1
|
Akhilesh, Singh AK, Tiwari V. TRPA1 siRNA-Loaded Nanoformulation Ameliorates Chemotherapy-Induced Peripheral Neuropathy. ACS Chem Neurosci 2025. [PMID: 40421769 DOI: 10.1021/acschemneuro.5c00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
Small interfering RNA (siRNA) has emerged as a cutting-edge therapeutic strategy, with significant promise for addressing peripheral neuropathies. Despite its immense revolutionary therapeutic potential, the application and sustained release of siRNA for the treatment of chronic pain remain an arduous scientific challenge. This study introduces a novel cationic lipid-based siRNA formulation specifically targeting transient receptor potential ankyrin 1 (TRPA1) for the systemic treatment of chemotherapy-induced neuropathic pain (CINP), a condition with no US-FDA-approved therapeutic options. CINP involves the upregulation of the TRPA1 channel, a key player in nociceptive signaling. Our approach leverages the selective silencing of the TRPA1 gene via siRNA encapsulated in liposomes, offering a targeted and safer therapeutic intervention. The proof-of-principle was established through in vivo experiments, demonstrating significant downregulation of TRPA1 mRNA and protein expressions in the spinal cord following intrathecal administration. Liposomal encapsulation improved siRNA stability and delivery, validated through sophisticated morphometric and analytical techniques. Behavioral assays revealed that both intravenous and intrathecal administrations of this TRPA1 siRNA formulation significantly reduced mechanical and cold hypersensitivity in CINP models. The sustained release profile of siRNA from liposomes ensured prolonged efficacy, contrasting sharply with the transient effects of nonencapsulated siRNA. Mechanistically, silencing of the TRPA1 gene led to decreased microglial activation and reduced expression of inflammatory markers such as ICAM-1 and iba1, mitigating neuroinflammatory responses in the dorsal root ganglia and spinal cord. Intravenous delivery notably outperformed intrathecal administration in downregulating TRPA1 and IL-6 expressions. Overall findings highlight the potential of this nanoengineered TRPA1 siRNA formulation to effectively modulate critical inflammatory pathways and manage CINP. This innovative and exciting strategy not only overcomes the limitations of conventional therapies but also paves the way for new approaches in chronic pain management with significant implications for future clinical applications.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| | - Anurag Kumar Singh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, UP India, 221005
| |
Collapse
|
2
|
Ten Barge JA, van den Bosch GE, Slater R, van den Hoogen NJ, Reiss IKM, Simons SHP. Visceral Pain in Preterm Infants with Necrotizing Enterocolitis: Underlying Mechanisms and Implications for Treatment. Paediatr Drugs 2025; 27:201-220. [PMID: 39752054 PMCID: PMC11829917 DOI: 10.1007/s40272-024-00676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
Necrotizing enterocolitis (NEC) is a relatively rare but very severe gastrointestinal disease primarily affecting very preterm infants. NEC is characterized by excessive inflammation and ischemia in the intestines, and is associated with prolonged, severe visceral pain. Despite its recognition as a highly painful disease, current pain management for NEC is often inadequate, and research on optimal analgesic therapy for these patients is lacking. Insight into the mechanisms underlying intestinal pain in infants with NEC-visceral pain-could help identify the most effective analgesics for these vulnerable patients. Therefore, this comprehensive review aims to provide an overview of visceral nociception, including transduction, transmission, modulation, and experience, and discuss the implications for analgesic therapy in preterm infants with NEC. The transmission of visceral pain differs from that of somatic pain, contributing to the diffuse nature of visceral pain. Studies evaluating the effectiveness of analgesics for treating visceral pain in infants are scarce. However, research in visceral pain models highlights agents that may be particularly effective for treating visceral pain based on their mechanisms of action. Further research is necessary to determine whether agents that have shown promise for treating visceral pain in preclinical studies and adults are effective in infants with NEC as well.
Collapse
Affiliation(s)
- Judith A Ten Barge
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Gerbrich E van den Bosch
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | - Irwin K M Reiss
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Sinno H P Simons
- Department of Neonatal and Pediatric Intensive Care, Division of Neonatology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Kim Y, Je MA, Jeong M, Kwon H, Jang A, Kim J, Choi GE. Upregulation of NGF/TrkA-Related Proteins in Dorsal Root Ganglion of Paclitaxel-Induced Peripheral Neuropathy Animal Model. J Pain Res 2024; 17:3919-3932. [PMID: 39588524 PMCID: PMC11586490 DOI: 10.2147/jpr.s470671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024] Open
Abstract
Background Paclitaxel (PTX) can induce chemotherapy-induced peripheral neuropathy (CIPN) as a side effect. The aim of this study was to understand the neurochemical changes induced by NGF/TrkA signaling in PTX-induced neuropathic pain. Methods The PTX-induced CIPN mouse model was evaluated using nerve conduction velocity (NCV) and behavioral tests. Protein expression in mouse DRG was observed by Western blotting and immunohistochemistry. Nerve growth factor (NGF), IL-6, and IL-1β mRNA levels were determined using qRT-PCR by isolating total RNA from whole blood. Results PTX showed low amplitude and high latency values in NCV in mice, and induced cold allodynia and thermal hyperalgesia in behavioral assessment. Activating transcription factor 3 (ATF3) and MAPK pathway related proteins (ERK1/2), tropomyosin receptor kinase A (TrkA), calcitonin gene related peptide (CGRP) and transient receptor potential vanilloid 1 (TRPV1) were upregulated 7th and 14th days after 2 mg/kg and 10 mg/kg of PTX administration. Protein kinase C (PKC) was upregulated 7th days after 10 mg/kg PTX treatment and 14th days after 2 mg/kg and 10 mg/kg PTX administration. NGF, IL-6, and IL-1β fold change values also showed a time- and dose-dependent increase. Conclusion Taken together, our findings may improve our understanding of the nociceptive symptoms associated with PTX-induced neuropathic pain and lead to the development of new treatments for peripheral neuropathy.
Collapse
Affiliation(s)
- Yeeun Kim
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Min-A Je
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Myeongguk Jeong
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Hyeokjin Kwon
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Aelee Jang
- Department of Nursing, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jungho Kim
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| | - Go-Eun Choi
- Department of Clinical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan, 46252, Republic of Korea
- Next-Generation Industrial Field-Based Specialist Program for Molecular Diagnostics, Brain Busan 21 Plus Project, Graduate School, Catholic University of Pusan, Busan, 46252, Republic of Korea
| |
Collapse
|
4
|
Saha P, Sharma SS. RNA Interference Unleashed: Current Perspective of Small Interfering RNA (siRNA) Therapeutics in the Treatment of Neuropathic Pain. ACS Pharmacol Transl Sci 2024; 7:2951-2970. [PMID: 39416962 PMCID: PMC11475279 DOI: 10.1021/acsptsci.4c00329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/12/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024]
Abstract
Neuropathic pain (NP) is one of the debilitating pain phenotypes that leads to the progressive degeneration of the central as well as peripheral nervous system. NP is often associated with hyperalgesia, allodynia, paresthesia, tingling, and burning sensations leading to disability, motor dysfunction, and compromised psychological state of the patients. Most of the conventional pharmacological agents are unable to improve the devastating conditions of pain because of their limited efficacy, undesirable side effects, and multifaceted pathophysiology of the diseased condition. A rapid rise in new cases of NP warrants further research for identifying the potential novel therapeutic modalities for treating NP. Recently, small interfering RNA (siRNA) approach has shown therapeutic potential in many disease conditions including NP. Delivery of siRNAs led to potential and selective downregulation of target mRNA and abolished the pain-related behaviors/pathophysiological pain response. The crucial role of siRNA in the treatment of NP by considering all of the pathways associated with NP that could be managed by siRNA therapeutics has been discussed. However, their therapeutic use is limited by several hurdles such as instability in systemic circulation due to their negative charge and membrane impermeability, off-target effects, immunogenicity, and inability to reach the intended site of action. This review also emphasizes several strategies and techniques to overcome these hurdles for translating these therapeutic siRNAs from bench to bedside by opening a new avenue for obtaining a potential therapeutic approach for treating NP.
Collapse
Affiliation(s)
- Priya Saha
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Shyam S. Sharma
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab 160062, India
| |
Collapse
|
5
|
Jiang Y, Shen L, Wang B. Non-electrophysiological techniques targeting transient receptor potential (TRP) gene of gastrointestinal tract. Int J Biol Macromol 2024; 262:129551. [PMID: 38367416 DOI: 10.1016/j.ijbiomac.2024.129551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/19/2024]
Abstract
Transient receptor potential (TRP) channels are cation channels related to a wide range of physical and chemical stimuli, they are expressed all along the gastrointestinal system, and a myriad of diseases are often associated with aberrant expression or mutation of the TRP gene, suggesting that TRPs are promising targets for drug therapy. Therefore, a better understanding of the information of TRPs in health and disease could facilitate the development of effective drugs for the treatment of gastrointestinal diseases like IBD. But there are very few generalizations about the experimental techniques studied in this field. In view of the promise of TRP as a therapeutic target, we discuss experimental methods that can be used for TRPs including their distribution, function and interaction with other proteins, as well as some promising emerging technologies to provide experimental methods for future studies.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China.
| |
Collapse
|
6
|
Ovsepian SV, Waxman SG. Gene therapy for chronic pain: emerging opportunities in target-rich peripheral nociceptors. Nat Rev Neurosci 2023; 24:252-265. [PMID: 36658346 DOI: 10.1038/s41583-022-00673-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
With sweeping advances in precision delivery systems and manipulation of the genomes and transcriptomes of various cell types, medical biotechnology offers unprecedented selectivity for and control of a wide variety of biological processes, forging new opportunities for therapeutic interventions. This perspective summarizes state-of-the-art gene therapies enabled by recent innovations, with an emphasis on the expanding universe of molecular targets that govern the activity and function of primary sensory neurons and which might be exploited to effectively treat chronic pain.
Collapse
Affiliation(s)
- Saak V Ovsepian
- School of Science, Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, UK.
