 0.001). Four weeks after the intra-articular injection, fractures and defects on the surface of the articular cartilage, bone hyperplasia, and blurred tidal line were observed in the rats in OA group, but no obvious pathological changes were detected in the control or saline groups. Compared with those in the control group, the expressions of ATF-3 and iNOS were significantly increased (P < 0.01) at 4 weeks after injection; the expression of Kcna2 mRNA at 2, 4 and 6 weeks and the expression of KCNA2 protein at 4 weeks were all significantly decreased (P < 0.05 or P < 0.01), and the methylation level of Kcna2 gene was significantly increased at 4 weeks after the injection in OA group (P < 0.01). CONCLUSIONS The expression of KCNA2 is decreased in the DRG neurons of rats with OA pain likely as a result of enhanced methylation of Kcna2 promoter region.
Collapse
|
27
|
Riccio D, Andersen HH, Arendt-Nielsen L. Antipruritic effects of transient heat stimulation on histaminergic and nonhistaminergic itch. Br J Dermatol 2019; 181:786-795. [PMID: 30802929 DOI: 10.1111/bjd.17825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND Chronic itch is notoriously difficult to treat. Counterstimuli are able to inhibit itch, but this principle is difficult to apply in clinical practice, and the mechanisms behind counterstimulation-induced itch suppression in humans are unclear. OBJECTIVES Firstly, to analyse the stimulus-response effects of transient heat stimuli on histaminergic and nonhistaminergic itch, and secondly, to investigate whether the antipruritic effect depends on homotopic (peripheral mediation) or heterotopic (central mediation) counterstimulation relative to the itch provocation site. METHODS Eighteen healthy volunteers participated (eight female, mean age 25·7 ± 0·8 years). Itch was evoked on premarked areas of the volar forearms, by either histamine (1% solution) or cowhage (35-40 spicules). In addition to the itch provocations (experiment 1), 5-s homotopic heat stimuli at 32, 40, 45 or 50 °C were applied. In experiment 2, heat stimuli were applied either homotopically, intrasegmentally (next to the provocation site) or extrasegmentally (dorsal forearm). Itch intensity was evaluated throughout the procedures using a digital visual analogue scale. RESULTS Homotopic counterstimuli inhibited histaminergic itch by 41·3% at 45 °C (P < 0·01) and by 76·7% at 50 °C (P < 0·001). Cowhage-induced itch was less prone to counterstimulation and was significantly diminished only at 50 °C, by 43·6% (P = 0·009). Counterstimulations applied heterotopically were not able to inhibit itch significantly. CONCLUSIONS Itch pathway-specific effects of counterstimuli were observed between homo- and heterotopic stimulation. Histaminergic itch was robustly inhibited by short-term homotopic noxious heat stimuli for up to 10 min. Nonhistaminergic itch was only weakly inhibited. The inhibitory effects exerted by the short-term heat stimuli only occurred following homotopic counterstimulation.
Collapse
Affiliation(s)
- D Riccio
- Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark.,Laboratory for Experimental Cutaneous Pain and Itch Research, SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - H H Andersen
- Laboratory for Experimental Cutaneous Pain and Itch Research, SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - L Arendt-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Aalborg University, Aalborg, Denmark.,Laboratory for Experimental Cutaneous Pain and Itch Research, SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
28
|
Inomata K, Tsuji K, Onuma H, Hoshino T, Udo M, Akiyama M, Nakagawa Y, Katagiri H, Miyatake K, Sekiya I, Muneta T, Koga H. Time course analyses of structural changes in the infrapatellar fat pad and synovial membrane during inflammation-induced persistent pain development in rat knee joint. BMC Musculoskelet Disord 2019; 20:8. [PMID: 30611247 PMCID: PMC6320593 DOI: 10.1186/s12891-018-2391-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/21/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common joint disease in aging societies, which is accompanied by chronic inflammation and degeneration of the joint structure. Inflammation of the infrapatellar fat pad (IFP) and synovial membrane (IFP surface) plays essential roles in persistent pain development in patients with OA. To identify the point during the inflammatory process critical for persistent pain development, we performed a time course histological analysis in a rat arthritis model. METHODS Wistar rats received single intra-articular injection of monoiodoacetic acid (MIA, 0.2 or 1.0 mg/30 μL) in the right knees or phosphate-buffered saline (PBS, 30 μL) as a control in the left knees. Pain avoidance behaviors (weight-bearing asymmetry and tactile hypersensitivity of the plantar surface of the hind paw) were evaluated on days 0, 1, 3, 5, 7, and 14 after injection. Histological assessments of the knee joint were performed on days 0, 1, 3, 5, and 7 after MIA injection. RESULTS Weight-bearing asymmetry was observed along with the onset of acute inflammation in both the low- (0.2 mg) and high-dose (1.0 mg) groups. In the low-dose group, weight-bearing asymmetry was completely reversed on day 10, indicating that joint pain seemed to alleviate between days 7 and 10. In contrast, we observed persistent joint pain after day 10 in the high-dose group. Histological assessments of the high-dose group indicated that the initial sign of inflammatory responses was observed in the perivascular region inside the IFP. Inflammatory cell infiltration from the perivascular region to the parenchymal region of the IFP was observed on day 3 and reached the IFP surface (synovial membrane) on day 7. Extensive fibrosis throughout the IFP was observed between days 5 and 7 after MIA injection. CONCLUSION Our data indicated that acute joint pain occurs along with the onset of acute inflammatory process. Irreversible structural changes in the IFP, such as extensive fibrosis, are observed prior to persistent pain development. Thus, we consider that this process may play important roles in persistent pain development.