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Silva Santos Ribeiro P, Willemen HLDM, Eijkelkamp N. Mitochondria and sensory processing in inflammatory and neuropathic pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1013577. [PMID: 36324872 PMCID: PMC9619239 DOI: 10.3389/fpain.2022.1013577] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023] Open
Abstract
Rheumatic diseases, such as osteoarthritis and rheumatoid arthritis, affect over 750 million people worldwide and contribute to approximately 40% of chronic pain cases. Inflammation and tissue damage contribute to pain in rheumatic diseases, but pain often persists even when inflammation/damage is resolved. Mechanisms that cause this persistent pain are still unclear. Mitochondria are essential for a myriad of cellular processes and regulate neuronal functions. Mitochondrial dysfunction has been implicated in multiple neurological disorders, but its role in sensory processing and pain in rheumatic diseases is relatively unexplored. This review provides a comprehensive understanding of how mitochondrial dysfunction connects inflammation and damage-associated pathways to neuronal sensitization and persistent pain. To provide an overall framework on how mitochondria control pain, we explored recent evidence in inflammatory and neuropathic pain conditions. Mitochondria have intrinsic quality control mechanisms to prevent functional deficits and cellular damage. We will discuss the link between neuronal activity, mitochondrial dysfunction and chronic pain. Lastly, pharmacological strategies aimed at reestablishing mitochondrial functions or boosting mitochondrial dynamics as therapeutic interventions for chronic pain are discussed. The evidence presented in this review shows that mitochondria dysfunction may play a role in rheumatic pain. The dysfunction is not restricted to neuronal cells in the peripheral and central nervous system, but also includes blood cells and cells at the joint level that may affect pain pathways indirectly. Pre-clinical and clinical data suggest that modulation of mitochondrial functions can be used to attenuate or eliminate pain, which could be beneficial for multiple rheumatic diseases.
Collapse
Affiliation(s)
| | | | - Niels Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
8
|
Zarei MM, Abdolmaleki Z, Shahidi S. Bioflavonoid exerts analgesic and anti-inflammatory effects via transient receptor potential 1 channel in a rat model. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:900-907. [PMID: 36351417 PMCID: PMC9770082 DOI: 10.1055/s-0042-1755321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/26/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND Pain is an uncomfortable sensation in the body. Kaempferol is a flavonoid with antinociceptive effects. Transient receptor potential (TRP) channels have been characterized in the sensory system. OBJECTIVE This study evaluated the central antinociceptive effect of Kaempferol and possible mechanisms of action of transient receptor potential cation channel subfamily V member 1 (TRPV1). METHODS Capsaicin as a TRPV agonist (5 μg/μL, intracerebroventricular [ICV]) and capsazepine as its antagonist (10 μg/μL, icv) were used to test the analgesic effect of kaempferol (1.5 mg, ICV). Morphine (10 μg, ICV) was used as a positive control. The other groups were treated with a combination of kaempferol and capsaicin, kaempferol and capsazepine, and capsaicin and capsazepine. The cannula was implanted in the cerebroventricular area. The tail-flick, acetic acid, and formalin tests were used to assess analgesic activity. For evaluation of antiinflammatory effect, the formalin-induced rat paw edema was used. RESULTS Kaempferol significantly decreased pain in the acute pain models, including the tail-flick and the first phase of the formalin test. In the late phase of the formalin test, as a valid model of nociception, capsazepine inhibited the antinociceptive effect of kaempferol. CONCLUSIONS Kaempferol has an analgesic effect in the acute pain model and can affect inflammatory pain. Also, the TRPV1 channel plays a role in the antinociceptive activity of kaempferol.
Collapse
Affiliation(s)
| | | | - Siamak Shahidi
- University of Medical Sciences, School of Medicine, Department of Physiology, Hamadan, Iran.
- Hamadan University of Medical Sciences, Neurophysiology Research Center, Hamadan, Iran.
| |
Collapse
|
9
|
Pasdaran A, Butovska D, Kerr P, Naychov Z, Aneva I, Kozuharova E. Gentians, natural remedies for future of visceral pain control; an ethnopharmacological review with an in silico approach. Biol Futur 2022; 73:219-227. [PMID: 35318616 DOI: 10.1007/s42977-022-00114-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 02/17/2022] [Indexed: 10/18/2022]
Abstract
Different gentian preparations are used as traditional remedies for internal pain control in: Persian traditional medicine (PTM), Chinese traditional medicine (CTM) and Ancient Greek medicine (AGM) from the time of the Roman Empire. Objective: To present a survey of the ethnopharmacological applications of gentians recorded as being used in Eastern and Western traditional medical systems (PTM, CTM and AGM) and their pharmacological effects, chemical composition as well as an in silico investigation of the possible active component/s for the alleviation of internal pain via molecular docking studies. Major traditional medicine literature (PTM, CTM and AGM, 50 AD- 1770) and ethnobotanical studies for the application of gentians were reviewed. Nine European species representing 5 of the 13 sections currently attributed to Gentiana were selected. Chemical compounds and pharmacological activity data of these species were gathered from different databases including Google Scholar, PubChem, PubMed and Web of Science (between 1972 and 2020). The possible active constituents of gentians on visceral pain receptors were investigated, in silico. In all investigated literature, traditional uses of gentian were indicated to have anti-nociceptive effects on visceral pain and possess diuretic action. According to our computational study, acylated flavonoid glycosides, viz. trans-feruloyl-2"-isovitexin (33), trans-feruloyl-2"-isovitexin-4'-O-β-D-glucoside (34), iso-orientin-4'-O-glucoside (38), trans-caffeoyl-2"-iso-orientin-4'-O-β-D-glucoside (39), iso-orientin-2"-O-β-D-glucoside (40) and isoscoparin (41), might be responsible for visceral pain reduction by interacting with the purinergic receptor (P2X3) and vanilloid receptor 1 (TrpV1). This finding shows a good correlation with different traditional gentian uses in Persian, Chinese and European ethnomedicine for visceral pain control.
Collapse
Affiliation(s)
- Ardalan Pasdaran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | | - Philip Kerr
- School of Biomedical Sciences & Graham Centre, Charles Sturt University, Boorooma St, Locked Bag 588, Wagga Wagga, NSW, 2678, Australia
| | | | - Ina Aneva
- Institute of Biodiversity and Ecosystem Research, Bulgarian Academy of Sciences, G. Bončev Str, Sofia, Bulgaria
| | - Ekaterina Kozuharova
- Faculty of Pharmacy, Department of Pharmacognosy, Medical University of Sofia, Sofia, 1000, Bulgaria.
| |
Collapse
|
10
|
Wang J, Zhou F, Zhang S, Mao M, Feng S, Wang X. Participation of transient receptor potential vanilloid 1 in the analgesic effect of duloxetine for paclitaxel induced peripheral neuropathic pain. Neurosci Lett 2022; 773:136512. [PMID: 35149198 DOI: 10.1016/j.neulet.2022.136512] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
Abstract
Painful peripheral neuropathy is a common dose-limiting side effect of chemotherapeutic paclitaxel (PTX) treatment. The American Society of Clinical Oncology (ASCO) recommends duloxetine (DUL) as a promising treatment alternative for chemotherapy-induced peripheral neuropathic pain. However, this recommendation lacks a robust theoretical basis and supporting data. To elucidate the involvement of transient receptor potential vanilloid 1 (TRPV1) in the analgesic effect of DUL for PTX-induced neuropathic pain, TRPV1 expression in the lumbar dorsal root ganglion (DRG) and spinal cord was evaluated following intraperitoneal administration of PTX (2 mg/kg/day) for four alternate days in rats. Western blot and immunohistochemistry suggested that a cumulative dosage of PTX (8 mg/kg) upregulated TRPV1 expression in the lumbar DRG and spinal dorsal horn (SDH) at day 14 post treatment. TRPV1 upregulation in the DRG was mainly expressed in calcitonin gene-related peptide (CGRP) and IB-4 positive small-size sensory neurons. Additionally, PTX increased CGRP and substance P (SP) expression in the DRG and SDH, induced SDH microglia and astrocyte activation, and upregulated spinal tumor necrosis factor-α (TNF-α) but not IL-1β or IL-10 expression. Behavioral detection showed that PTX-related mechanical and thermal hyperalgesia was significantly inhibited by consecutive administration of DUL 20 mg/kg/day greater than 10 mg/kg/day for 5 days starting at day 10 post PTX injection. Furthermore, DUL (20 mg/kg/day) for 5 days markedly ameliorated PTX-induced TRPV1, CGRP, and SP upregulation in the DRG and SDH, and mitigated PTX-induced spinal cord glia activation and TNF-α expression. Moreover, the pharmacological blockade of TRPV1 resulted in an analgesic effect on PTX-induced hyperalgesia. Collectively, these results suggest that DUL alleviates PTX-induced peripheral neuropathic pain by suppressing TRPV1 upregulation in the lumbar DRG and SDH, which is followed by a reduction in CGRP and SP release, as well as spinal glia activation and TNF-α expression.
Collapse
Affiliation(s)
- Jing Wang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Feng Zhou
- Department of Anesthesiology, Jinan Maternity and Child Health Care Hospital, Jinan, China
| | - Susu Zhang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Mao Mao
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Shanwu Feng
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Xian Wang
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| |
Collapse
|
11
|
Akhilesh, Uniyal A, Gadepalli A, Tiwari V, Allani M, Chouhan D, Ummadisetty O, Verma N, Tiwari V. Unlocking the potential of TRPV1 based siRNA therapeutics for the treatment of chemotherapy-induced neuropathic pain. Life Sci 2022; 288:120187. [PMID: 34856209 DOI: 10.1016/j.lfs.2021.120187] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 01/23/2023]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is among the most common clinical complications associated with the use of anti-cancer drugs. CINP occurs in nearly 68.1% of the cancer patients receiving chemotherapeutic drugs. Most of the clinically available analgesics are ineffective in the case of CINP patients as the pathological mechanisms involved with different chemotherapeutic drugs are distinct from each other. CINP triggers the somatosensory nervous system, increases the neuronal firing and activation of nociceptive mediators including transient receptor protein vanilloid 1 (TRPV1). TRPV1 is widely present in the peripheral nociceptive nerve cells and it has been reported that the higher expression of TRPV1 in DRGs serves a critical role in the potentiation of CINP. The therapeutic glory of TRPV1 is well recognized in clinics which gives a promising insight into the treatment of pain. But the adverse effects associated with some of the antagonists directed the scientists towards RNA interference (RNAi), a tool to silence gene expression. Thus, ongoing research is focused on developing small interfering RNA (siRNA)-based therapeutics targeting TRPV1. In this review, we have discussed the involvement of TRPV1 in the nociceptive signaling associated with CINP and targeting this nociceptor, using siRNA will potentially arm us with effective therapeutic interventions for the clinical management of CINP.
Collapse
Affiliation(s)
- Akhilesh
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ankit Uniyal
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anagha Gadepalli
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vineeta Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Meghana Allani
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Deepak Chouhan
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Obulapathi Ummadisetty
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Nimisha Verma
- Department of Anaesthesiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
12
|
Tamada M, Ohi Y, Kodama D, Miyazawa K, Goto S, Haji A. Modulation of excitatory synaptic transmissions by TRPV1 in the spinal trigeminal subnucleus caudalis neurons of neuropathic pain rats. Eur J Pharmacol 2021; 913:174625. [PMID: 34758353 DOI: 10.1016/j.ejphar.2021.174625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 10/29/2021] [Accepted: 11/04/2021] [Indexed: 01/16/2023]
Abstract
The present study examined contribution of the transient receptor potential vanilloid 1 channel (TRPV1) to the chronic orofacial pain. Bilateral partial nerve ligation (PNL) of the mental nerve, a branch of trigeminal nerve, was performed to induce neuropathic pain. The withdrawal threshold in response to mechanical stimulation of the lower lip skin was substantially reduced after the surgery in the PNL rats while it remained unchanged in the sham rats. This reduction in the PNL rats was alleviated by pregabalin injected intraperitoneally (10 mg/kg) and intracisternally (10, 30, 100 μg). Furthermore, an intracisternal injection of AMG9810, an antagonist of TRPV1, (1.5, 5.0 μg) attenuated the reduction of withdrawal threshold. Spontaneous and miniature excitatory postsynaptic currents (sEPSCs and mEPSCs) were recorded from the spinal trigeminal subnucleus caudalis (Vc) neurons in the brainstem slice, which receive the orofacial nociceptive signals. In the PNL rats, superfusion of capsaicin (0.03, 0.1 μM) enhanced their frequency without effect on the amplitude and the highest concentration (0.3 μM) increased both the frequency and amplitude. In the sham rats, only 0.3 μM capsaicin increased their frequency. Thus, capsaicin-induced facilitation of sEPSCs and mEPSCs in the PNL rats was significantly stronger than that in the sham rats. AMG9810 (0.1 μM) attenuated the capsaicin's effect. Capsaicin was ineffective on the trigeminal tract-evoked EPSCs in the PNL and sham rats. These results suggest that the chronic orofacial pain in the PNL model results from facilitation of the spontaneous excitatory synaptic transmission in the Vc region through TRPV1 at least partly.
Collapse
Affiliation(s)
- Mayo Tamada
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan; Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Yoshiaki Ohi
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Daisuke Kodama
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Ken Miyazawa
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Shigemi Goto
- Department of Orthodontics, School of Dentistry, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| | - Akira Haji
- Laboratory of Neuropharmacology, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya, 464-8650, Japan.
| |
Collapse
|
13
|
Luo J, Chen J, Yang C, Tan J, Zhao J, Jiang N, Zhao Y. 6-Gingerol protects against cerebral ischemia/reperfusion injury by inhibiting NLRP3 inflammasome and apoptosis via TRPV1 / FAF1 complex dissociation-mediated autophagy. Int Immunopharmacol 2021; 100:108146. [PMID: 34537481 DOI: 10.1016/j.intimp.2021.108146] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/22/2021] [Accepted: 09/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our previous studies demonstrated that autophagy alleviates cerebral I/R injury by inhibiting NLRP3 inflammasome-mediated inflammation. 6-Gingerol, a phenolic compound extracted from ginger, was reported to possess potent antiapoptotic and anti-inflammatory activities and is associated with autophagy. However, the effects of 6-Gingerol in cerebral I/R injury have not been elucidated, and whether they involve autophagy-induced NLRP3 inflammasome inhibition remains unclear. METHODS Adult male Sprague-Dawley (SD) rats were subjected to middle cerebral artery occlusion (MCAO) for 1 h, followed by reperfusion for 24 h. 6-Gingerol and 3-methyladenine (3-MA) were injected intraperitoneally, and si-TRPV1 was injected via the lateral ventricle. Cerebral infarct volume, brain edema, neurological deficits, HE and Nissl were used to evaluate the morphological and functional changes of brain tissue, respectively. TRPV1, FAF1, autophagy related (LC3II/I, P62, Beclin1), inflammation related (NLRP3, cleaved-caspase-1, caspase-1, cleaved-IL-1β, IL-1β, cleaved-IL-18, IL-18) and apoptosis related (Bcl-2, Bax, cleaved-caspase-3) proteins were assessed by Western blot, immunofluorescence staining and coimmunoprecipitation, respectively. Enzyme linked immunosorbent assay (ELISA) was used to evaluate the changes in the expression levels of interleukin-1 (IL-1β) and interleukin-18(IL-18), respectively. The degree of neuronal apoptosis was evaluated by TUNEL staining. Neuronal ultrastructure was examined by transmission electron microscopy. RESULT 6-Gingerol treatment significantly reduced cerebral infarct volume, improved brain edema and neurological scores, and reversed brain histomorphological damage after I/R injury. In addition, 6-Gingerol significantly reduced NLRP3 inflammasome-derived inflammation and neuronal apoptosis and upregulated autophagy. The autophagy inhibitor 3-MA rescued the effects of 6-Gingerol on the NLRP3 inflammasome and apoptosis. Moreover, the findings illustrated that 6-Gingerol inhibited autophagy-induced NLRP3 inflammasome activation and apoptosis through the dissociation of TRPV1 from FAF1. CONCLUSION In brief, 6-Gingerol exerts antiapoptotic and anti-inflammatory effects via TRPV1/FAF1 complex dissociation-mediated autophagy during cerebral I/R injury. Therefore, 6-Gingerol may be an effective drug for the treatment of I/R injury.
Collapse
Affiliation(s)
- Jing Luo
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- Department of First Clinical College, Chongqing Medical University, Chongqing 400016, China
| | - Changhong Yang
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, China
| | - Junyi Tan
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China
| | - Ning Jiang
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
14
|
Wanasuntronwong A, Punyawattananon V, Rotpenpian N, Meepong R, Srikiatkhachorn A. Nociceptive receptors are expressed differently in trigeminal nociception after lingual nerve injury and unilateral external carotid artery occlusion in rats. Arch Oral Biol 2021; 126:105128. [PMID: 33895544 DOI: 10.1016/j.archoralbio.2021.105128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To investigate the different changes in nociceptive activity between two animal models of trigeminal neuropathic pain: unilateral external carotid artery ischemic reperfusion and lingual nerve crush in rats. DESIGN In this study, changes in nociceptive activity were investigated in unilateral external carotid artery ischemic reperfusion and lingual nerve crush models of trigeminal neuropathic pain in rats. Field excitatory postsynaptic potentials (fEPSPs) evoked by capsaicin application on the tongue of rats were recorded in the trigeminal nucleus caudalis. In addition, immunohistochemistry was performed in the trigeminal ganglia and trigeminal nucleus caudalis. RESULTS The fEPSP in unilateral external carotid artery ischemic reperfusion and lingual nerve crush rats was irregular relative to that in sham rats. In particular, the fEPSP spike in lingual nerve crush rats had a higher amplitude and shorter duration than that in sham rats. Unilateral external carotid artery ischemic reperfusion and lingual nerve crush also increased c-fos expression in the trigeminal nucleus caudalis. Upregulation of transient receptor potential vanilloid 1 in trigeminal ganglion was observed in unilateral external carotid artery ischemic reperfusion and lingual nerve crush rats, whereas upregulation of purinergic receptor subtype 3 in trigeminal ganglion was observed only in lingual nerve crush rats. CONCLUSIONS Although unilateral external carotid artery ischemic reperfusion and lingual nerve crush similarly increased nociceptive activity at the trigeminal nucleus caudalis, the fEPSPs and expression of nociceptive peripheral afferent neurons were different. Therefore, direct and indirect nerve injuries apparently induced the same nociceptive activity by different signaling responses dependent on nociceptive receptors.
Collapse
Affiliation(s)
| | | | | | | | - Anan Srikiatkhachorn
- Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| |
Collapse
|
15
|
Paclitaxel Induces Upregulation of Transient Receptor Potential Vanilloid 1 Expression in the Rat Spinal Cord. Int J Mol Sci 2020; 21:ijms21124341. [PMID: 32570786 PMCID: PMC7352737 DOI: 10.3390/ijms21124341] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Painful peripheral neuropathy is a common adverse effect of paclitaxel (PTX) treatment. To analyze the contribution of transient receptor potential vanilloid 1 (TRPV1) in the development of PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia, TRPV1 expression in the rat spinal cord was analyzed after intraperitoneal administration of 2 and 4 mg/kg PTX. PTX treatment increased the expression of TRPV1 protein in the spinal cord. Immunohistochemistry showed that PTX (4 mg/kg) treatment increased TRPV1 protein expression in the superficial layers of the spinal dorsal horn 14 days after treatment. Behavioral assessment using the paw withdrawal response showed that PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia after 14 days was significantly inhibited by oral or intrathecal administration of the TRPV1 antagonist AMG9810. We found that intrathecal administration of small interfering RNA (siRNA) to knock down TRPV1 protein expression in the spinal cord significantly decreased PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia. Together, these results demonstrate that TRPV1 receptor expression in spinal cord contributes, at least in part, to the development of PTX-induced painful peripheral neuropathy. TRPV1 receptor antagonists may be useful in the prevention and treatment of PTX-induced peripheral neuropathic pain.
Collapse
|
16
|
Valdor M, Wagner A, Fischer H, Röhrs V, Schröder W, Bahrenberg G, Welbers A, Fechner H, Kurreck J, Tzschentke TM, Christoph T. RNA interference-mediated silencing of Kv7.2 in rat dorsal root ganglion neurons abolishes the anti-nociceptive effect of a selective channel opener. J Pharmacol Toxicol Methods 2020; 103:106693. [DOI: 10.1016/j.vascn.2020.106693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/25/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023]
|
17
|
Pain Relief and Kaempferol: Activation of Transient Receptors Potential Vanilloid Type 1 in Male Rats. PAJOUHAN SCIENTIFIC JOURNAL 2020. [DOI: 10.52547/psj.18.2.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
18
|
Schiene K, Schröder W, Linz K, Frosch S, Tzschentke TM, Christoph T, Xie JY, Porreca F. Inhibition of experimental visceral pain in rodents by cebranopadol. Behav Pharmacol 2019; 30:320-326. [DOI: 10.1097/fbp.0000000000000420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
19
|
de la Peña JBI, Song JJ, Campbell ZT. RNA control in pain: Blame it on the messenger. WILEY INTERDISCIPLINARY REVIEWS-RNA 2019; 10:e1546. [PMID: 31090211 DOI: 10.1002/wrna.1546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/12/2022]
Abstract
mRNA function is meticulously controlled. We provide an overview of the integral role that posttranscriptional controls play in the perception of painful stimuli by sensory neurons. These specialized cells, termed nociceptors, precisely regulate mRNA polarity, translation, and stability. A growing body of evidence has revealed that targeted disruption of mRNAs and RNA-binding proteins robustly diminishes pain-associated behaviors. We propose that the use of multiple independent regulatory paradigms facilitates robust temporal and spatial precision of protein expression in response to a range of pain-promoting stimuli. This article is categorized under: RNA in Disease and Development > RNA in Disease Translation > Translation Regulation RNA Turnover and Surveillance > Regulation of RNA Stability.