Collapse
Affiliation(s)
- Kei Inomata
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Hiroaki Onuma
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takashi Hoshino
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mio Udo
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masako Akiyama
- Research Administration Unit, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Nakagawa
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hiroki Katagiri
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazumasa Miyatake
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Muneta
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.,National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Hideyuki Koga
- Department of Joint Surgery and Sports Medicine, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
29
|
Fang J, Du J, Fang J, Xiao T, Le X, Pan N, Yu J, Liu B. Parameter-specific analgesic effects of electroacupuncture mediated by degree of regulation TRPV1 and P2X3 in inflammatory pain in rats. Life Sci 2018; 200:69-80. [DOI: 10.1016/j.lfs.2018.03.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/11/2017] [Accepted: 03/07/2018] [Indexed: 12/21/2022]
|
30
|
Molecular Understanding of the Activation of CB1 and Blockade of TRPV1 Receptors: Implications for Novel Treatment Strategies in Osteoarthritis. Int J Mol Sci 2018; 19:ijms19020342. [PMID: 29364174 PMCID: PMC5855564 DOI: 10.3390/ijms19020342] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/18/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is a joint disease in which cartilage degenerates as a result of mechanical and biochemical changes. The main OA symptom is chronic pain involving both peripheral and central mechanisms of nociceptive processing. Our previous studies have implicated the benefits of dual- over single-acting compounds interacting with the endocannabinoid system (ECS) in OA treatment. In the present study, we focused on the specific molecular alterations associated with pharmacological treatment. OA was induced in Wistar rats by intra-articular injection of 3 mg of monoiodoacetate (MIA). Single target compounds (URB597, an FAAH inhibitor, and SB366791, a TRPV1 antagonist) and a dual-acting compound OMDM198 (FAAH inhibitor/TRPV1 antagonist) were used in the present study. At day 21 post-MIA injection, rats were sacrificed 1 h after i.p. treatment, and changes in mRNA expression were evaluated in the lumbar spinal cord by RT-qPCR. Following MIA administration, we observed 2-4-fold increase in mRNA expression of targeted receptors (Cnr1, Cnr2, and Trpv1), endocannabinoid degradation enzymes (Faah, Ptgs2, and Alox12), and TRPV1 sensitizing kinases (Mapk3, Mapk14, Prkcg, and Prkaca). OMDM198 treatment reversed some of the MIA effects on the spinal cord towards intact levels (Alox12, Mapk14, and Prkcg). Apparent regulation of ECS and TRPV1 in response to pharmacological intervention is a strong justification for novel ECS-based multi-target drug treatment in OA.
Collapse
|
31
|
Bao D, Zhao W, Dai C, Wan H, Cao Y. H89 dihydrochloride hydrate and calphostin C lower the body temperature through TRPV1. Mol Med Rep 2017; 17:1599-1608. [PMID: 29257197 PMCID: PMC5780100 DOI: 10.3892/mmr.2017.8078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/31/2017] [Indexed: 01/04/2023] Open
Abstract
The transient receptor potential vanilloid (TRPV1) serves as a negative regulator of body temperature, and during fever conditions its expression can lead to a decrease in temperature. TRPV1 is regulated by a variety of enzymes; however, it is currently unclear whether the regulation of TRPV1 phosphorylation may serve a role in the increase in TRPV1 expression during fever. In the present study, using an in vivo experimental method, rat brain ventricles were injected with the protein kinase A (PKA) antagonist, H89, and the protein kinase C (PKC) antagonist, calphostin C, and fever was induced using lipopolysaccharide (LPS) in order to detect the expression of TRPV1 and phosphorylated (p-)TRPV1, the intracellular Ca2+ concentration [(Ca2+)i] of hypothalami and rat body temperature. The results demonstrated that following the generation of fever using LPS, the expressions of TRPV1 and p-TRPV1, and hypothalamic [Ca2+]i markedly increased. In addition, following an injection with the PKA or PKC antagonist, the temperature increased further due to the inhibition of p-TRPV1. Thus, it was hypothesized that PKA and PKC may be involved in TRPV1 phosphorylation, resulting in a temperature reduction during LPS-induced fever conditions.