Collapse
Affiliation(s)
- June Bryan I de la Peña
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Jane J Song
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences and the Center for Advanced Pain Studies, University of Texas, Dallas, Richardson, Texas
| |
Collapse
|
20
|
Guo SH, Lin JP, Huang LE, Yang Y, Chen CQ, Li NN, Su MY, Zhao X, Zhu SM, Yao YX. Silencing of spinal Trpv1 attenuates neuropathic pain in rats by inhibiting CAMKII expression and ERK2 phosphorylation. Sci Rep 2019; 9:2769. [PMID: 30808963 PMCID: PMC6391380 DOI: 10.1038/s41598-019-39184-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/14/2019] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence suggests a potential role of transient receptor potential vanilloid 1 (TRPV1) channels in inflammatory and cancer-related pain. However, the role of TRPV1 in the maintenance of neuropathic pain remains elusive. The current study investigated the effects of transient Trpv1 gene silencing using a small interference RNA (siRNA) on neuropathic pain induced by chronic constriction injury (CCI) of the sciatic nerve in rats. Seven days after CCI, the TRPV1 siRNA was intrathecally administered (5 µg/15 µl, once daily for 2 days). TRPV1 and Ca2+/calmodulin-dependent protein kinase II (CAMKII) expression and extracellular signal-regulated kinase (ERK) phosphorylation in the spinal cord were detected using western blotting. The thresholds to mechanical and thermal stimuli were determined before and after intrathecal TRPV1 siRNA administration. TRPV1 and CAMKII expression and ERK2 phosphorylation in the spinal cord were upregulated after CCI. Intrathecal administration of the TRPV1 siRNA not only attenuated behavioural hyperalgesia but also reduced the expression of TRPV1 and CAMKII, as well as ERK2 phosphorylation. Based on these results, silencing of the TRPV1 gene in the spinal cord attenuates the maintenance of neuropathic pain by inhibiting CAMKII/ERK2 activation and suggests that TRPV1 represents a potential target in pain therapy.
Collapse
Affiliation(s)
- Shao-Hui Guo
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Jia-Piao Lin
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Ling-Er Huang
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yan Yang
- Zhejiang University School of Medicine, Centre for Neuroscience, Hangzhou, 310016, P. R. China
| | - Chao-Qin Chen
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Na-Na Li
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Meng-Yun Su
- Department of Anaesthesia, Shulan (Hangzhou) Hospital, Hangzhou, 310022, P. R. China
| | - Xian Zhao
- Department of Anaesthesia, Shulan (Hangzhou) Hospital, Hangzhou, 310022, P. R. China
| | - Sheng-Mei Zhu
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China
| | - Yong-Xing Yao
- Department of Anaesthesia, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, P. R. China.
| |
Collapse
|
21
|
Guo ZL, Fu LW, Su HF, Tjen-A-Looi SC, Longhurst JC. Role of TRPV1 in acupuncture modulation of reflex excitatory cardiovascular responses. Am J Physiol Regul Integr Comp Physiol 2018; 314:R655-R666. [PMID: 29351423 PMCID: PMC6008114 DOI: 10.1152/ajpregu.00405.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 01/17/2023]
Abstract
We have shown that acupuncture, including manual and electroacupuncture (MA and EA), at the P5-6 acupoints stimulates afferent fibers in the median nerve (MN) to modulate sympathoexcitatory cardiovascular reflexes through central regulation of autonomic function. However, the mechanisms underlying acupuncture activation of these sensory afferent nerves and their cell bodies in the dorsal root ganglia (DRG) are unclear. Transient receptor potential vanilloid type 1 (TRPV1) is present in sensory nerve fibers distributed in the general region of acupoints like ST36 and BL 40 located in the hindlimb. However, the contribution of TRPV1 to activation of sensory nerves by acupuncture, leading to modulation of pressor responses, has not been studied. We hypothesized that TRPV1 participates in acupuncture's activation of sensory afferents and their associated cell bodies in the DRG to modulate pressor reflexes. Local injection of iodoresiniferatoxin (Iodo-RTX; a selective TRPV1 antagonist), but not 5% DMSO (vehicle), into the P6 acupoint on the forelimb reversed the MA's inhibition of pressor reflexes induced by gastric distension (GD). Conversely, inhibition of GD-induced sympathoexcitatory responses by EA at P5-6 was unchanged after administration of Iodo-RTX into P5-6. Single-unit activity of Group III or IV bimodal afferents sensitive to both mechanical and capsaicin stimuli responded to MA stimulation at P6. MA-evoked activity was attenuated significantly ( P < 0.05) by local administration of Iodo-RTX ( n = 12) but not by 5% DMSO ( n = 12) into the region of the P6 acupoint in rats. Administration of Iodo-RTX into P5-6 did not reduce bimodal afferent activity evoked by EA stimulation ( n = 8). Finally, MA at P6 and EA at P5-6 induced phosphorylation of extracellular signal-regulated kinases (ERK; an intracellular signaling messenger involved in cellular excitation) in DRG neurons located at C7-8 spinal levels receiving MN inputs. After TRPV1 was knocked down in the DRG at these spinal levels with intrathecal injection of TRPV1-siRNA, expression of phosphorylated ERK in the DRG neuron was reduced in MA-treated, but not EA-treated animals. These data suggest that TRPV1 in Group III and IV bimodal sensory afferent nerves contributes to acupuncture inhibition of reflex increases in blood pressure and specifically plays an important role during MA but not EA.
Collapse
Affiliation(s)
- Zhi-Ling Guo
- Department of Medicine and Susan-Samueli Institute for Integrative Health, School of Medicine, University of California at Irvine , Irvine, California
| | - Liang-Wu Fu
- Department of Medicine and Susan-Samueli Institute for Integrative Health, School of Medicine, University of California at Irvine , Irvine, California
| | - Hou-Fen Su
- Department of Medicine and Susan-Samueli Institute for Integrative Health, School of Medicine, University of California at Irvine , Irvine, California
| | - Stephanie C Tjen-A-Looi
- Department of Medicine and Susan-Samueli Institute for Integrative Health, School of Medicine, University of California at Irvine , Irvine, California
| | - John C Longhurst
- Department of Medicine and Susan-Samueli Institute for Integrative Health, School of Medicine, University of California at Irvine , Irvine, California
| |
Collapse
|
22
|
Investigation of TRPV1 loss-of-function phenotypes in TRPV1 Leu206Stop mice generated by N-ethyl-N-nitrosourea mutagenesis. Biochem Biophys Res Commun 2018; 500:456-461. [PMID: 29660342 DOI: 10.1016/j.bbrc.2018.04.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/13/2018] [Indexed: 11/23/2022]
Abstract
N-ethyl-N-nitrosourea (ENU) random mutagenesis was used to generate a mouse model for the analysis of the transient receptor potential vanilloid 1 (TRPV1) cation channel. A transversion from T→A in exon 4 led to a Leu206Stop mutation generating a loss-of-function mutant. The TRPV1 agonist capsaicin was used to analyze functional and nociceptive parameters in vitro and in vivo in TRPV1 Leu206Stop mice and congenic C3HeB/FeJ controls. Capsaicin-induced [Ca2+]i changes in small diameter DRG neurons were significantly diminished in TRPV1 Leu206Stop mice and administration of capsaicin induced neither hypothermia nor nocifensive behaviour in vivo. TRPV1 Leu206Stop mice were tested in the spinal nerve ligation of mononeuropathic pain and developed mechanical hypersensitivity two weeks after nerve injury. In the open field test, a significant increase in spontaneous locomotion was detected in TRPV1 Leu206Stop mice as compared to wildtype controls. TRPV1 knockout mice have been reported to carry a similar phenotype regarding capsaicin-evoked responses in vitro and in vivo. However, in contrast to TRPV1 Leu206Stop mice, TRPV1 knockout mice did not differ in spontaneous locomotion as compared to congenic C57BL/6 mice, suggesting subtle ENU-dependent or independent strain differences between TRPV1 Leu206Stop mice and their wildtype controls. In summary, these data revealed a target-related (i.e. capsaicin-evoked) phenotype of TRPV1 Leu206Stop mice closely resembling that of published TRPV1 knockout mice. However, since ENU-mutant mice are congenic with the mouse strain initially used in random mutagenesis, direct phenotypic comparison with the respective wildtype controls is possible, and the time-consuming backcrossing in lines with targeted mutations is avoided.
Collapse
|
23
|
Rossato MF, Rigo FK, Oliveira SM, Guerra GP, Silva CR, Cunha TM, Gomez MV, Ferreira J, Trevisan G. Participation of transient receptor potential vanilloid 1 in paclitaxel-induced acute visceral and peripheral nociception in rodents. Eur J Pharmacol 2018; 828:42-51. [PMID: 29577893 DOI: 10.1016/j.ejphar.2018.03.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 10/17/2022]
Abstract
The clinical use of paclitaxel as a chemotherapeutic agent is limited by the severe acute and chronic hypersensitivity caused when it is administered via intraperitoneal or intravenous routes. Thus far, evidence has suggested that transient receptor potential vanilloid-1 (TRPV1) has a key role in the chronic neuropathy induced by paclitaxel. Despite this, the role of TRPV1 in paclitaxel -related acute nociception, especially the development of visceral nociception, has not been evaluated. Thus, the goal of this study was to evaluate the participation of TRPV1 in a model of acute nociception induced by paclitaxel in rats and mice. A single intraperitoneal (i.p.) paclitaxel administration (1 mg/kg, i.p.) produced an immediate visceral nociception response 1 h after administration, caused mechanical and heat hypersensitivity, and diminished burrowing behaviour 24 h after administration. These nociceptive responses were reduced by SB-366791 treatment (0.5 mg/kg, i.p., a TRPV1 antagonist). In addition, TRPV1-positive sensory fibre ablation (using resiniferatoxin, 200 µg/kg, s.c.) reduced visceral nociception and mechanical or heat hypersensitivity caused by paclitaxel injection. Similarly, TRPV1 deficient mice showed a pronounced reduction in mechanical allodynia to paclitaxel acute injection and did not develop heat hypersensitivity. Moreover, 24 h after its injection, paclitaxel induced chemical hypersensitivity to capsaicin (a TRPV1 agonist, 0.01 nmol/site) and increased TRPV1 immunoreactivity in the dorsal root ganglion and sciatic nerve. In conclusion, TRPV1 is involved in mechanical and heat hypersensitivity and spontaneous-pain behaviour induced 24 h after a single paclitaxel injection. This receptor is also involved in visceral nociception induced immediately after paclitaxel administration.
Collapse
Affiliation(s)
- Mateus Fortes Rossato
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Flavia Karine Rigo
- Graduate Program in Health Science, University of the Extreme South of Santa Catarina (Unesc), 88806-000 Criciúma, SC, Brazil; Teaching and Research Institute, Santa Casa de Misericórdia de Belo Horizonte, 30150-221 Belo Horizonte, MG, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Gustavo Petri Guerra
- Federal University of Technology of Paraná (UTFPR), 85884-000 Medianeira, PR, Brazil
| | - Cássia Regina Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil
| | - Marcus Vinícius Gomez
- Teaching and Research Institute, Santa Casa de Misericórdia de Belo Horizonte, 30150-221 Belo Horizonte, MG, Brazil
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), 88049-900 Florianópolis, SC, Brazil
| | - Gabriela Trevisan
- Graduate Program in Health Science, University of the Extreme South of Santa Catarina (Unesc), 88806-000 Criciúma, SC, Brazil; Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil.
| |
Collapse
|
24
|
Rivat C, Sar C, Mechaly I, Leyris JP, Diouloufet L, Sonrier C, Philipson Y, Lucas O, Mallié S, Jouvenel A, Tassou A, Haton H, Venteo S, Pin JP, Trinquet E, Charrier-Savournin F, Mezghrani A, Joly W, Mion J, Schmitt M, Pattyn A, Marmigère F, Sokoloff P, Carroll P, Rognan D, Valmier J. Inhibition of neuronal FLT3 receptor tyrosine kinase alleviates peripheral neuropathic pain in mice. Nat Commun 2018. [PMID: 29531216 PMCID: PMC5847526 DOI: 10.1038/s41467-018-03496-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathic pain (PNP) is a debilitating and intractable chronic disease, for which sensitization of somatosensory neurons present in dorsal root ganglia that project to the dorsal spinal cord is a key physiopathological process. Here, we show that hematopoietic cells present at the nerve injury site express the cytokine FL, the ligand of fms-like tyrosine kinase 3 receptor (FLT3). FLT3 activation by intra-sciatic nerve injection of FL is sufficient to produce pain hypersensitivity, activate PNP-associated gene expression and generate short-term and long-term sensitization of sensory neurons. Nerve injury-induced PNP symptoms and associated-molecular changes were strongly altered in Flt3-deficient mice or reversed after neuronal FLT3 downregulation in wild-type mice. A first-in-class FLT3 negative allosteric modulator, discovered by structure-based in silico screening, strongly reduced nerve injury-induced sensory hypersensitivity, but had no effect on nociception in non-injured animals. Collectively, our data suggest a new and specific therapeutic approach for PNP. Sensitisation of dorsal root ganglia neurons contributes to neuropathic pain. Here the authors demonstrate the cytokine FL contributes to sensitisation of DRGs via its receptor FLT3 expressed on neurons, and identify a novel FLT3 inhibitor that attenuates neuropathic pain in mice.
Collapse
Affiliation(s)
- Cyril Rivat
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Chamroeun Sar
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Ilana Mechaly
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Jean-Philippe Leyris
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Biodol Therapeutics, Cap Alpha, Clapiers, 34830, France
| | - Lucie Diouloufet
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Corinne Sonrier
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Biodol Therapeutics, Cap Alpha, Clapiers, 34830, France
| | - Yann Philipson
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS-Université de Strasbourg, Illkirch, 67400, France
| | - Olivier Lucas
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Sylvie Mallié
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Antoine Jouvenel
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Adrien Tassou
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Henri Haton
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France.,Université de Montpellier, Montpellier, 34000, France
| | - Stéphanie Venteo
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Jean-Philippe Pin
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Univ. Montpellier, 34094, Montpellier, France
| | - Eric Trinquet
- Cisbio Bioassays, Parc Marcel Boiteux, BP84175, 30200, Codolet, France
| | | | - Alexandre Mezghrani
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Willy Joly
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Julie Mion
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Martine Schmitt
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS-Université de Strasbourg, Illkirch, 67400, France
| | - Alexandre Pattyn
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Frédéric Marmigère
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | | | - Patrick Carroll
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France
| | - Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR7200, CNRS-Université de Strasbourg, Illkirch, 67400, France.
| | - Jean Valmier
- Institute for Neurosciences of Montpellier, INSERM, Institut National de la Santé et de la Recherche Médicale, UMR1051, Hôpital Saint-Eloi, Montpellier, 34000, France. .,Université de Montpellier, Montpellier, 34000, France.
| |
Collapse
|
25
|
Backes TM, Rössler OG, Hui X, Grötzinger C, Lipp P, Thiel G. Stimulation of TRPV1 channels activates the AP-1 transcription factor. Biochem Pharmacol 2018; 150:160-169. [PMID: 29452097 DOI: 10.1016/j.bcp.2018.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/08/2018] [Indexed: 02/09/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) channels were originally described as the receptors of capsaicin, the main constituent of hot chili pepper. The biological functions of TRPV1 channels include pain sensation and inflammatory thermal hyperalgesia. Here, we show that stimulation of HEK293 cells expressing TRPV1 channels (H2C1 cells) with capsaicin or the TRPV1 ligand resiniferatoxin activated transcription mediated by the transcription factor AP-1. No cell death was occurring under these experimental conditions. The AP-1 activity was not altered in capsaicin or resiniferatoxin-stimulated HEK293 cells lacking TRPV1. We identified the AP-1 DNA binding site as the capsaicin/resiniferatoxin-responsive element. Stimulation with the TRPV1 ligand N-arachidonoyldopamine increased AP-1 activity in a TRPV1-dependent and TRPV1-independent manner. Stimulation of TRPV1 channels induced an influx of Ca2+ into the cells and this rise in intracellular Ca2+ was essential for activating AP-1 in capsaicin or resiniferatoxin-stimulated cells. N-arachidonoyldopamine stimulation induced a rise in intracellular Ca2+ in a TRPV-1 dependent and independent manner. AP-1 is a dimeric transcription factor, composed of proteins of the c-Jun, c-Fos and ATF families. Stimulation of TRPV1 channels with capsaicin increased c-Jun and c-Fos biosynthesis in H2C1 cells. The signal transduction of capsaicin, leading to enhanced AP-1-mediated transcription, required extracellular signal-regulated protein kinase ERK1/2 as a signal transducer and the activation of the transcription factors c-Jun and ternary complex factor. Together, these data suggest that the intracellular functions of TRPV1 stimulation may rely on the activation of a stimulus-regulated protein kinase and stimulus-responsive transcription factors.
Collapse
Affiliation(s)
- Tobias M Backes
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Oliver G Rössler
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Xin Hui
- Anatomy and Cell Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Carsten Grötzinger
- Department of Internal Medicine, Division of Hepatology and Gastroenterology, Charité, D-13353 Berlin, Germany
| | - Peter Lipp
- Anatomy and Cell Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany
| | - Gerald Thiel
- Department of Medical Biochemistry and Molecular Biology, Saarland University Medical Faculty, D-66421 Homburg, Germany.
| |
Collapse
|
26
|
Valdor M, Wagner A, Röhrs V, Berg J, Fechner H, Schröder W, Tzschentke TM, Bahrenberg G, Christoph T, Kurreck J. RNA interference-based functional knockdown of the voltage-gated potassium channel Kv7.2 in dorsal root ganglion neurons after in vitro and in vivo gene transfer by adeno-associated virus vectors. Mol Pain 2017; 14:1744806917749669. [PMID: 29212407 PMCID: PMC5805000 DOI: 10.1177/1744806917749669] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activation of the neuronal potassium channel Kv7.2 encoded by the KCNQ2 gene has recently been shown to be an attractive mechanism to inhibit nociceptive transmission. However, potent, selective, and clinically proven activators of Kv7.2/Kv7.3 currents with analgesic properties are still lacking. An important prerequisite for the development of new drugs is a model to test the selectivity of novel agonists by abrogating Kv7.2/Kv7.3 function. Since constitutive knockout mice are not viable, we developed a model based on RNA interference-mediated silencing of KCNQ2. By delivery of a KCNQ2-specific short hairpin RNA with adeno-associated virus vectors, we completely abolished the activity of the specific Kv7.2/Kv7.3-opener ICA-27243 in rat sensory neurons. Results obtained in the silencing experiments were consistent between freshly prepared and cryopreserved dorsal root ganglion neurons, as well as in dorsal root ganglion neurons dissociated and cultured after in vivo administration of the silencing vector by intrathecal injections into rats. Interestingly, the tested associated virus serotypes substantially differed with respect to their transduction capability in cultured neuronal cell lines and primary dorsal root ganglion neurons and the in vivo transfer of transgenes by intrathecal injection of associated virus vectors. However, our study provides the proof-of-concept that RNA interference-mediated silencing of KCNQ2 is a suitable approach to create an ex vivo model for testing the specificity of novel Kv7.2/Kv7.3 agonists.