Collapse
Affiliation(s)
- Dongyan Bao
- Department of Physiology, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Wenqing Zhao
- Department of Physiology, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Congcong Dai
- Department of Physiology, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Hongmei Wan
- Department of Physiology, China Medical University, Shenyang, Liaoning 110000, P.R. China
| | - Yu Cao
- Department of Physiology, China Medical University, Shenyang, Liaoning 110000, P.R. China
| |
Collapse
|
32
|
Hang LH, Li SN, Dan X, Shu WW, Luo H, Shao DH. Involvement of Spinal CCR5/PKCγ Signaling Pathway in the Maintenance of Cancer-Induced Bone Pain. Neurochem Res 2016; 42:563-571. [PMID: 27848062 DOI: 10.1007/s11064-016-2108-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/21/2016] [Accepted: 11/10/2016] [Indexed: 01/24/2023]
Abstract
Cancer-induced bone pain (CIBP) is a challenging medical problem that considerably influences cancer patients' quality of life. Currently, few treatments have been developed to conquer CIBP because of a poor understanding of the potential mechanisms. Our previous work has proved that spinal RANTES (a major ligand for CCR5) was involved in the maintenance of CIBP. In this study, we attempted to investigate whether spinal CCR5 and its downstream PKCγ pathway is involved in the maintenance of CIBP. Inoculation of Walker 256 cells into the tibia could induce a marked mechanical allodynia with concomitant upregulation of spinal CCR5 and p-PKCγ expression from day 6 to day 15 after inoculation. Spinal CCR5 was prominently expressed in microglia, and mechanical allodynia was attenuated by intrathecal injection of DAPTA (a specific antagonist of CCR5) with downregulation of spinal CCR5 and p-PKCγ expression levels at day 15 in inoculated rats. Pre-intrathecal injection of RANTES could reverse the anti-allodynia effects of DAPTA. Intrathecal administration of GF109203X (an inhibitor of PKC) could alleviate mechanical allodynia as well as decrease of spinal p-PKCγ expression level, but no influence on spinal CCR5 level. Our findings suggest that CCR5/PKCγ signaling pathway in microglia may contribute to the maintenance of CIBP in rats.
Collapse
Affiliation(s)
- Li-Hua Hang
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China.
| | - Shu-Na Li
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China
| | - Xiang Dan
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China
| | - Wei-Wei Shu
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China
| | - Hong Luo
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China
| | - Dong-Hua Shao
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, People's Republic of China
| |
Collapse
|
33
|
Ghosh D, Syed AU, Prada MP, Nystoriak MA, Santana LF, Nieves-Cintrón M, Navedo MF. Calcium Channels in Vascular Smooth Muscle. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:49-87. [PMID: 28212803 DOI: 10.1016/bs.apha.2016.08.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Calcium (Ca2+) plays a central role in excitation, contraction, transcription, and proliferation of vascular smooth muscle cells (VSMs). Precise regulation of intracellular Ca2+ concentration ([Ca2+]i) is crucial for proper physiological VSM function. Studies over the last several decades have revealed that VSMs express a variety of Ca2+-permeable channels that orchestrate a dynamic, yet finely tuned regulation of [Ca2+]i. In this review, we discuss the major Ca2+-permeable channels expressed in VSM and their contribution to vascular physiology and pathology.
Collapse
Affiliation(s)
- D Ghosh
- University of California, Davis, CA, United States
| | - A U Syed
- University of California, Davis, CA, United States
| | - M P Prada
- University of California, Davis, CA, United States
| | - M A Nystoriak
- Diabetes and Obesity Center, University of Louisville, Louisville, KY, United States
| | - L F Santana
- University of California, Davis, CA, United States
| | | | - M F Navedo
- University of California, Davis, CA, United States.
| |
Collapse
|
34
|
Fattori V, Hohmann MSN, Rossaneis AC, Pinho-Ribeiro FA, Verri WA. Capsaicin: Current Understanding of Its Mechanisms and Therapy of Pain and Other Pre-Clinical and Clinical Uses. Molecules 2016; 21:E844. [PMID: 27367653 PMCID: PMC6273101 DOI: 10.3390/molecules21070844] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023] Open
Abstract
In this review, we discuss the importance of capsaicin to the current understanding of neuronal modulation of pain and explore the mechanisms of capsaicin-induced pain. We will focus on the analgesic effects of capsaicin and its clinical applicability in treating pain. Furthermore, we will draw attention to the rationale for other clinical therapeutic uses and implications of capsaicin in diseases such as obesity, diabetes, cardiovascular conditions, cancer, airway diseases, itch, gastric, and urological disorders.
Collapse
Affiliation(s)
- Victor Fattori
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Miriam S N Hohmann
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Ana C Rossaneis
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rodovia Celso Garcia Cid KM480 PR445, Caixa Postal 10.011, 86057-970 Londrina, Paraná, Brazil.
| |
Collapse
|