Collapse
Affiliation(s)
- Markus Valdor
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Anke Wagner
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Viola Röhrs
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Johanna Berg
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Henry Fechner
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| | - Wolfgang Schröder
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Thomas M Tzschentke
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | | | - Thomas Christoph
- 1 14938 Grünenthal GmbH , Pharmacology and Biomarker Development, Aachen, Germany
| | - Jens Kurreck
- 2 Department of Applied Biochemistry, Institute of Biotechnology, Berlin University of Technology, Berlin, Germany
| |
Collapse
|
27
|
Hellman KM, Yu PY, Oladosu FA, Segel C, Han A, Prasad PV, Jilling T, Tu FF. The Effects of Platelet-Activating Factor on Uterine Contractility, Perfusion, Hypoxia, and Pain in Mice. Reprod Sci 2017. [PMID: 28631554 DOI: 10.1177/1933719117715122] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is widely hypothesized that menstrual pain is triggered by prostaglandin synthesis that evokes high-pressure uterine contractions and ischemia. However, the effects of molecules implicated in menstrual pain on uterine contractility, perfusion, and oxygenation in vivo have been rarely demonstrated. Studies in women that do not respond to nonsteroidal anti-inflammatory drugs (NSAIDs) have reported elevated levels of platelet-activating factor (PAF). To establish in vivo evidence of PAF's capability to impair uterine homeostasis and to elicit visceral pain, we examined the effects of the PAF receptor agonist (carbamyl PAF [CPAF]) in comparison to other molecules hypothesized to play a role in uterine pain in mice. Uterine pressure was increased by oxytocin, prostaglandin F2α (PGF2α), and CPAF. Even in the absence of inflammatory molecules, uterine contractions reduced uterine oxygenation by 38%. CPAF reduced uterine perfusion by 40% ± 8% and elicited further oxygen desaturation approaching hypoxia (9.4 ± 3.4 mm Hg Pao2). Intraperitoneal injections of CPAF and PGF2α evoked visceral pain and pelvic hyperalgesia in awake wild-type mice. However, pain was not observed in identically injected PAF-receptor knockout mice. Thus, our model provides a demonstration that a molecule implicated in NSAID-resistant dysmenorrhea has a detrimental effect on uterine homeostasis and is capable of causing visceral pain. Our results support the general hypothesis that menstrual cramps are caused by uterine contractions, impaired perfusion, and reduced oxygenation. Since this study was limited to mice, confirmation of these results in humans would be valuable for development of novel therapeutics targeted at inflammatory precursors, contractility, perfusion, and tissue oxygenation.
Collapse
Affiliation(s)
- Kevin M Hellman
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
- 2 Deptartment of Obstetrics and Gynecology, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Peter Y Yu
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Folabomi A Oladosu
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
- 2 Deptartment of Obstetrics and Gynecology, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Chaya Segel
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Alice Han
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Pottumarthi V Prasad
- 3 Department of Radiology, NorthShore University HealthSystem, Evanston, IL, USA
| | - Tamas Jilling
- 4 Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Frank F Tu
- 1 Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, IL, USA
- 2 Deptartment of Obstetrics and Gynecology, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| |
Collapse
|
28
|
Jardín I, López JJ, Diez R, Sánchez-Collado J, Cantonero C, Albarrán L, Woodard GE, Redondo PC, Salido GM, Smani T, Rosado JA. TRPs in Pain Sensation. Front Physiol 2017. [PMID: 28649203 PMCID: PMC5465271 DOI: 10.3389/fphys.2017.00392] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
According to the International Association for the Study of Pain (IASP) pain is characterized as an "unpleasant sensory and emotional experience associated with actual or potential tissue damage". The TRP super-family, compressing up to 28 isoforms in mammals, mediates a myriad of physiological and pathophysiological processes, pain among them. TRP channel might be constituted by similar or different TRP subunits, which will result in the formation of homomeric or heteromeric channels with distinct properties and functions. In this review we will discuss about the function of TRPs in pain, focusing on TRP channles that participate in the transduction of noxious sensation, especially TRPV1 and TRPA1, their expression in nociceptors and their sensitivity to a large number of physical and chemical stimuli.
Collapse
Affiliation(s)
- Isaac Jardín
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José J López
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Raquel Diez
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - José Sánchez-Collado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Carlos Cantonero
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Letizia Albarrán
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Geoffrey E Woodard
- Department of Surgery, Uniformed Services University of the Health SciencesBethesda, MD, United States
| | - Pedro C Redondo
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Ginés M Salido
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysics, Institute of Biomedicine of Sevilla, University of SevilleSevilla, Spain
| | - Juan A Rosado
- Cell Physiology Research Group, Department of Physiology, University of ExtremaduraCáceres, Spain
| |
Collapse
|
29
|
Alsalem M, Millns P, Altarifi A, El-Salem K, Chapman V, Kendall DA. Anti-nociceptive and desensitizing effects of olvanil on capsaicin-induced thermal hyperalgesia in the rat. BMC Pharmacol Toxicol 2016; 17:31. [PMID: 27439609 PMCID: PMC4955132 DOI: 10.1186/s40360-016-0074-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/25/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Olvanil (NE 19550) is a non-pungent synthetic analogue of capsaicin, the natural pungent ingredient of capsicum which activates the transient receptor potential vanilloid type-1 (TRPV1) channel and was developed as a potential analgesic compound. Olvanil has potent anti-hyperalgesic effects in several experimental models of chronic pain. Here we report the inhibitory effects of olvanil on nociceptive processing using cultured dorsal root ganglion (DRG) neurons and compare the effects of capsaicin and olvanil on thermal nociceptive processing in vivo; potential contributions of the cannabinoid CB1 receptor to olvanil's anti-hyperalgesic effects were also investigated. METHODS A hot plate analgesia meter was used to evaluate the anti-nociceptive effects of olvanil on capsaicin-induced thermal hyperalgesia and the role played by CB1 receptors in mediating these effects. Single cell calcium imaging studies of DRG neurons were employed to determine the desensitizing effects of olvanil on capsaicin-evoked calcium responses. Statistical analysis used Student's t test or one way ANOVA followed by Dunnett's post-hoc test as appropriate. RESULTS Both olvanil (100 nM) and capsaicin (100 nM) produced significant increases in intracellular calcium concentrations [Ca(2+)]i in cultured DRG neurons. Olvanil was able to desensitise TRPV1 responses to further capsaicin exposure more effectively than capsaicin. Intraplantar injection of capsaicin (0.1, 0.3 and 1 μg) produced a robust TRPV1-dependant thermal hyperalgesia in rats, whilst olvanil (0.1, 0.3 and 1 μg) produced no hyperalgesia, emphasizing its lack of pungency. The highest dose of olvanil significantly reduced the hyperalgesic effects of capsaicin in vivo. Intraplantar injection of the selective cannabinoid CB1 receptor antagonist rimonabant (1 μg) altered neither capsaicin-induced thermal hyperalgesia nor the desensitizing properties of olvanil, indicating a lack of involvement of CB1 receptors. CONCLUSIONS Olvanil is effective in reducing capsaicin-induced thermal hyperalgesia, probably via directly desensitizing TRPV1 channels in a CB1 receptor-independent fashion. The results presented clearly support the potential for olvanil in the development of new topical analgesic preparations for treating chronic pain conditions while avoiding the unwanted side effects of capsaicin treatments.
Collapse
Affiliation(s)
- Mohammad Alsalem
- Department of Anatomy and Histology, Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan.
| | - Paul Millns
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| | - Ahmad Altarifi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Khalid El-Salem
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Victoria Chapman
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.,Arthritis Research UK Pain Centre, Nottingham, UK
| | - David A Kendall
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK
| |
Collapse
|
30
|
Li W, Wang JX, Zhou ZH, Lu Y, Li XQ, Liu BJ, Chen HS. Contribution of capsaicin-sensitive primary afferents to mechanical hyperalgesia induced by ventral root transection in rats: the possible role of BDNF. Neurol Res 2016; 38:80-5. [DOI: 10.1080/01616412.2015.1135570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
31
|
Emerging Role of Spinal Cord TRPV1 in Pain Exacerbation. Neural Plast 2016; 2016:5954890. [PMID: 26885404 PMCID: PMC4738952 DOI: 10.1155/2016/5954890] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/20/2015] [Accepted: 08/12/2015] [Indexed: 12/25/2022] Open
Abstract
TRPV1 is well known as a sensor ion channel that transduces a potentially harmful environment into electrical depolarization of the peripheral terminal of the nociceptive primary afferents. Although TRPV1 is also expressed in central regions of the nervous system, its roles in the area remain unclear. A series of recent reports on the spinal cord synapses have provided evidence that TRPV1 plays an important role in synaptic transmission in the pain pathway. Particularly, in pathologic pain states, TRPV1 in the central terminal of sensory neurons and interneurons is suggested to commonly contribute to pain exacerbation. These observations may lead to insights regarding novel synaptic mechanisms revealing veiled roles of spinal cord TRPV1 and may offer another opportunity to modulate pathological pain by controlling TRPV1. In this review, we introduce historical perspectives of this view and details of the recent promising results. We also focus on extended issues and unsolved problems to fully understand the role of TRPV1 in pathological pain. Together with recent findings, further efforts for fine analysis of TRPV1's plastic roles in pain synapses at different levels in the central nervous system will promote a better understanding of pathologic pain mechanisms and assist in developing novel analgesic strategies.
Collapse
|
32
|
Fernandes ES, Cerqueira ARA, Soares AG, Costa SKP. Capsaicin and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 929:91-125. [PMID: 27771922 DOI: 10.1007/978-3-319-41342-6_5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A significant number of experimental and clinical studies published in peer-reviewed journals have demonstrated promising pharmacological properties of capsaicin in relieving signs and symptoms of non-communicable diseases (chronic diseases). This chapter provides an overview made from basic and clinical research studies of the potential therapeutic effects of capsaicin, loaded in different application forms, such as solution and cream, on chronic diseases (e.g. arthritis, chronic pain, functional gastrointestinal disorders and cancer). In addition to the anti-inflammatory and analgesic properties of capsaicin largely recognized via, mainly, interaction with the TRPV1, the effects of capsaicin on different cell signalling pathways will be further discussed here. The analgesic, anti-inflammatory or apoptotic effects of capsaicin show promising results in arthritis, neuropathic pain, gastrointestinal disorders or cancer, since evidence demonstrates that the oral or local application of capsaicin reduce inflammation and pain in rheumatoid arthritis, promotes gastric protection against ulcer and induces apoptosis of the tumour cells. Sadly, these results have been paralleled by conflicting studies, which indicate that high concentrations of capsaicin are likely to evoke deleterious effects, thus suggesting that capsaicin activates different pathways at different concentrations in both human and rodent tissues. Thus, to establish effective capsaicin doses for chronic conditions, which can be benefited from capsaicin therapeutic effects, is a real challenge that must be pursued.
Collapse
Affiliation(s)
- E S Fernandes
- Programa de Pós-Graduação, Universidade Ceuma, São Luís-MA, Brazil.,Vascular Biology Section, Cardiovascular Division, King's College London, London, UK
| | - A R A Cerqueira
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil
| | - A G Soares
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil
| | - Soraia K P Costa
- Department of Pharmacology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), Av. Prof. Lineu Prestes, 1524 - Room 326, Butantan, São Paulo, 05508-900, Sao Paulo, Brazil.
| |
Collapse
|
33
|
Muley MM, Krustev E, McDougall JJ. Preclinical Assessment of Inflammatory Pain. CNS Neurosci Ther 2015; 22:88-101. [PMID: 26663896 DOI: 10.1111/cns.12486] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 12/21/2022] Open
Abstract
While acute inflammation is a natural physiological response to tissue injury or infection, chronic inflammation is maladaptive and engenders a considerable amount of adverse pain. The chemical mediators responsible for tissue inflammation act on nociceptive nerve endings to lower neuronal excitation threshold and sensitize afferent firing rate leading to the development of allodynia and hyperalgesia, respectively. Animal models have aided in our understanding of the pathophysiological mechanisms responsible for the generation of chronic inflammatory pain and allowed us to identify and validate numerous analgesic drug candidates. Here we review some of the commonly used models of skin, joint, and gut inflammatory pain along with their relative benefits and limitations. In addition, we describe and discuss several behavioral and electrophysiological approaches used to assess the inflammatory pain in these preclinical models. Despite significant advances having been made in this area, a gap still exists between fundamental research and the implementation of these findings into a clinical setting. As such we need to characterize inherent pathophysiological pathways and develop new endpoints in these animal models to improve their predictive value of human inflammatory diseases in order to design safer and more effective analgesics.
Collapse
Affiliation(s)
- Milind M Muley
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Eugene Krustev
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
34
|
Planells-Cases1 R, Ferrer-Montiel A. Drug design and development through the vanilloid receptor. Expert Opin Drug Discov 2015; 2:1053-63. [PMID: 23484872 DOI: 10.1517/17460441.2.8.1053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The vanilloid receptor (TRPV1) has attracted a great expectation in pain therapeutics for the treatment of chronic inflammatory conditions. As a result, several drug discovery programmes were launched in the past years that yielded a large number of receptor agonists and antagonists. However, despite the claimed therapeutic potential of TRPV1 modulators, a disappointing number of candidates have progressed into clinical trials and those were only for dental pain and migraine, indicating that our understanding of the role of TRPV1 in pain is still very limited. The widespread distribution of TRPV1 in different tissues suggests an involvement in body functions other than pain. Indeed, new findings indicate that TRPV1 is tonically active in physiological conditions and its pharmacological blockade leads to hyperthermia. Furthermore, the full abrogation of TRPV1 in some models of chronic pain results in enhanced pain. Therefore, a remaining challenge is the development of drugs that preserve the physiological activity of TRPV1 and downregulate the function of overactive receptors.
Collapse
|
35
|
Vizin RCL, Scarpellini CDS, Ishikawa DT, Correa GM, de Souza CO, Gargaglioni LH, Carrettiero DC, Bícego KC, Almeida MC. TRPV4 activates autonomic and behavioural warmth-defence responses in Wistar rats. Acta Physiol (Oxf) 2015; 214:275-89. [PMID: 25739906 DOI: 10.1111/apha.12477] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/10/2015] [Accepted: 02/26/2015] [Indexed: 11/28/2022]
Abstract
AIM In this study, we aimed at investigating the involvement of the warmth-sensitive channel - TRPV4 (in vitro sensitive to temperatures in the range of approx. 24-34 °C) - on the thermoregulatory mechanisms in rats. METHODS We treated rats with a chemical selective agonist (RN-1747) and two antagonists (RN-1734 and HC-067047) of the TRPV4 channel and measured core body temperature, metabolism, heat loss index and preferred ambient temperature. RESULTS Our data revealed that chemical activation of TRPV4 channels by topical application of RN-1747 on the skin leads to hypothermia and this effect was blocked by the pre-treatment with the selective antagonist of this channel. Intracerebroventricular treatment with RN-1747 did not cause hypothermia, indicating that the observed response was indeed due to activation of TRPV4 channels in the periphery. Intravenous blockade of this channel with HC-067047 caused an increase in core body temperature at ambient temperature of 26 and 30 °C, but not at 22 and 32 °C. At 26 °C, HC-067047-induced hyperthermia was accompanied by increase in oxygen consumption (an index of thermogenesis), while chemical stimulation of TRPV4 increased tail heat loss, indicating that these two autonomic thermoeffectors in the rat are modulated through TRPV4 channels. Furthermore, rats chemically stimulated with TRPV4 agonist choose colder ambient temperatures and cold-seeking behaviour after thermal stimulation (28-31 °C) was inhibited by TRPV4 antagonist. CONCLUSION Our results suggest, for the first time, that TRPV4 channel is involved in the recruitment of behavioural and autonomic warmth-defence responses to regulate core body temperature.
Collapse
Affiliation(s)
- R. C. L. Vizin
- Graduate Program on Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - C. da S. Scarpellini
- Department of Animal Morphology and Physiology; College of Agricultural and Veterinary Sciences; São Paulo State University; Jaboticabal SP Brazil
- Joint UFSCar-UNESP Graduate Program of Physiological Sciences; Sao Carlos SP Brazil
- National Institute of Science and Technology in Comparative Physiology (INCT - Fisiologia Comparada); Jaboticabal SP Brazil
| | - D. T. Ishikawa
- Graduate Program on Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - G. M. Correa
- Department of Animal Morphology and Physiology; College of Agricultural and Veterinary Sciences; São Paulo State University; Jaboticabal SP Brazil
- National Institute of Science and Technology in Comparative Physiology (INCT - Fisiologia Comparada); Jaboticabal SP Brazil
| | - C. O. de Souza
- Graduate Program on Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - L. H. Gargaglioni
- Department of Animal Morphology and Physiology; College of Agricultural and Veterinary Sciences; São Paulo State University; Jaboticabal SP Brazil
- National Institute of Science and Technology in Comparative Physiology (INCT - Fisiologia Comparada); Jaboticabal SP Brazil
| | - D. C. Carrettiero
- Graduate Program on Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
- Natural and Humanities Science Center; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| | - K. C. Bícego
- Department of Animal Morphology and Physiology; College of Agricultural and Veterinary Sciences; São Paulo State University; Jaboticabal SP Brazil
- National Institute of Science and Technology in Comparative Physiology (INCT - Fisiologia Comparada); Jaboticabal SP Brazil
| | - M. C. Almeida
- Graduate Program on Neuroscience and Cognition; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
- Natural and Humanities Science Center; Universidade Federal do ABC (UFABC); São Bernardo do Campo SP Brazil
| |
Collapse
|
36
|
Acupuncture for visceral pain: neural substrates and potential mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:609594. [PMID: 25614752 PMCID: PMC4295157 DOI: 10.1155/2014/609594] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/13/2014] [Accepted: 12/13/2014] [Indexed: 12/17/2022]
Abstract
Visceral pain is the most common form of pain caused by varied diseases and a major reason for patients to seek medical consultation. Despite much advances, the pathophysiological mechanism is still poorly understood comparing with its somatic counterpart and, as a result, the therapeutic efficacy is usually unsatisfactory. Acupuncture has long been used for the management of numerous disorders in particular pain and visceral pain, characterized by the high therapeutic benefits and low adverse effects. Previous findings suggest that acupuncture depresses pain via activation of a number of neurotransmitters or modulators including opioid peptides, serotonin, norepinephrine, and adenosine centrally and peripherally. It endows us, by advancing the understanding of the role of ion channels and gut microbiota in pain process, with novel perspectives to probe the mechanisms underlying acupuncture analgesia. In this review, after describing the visceral innervation and the relevant afferent pathways, in particular the ion channels in visceral nociception, we propose three principal mechanisms responsible for acupuncture induced benefits on visceral pain. Finally, potential topics are highlighted regarding the future studies in this field.
Collapse
|
37
|
Sousa-Valente J, Andreou AP, Urban L, Nagy I. Transient receptor potential ion channels in primary sensory neurons as targets for novel analgesics. Br J Pharmacol 2014; 171:2508-27. [PMID: 24283624 DOI: 10.1111/bph.12532] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/11/2013] [Accepted: 11/20/2013] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed an explosion in novel findings relating to the molecules involved in mediating the sensation of pain in humans. Transient receptor potential (TRP) ion channels emerged as the greatest group of molecules involved in the transduction of various physical stimuli into neuronal signals in primary sensory neurons, as well as, in the development of pain. Here, we review the role of TRP ion channels in primary sensory neurons in the development of pain associated with peripheral pathologies and possible strategies to translate preclinical data into the development of effective new analgesics. Based on available evidence, we argue that nociception-related TRP channels on primary sensory neurons provide highly valuable targets for the development of novel analgesics and that, in order to reduce possible undesirable side effects, novel analgesics should prevent the translocation from the cytoplasm to the cell membrane and the sensitization of the channels rather than blocking the channel pore or binding sites for exogenous or endogenous activators.
Collapse
Affiliation(s)
- J Sousa-Valente
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | | | | | | |
Collapse
|
38
|
Down-regulation of Stargazin inhibits the enhanced surface delivery of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor GluR1 subunit in rat dorsal horn and ameliorates postoperative pain. Anesthesiology 2014; 121:609-19. [PMID: 25093662 DOI: 10.1097/aln.0000000000000291] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Stargazin is the first transmembrane protein known to regulate synaptic targeting of α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors. However, it is unclear whether regulation of the surface delivery of spinal AMPA receptor subunits by stargazin contributes to postoperative pain development. METHODS Western blot analysis was used to examine changes in the surface delivery of AMPA receptor subunits, GluR1 and GluR2, in rat dorsal horn. The interaction between stargazin and GluR1 and GluR2 was examined by coimmunoprecipitation. Expression of stargazin was suppressed by intrathecal administration of small interfering RNA311. RESULTS Membrane-bound GluR1, but not GluR2, in ipsilateral dorsal horn was increased at 3 h (1.49 ± 0.15-fold of β-tubulin, mean ± SEM) and 1 day (1.03 ± 0.25) after incision, as compared with that in control rats (naive, 0.63 ± 0.23, P < 0.05, n = 6 per group). The amount of GluR1 coimmunoprecipitated with stargazin was greater at 3 h after incision (1.48 ± 0.31-fold of input) than that in control animals (0.45 ± 0.24, P < 0.05, n = 6 per group). Importantly, the increase in membrane GluR1 at 3 h after incision was normalized to near control level (0.72 ± 0.20-fold of β-tubulin) by pretreatment with intrathecal stargazin small interfering RNA311 (0.87 ± 0.09), but not scrambled small interfering RNA (1.48 ± 0.24) or vehicle (1.25 ± 0.13, P < 0.05, n = 6 per group). Stargazin small interfering RNA311 pretreatment prevented the increase in stargazin-GluR1 interaction and decreased postoperative pain after incision. CONCLUSIONS This study suggests a critical role of stargazin-mediated surface delivery of GluR1 subunit in the development of postoperative pain. A better therapeutic strategy for postoperative pain may involve selectively down-regulating spinal stargazin to inhibit synaptic targeting of GluR1 subunit.
Collapse
|
39
|
Ryskamp DA, Redmon S, Jo AO, Križaj D. TRPV1 and Endocannabinoids: Emerging Molecular Signals that Modulate Mammalian Vision. Cells 2014; 3:914-38. [PMID: 25222270 PMCID: PMC4197638 DOI: 10.3390/cells3030914] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/27/2014] [Accepted: 09/05/2014] [Indexed: 01/18/2023] Open
Abstract
Transient Receptor Potential Vanilloid 1 (TRPV1) subunits form a polymodal cation channel responsive to capsaicin, heat, acidity and endogenous metabolites of polyunsaturated fatty acids. While originally reported to serve as a pain and heat detector in the peripheral nervous system, TRPV1 has been implicated in the modulation of blood flow and osmoregulation but also neurotransmission, postsynaptic neuronal excitability and synaptic plasticity within the central nervous system. In addition to its central role in nociception, evidence is accumulating that TRPV1 contributes to stimulus transduction and/or processing in other sensory modalities, including thermosensation, mechanotransduction and vision. For example, TRPV1, in conjunction with intrinsic cannabinoid signaling, might contribute to retinal ganglion cell (RGC) axonal transport and excitability, cytokine release from microglial cells and regulation of retinal vasculature. While excessive TRPV1 activity was proposed to induce RGC excitotoxicity, physiological TRPV1 activity might serve a neuroprotective function within the complex context of retinal endocannabinoid signaling. In this review we evaluate the current evidence for localization and function of TRPV1 channels within the mammalian retina and explore the potential interaction of this intriguing nociceptor with endogenous agonists and modulators.
Collapse
Affiliation(s)
- Daniel A Ryskamp
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Sarah Redmon
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Andrew O Jo
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, Moran Eye Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| |
Collapse
|
40
|
Walter C, Oertel B, Lötsch J. THC may reproducibly induce electrical hyperalgesia in healthy volunteers. Eur J Pain 2014; 19:516-8. [DOI: 10.1002/ejp.575] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2014] [Indexed: 01/28/2023]
Affiliation(s)
- C. Walter
- Institute of Clinical Pharmacology; Goethe - University; Frankfurt am Main Germany
| | - B.G. Oertel
- Project Group Translational Medicine and Pharmacology (IME-TMP); Fraunhofer Institute of Molecular Biology and Applied Ecology; Frankfurt am Main Germany
| | - J. Lötsch
- Institute of Clinical Pharmacology; Goethe - University; Frankfurt am Main Germany
- Project Group Translational Medicine and Pharmacology (IME-TMP); Fraunhofer Institute of Molecular Biology and Applied Ecology; Frankfurt am Main Germany
| |
Collapse
|
41
|
Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev 2014; 66:676-814. [PMID: 24951385 DOI: 10.1124/pr.113.008268] [Citation(s) in RCA: 382] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The large Trp gene family encodes transient receptor potential (TRP) proteins that form novel cation-selective ion channels. In mammals, 28 Trp channel genes have been identified. TRP proteins exhibit diverse permeation and gating properties and are involved in a plethora of physiologic functions with a strong impact on cellular sensing and signaling pathways. Indeed, mutations in human genes encoding TRP channels, the so-called "TRP channelopathies," are responsible for a number of hereditary diseases that affect the musculoskeletal, cardiovascular, genitourinary, and nervous systems. This review gives an overview of the functional properties of mammalian TRP channels, describes their roles in acquired and hereditary diseases, and discusses their potential as drug targets for therapeutic intervention.
Collapse
Affiliation(s)
- Bernd Nilius
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| | - Arpad Szallasi
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Ion Channel Research, Campus Gasthuisberg, Leuven, Belgium (B.N.); and Department of Pathology, Monmouth Medical Center, Long Branch, New Jersey (A.S.)
| |
Collapse
|
42
|
Farrell KE, Callister RJ, Keely S. Understanding and targeting centrally mediated visceral pain in inflammatory bowel disease. Front Pharmacol 2014; 5:27. [PMID: 24634658 PMCID: PMC3942649 DOI: 10.3389/fphar.2014.00027] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/13/2014] [Indexed: 11/17/2022] Open
Affiliation(s)
- Kristen E Farrell
- School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia
| | - Robert J Callister
- School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, The University of Newcastle Callaghan, NSW, Australia ; Gastrointestinal Research Group, Viruses, Infection/Immunity, Vaccines and Asthma Program, Hunter Medical Research Institute New Lambton Heights, NSW, Australia
| |
Collapse
|
43
|
Luo J, Walters ET, Carlton SM, Hu H. Targeting Pain-evoking Transient Receptor Potential Channels for the Treatment of Pain. Curr Neuropharmacol 2014; 11:652-63. [PMID: 24396340 PMCID: PMC3849790 DOI: 10.2174/1570159x113119990040] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 06/19/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023] Open
Abstract
Chronic pain affects billions of lives globally and is a major public health problem in the United States. However, pain management is still a challenging task due to a lack of understanding of the fundamental mechanisms of pain. In the past decades transient receptor potential (TRP) channels have been identified as molecular sensors of tissue damage and inflammation. Activation/sensitization of TRP channels in peripheral nociceptors produces neurogenic inflammation and contributes to both somatic and visceral pain. Pharmacological and genetic studies have affirmed the role of TRP channels in multiple forms of inflammatory and neuropathic pain. Thus pain-evoking TRP channels emerge as promising therapeutic targets for a wide variety of pain and inflammatory conditions.
Collapse
Affiliation(s)
- Jialie Luo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| | - Susan M Carlton
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1069
| | - Hongzhen Hu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| |
Collapse
|
44
|
Matricon J, Muller E, Accarie A, Meleine M, Etienne M, Voilley N, Busserolles J, Eschalier A, Lazdunski M, Bourdu S, Gelot A, Ardid D. Peripheral contribution of NGF and ASIC1a to colonic hypersensitivity in a rat model of irritable bowel syndrome. Neurogastroenterol Motil 2013; 25:e740-54. [PMID: 23902154 DOI: 10.1111/nmo.12199] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder associated with idiopathic colonic hypersensitivity (CHS). However, recent studies suggest that low-grade inflammation could underlie CHS in IBS. The pro-inflammatory mediator nerve growth factor (NGF) plays a key role in the sensitization of peripheral pain pathways and several studies have reported its contribution to visceral pain development. NGF modulates the expression of Acid-Sensing Ion Channels (ASICs), which are proton sensors involved in sensory neurons sensitization. This study examined the peripheral contribution of NGF and ASICs to IBS-like CHS induced by butyrate enemas in the rat colon. METHODS Colorectal distension and immunohistochemical staining of sensory neurons were used to evaluate NGF and ASICs contribution to the development of butyrate-induced CHS. KEY RESULTS Systemic injection of anti-NGF antibodies or the ASICs inhibitor amiloride prevented the development of butyrate-induced CHS. A significant increase in NGF and ASIC1a protein expression levels was observed in sensory neurons of rats displaying butyrate-induced CHS. This increase was specific of small- and medium-diameter L1 + S1 sensory neurons, where ASIC1a was co-expressed with NGF or trkA in CGRP-immunoreactive somas. ASIC1a was also overexpressed in retrogradely labeled colon sensory neurons. Interestingly, anti-NGF antibody administration prevented ASIC1a overexpression in sensory neurons of butyrate-treated rats. CONCLUSIONS & INFERENCES Our data suggest that peripheral NGF and ASIC1a concomitantly contribute to the development of butyrate-induced CHS NGF-ASIC1a interplay may have a pivotal role in the sensitization of colonic sensory neurons and as such, could be considered as a potential new therapeutic target for IBS treatment.
Collapse
Affiliation(s)
- J Matricon
- NEURO-DOL, Pharmacologie Fondamentale et Clinique de la Douleur, Faculté de Médecine, INSERM/UdA, UMR 1107, Université d'Auvergne, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hirai T, Enomoto M, Kaburagi H, Sotome S, Yoshida-Tanaka K, Ukegawa M, Kuwahara H, Yamamoto M, Tajiri M, Miyata H, Hirai Y, Tominaga M, Shinomiya K, Mizusawa H, Okawa A, Yokota T. Intrathecal AAV serotype 9-mediated delivery of shRNA against TRPV1 attenuates thermal hyperalgesia in a mouse model of peripheral nerve injury. Mol Ther 2013; 22:409-419. [PMID: 24322332 DOI: 10.1038/mt.2013.247] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 10/17/2013] [Indexed: 01/03/2023] Open
Abstract
Gene therapy for neuropathic pain requires efficient gene delivery to both central and peripheral nervous systems. We previously showed that an adenoassociated virus serotype 9 (AAV9) vector expressing short-hairpin RNA (shRNA) could suppress target molecule expression in the dorsal root ganglia (DRG) and spinal cord upon intrathecal injection. To evaluate the therapeutic potential of this approach, we constructed an AAV9 vector encoding shRNA against vanilloid receptor 1 (TRPV1), which is an important target gene for acute pain, but its role in chronic neuropathic pain remains unclear. We intrathecally injected it into the subarachnoid space at the upper lumbar spine of mice 3 weeks after spared nerve injury (SNI). Delivered shTRPV1 effectively suppressed mRNA and protein expression of TRPV1 in the DRG and spinal cord, and it attenuated nerve injury-induced thermal allodynia 10-28 days after treatment. Our study provides important evidence for the contribution of TRPV1 to thermal hypersensitivity in neuropathic pain and thus establishes intrathecal AAV9-mediated gene delivery as an investigative and potentially therapeutic platform for the nervous system.
Collapse
Affiliation(s)
- Takashi Hirai
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuhiro Enomoto
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Hidetoshi Kaburagi
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinichi Sotome
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kie Yoshida-Tanaka
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Madoka Ukegawa
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroya Kuwahara
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mariko Yamamoto
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mio Tajiri
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Haruka Miyata
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yukihiko Hirai
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Okazaki, Japan
| | - Kenichi Shinomiya
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidehiro Mizusawa
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Okawa
- Department of Orthopaedic Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
46
|
Chen HS, Li FP, Li XQ, Liu BJ, Qu F, Wen WW, Wang Y, Lin Q. Acute stress regulates nociception and inflammatory response induced by bee venom in rats: possible mechanisms. Stress 2013; 16:557-63. [PMID: 23574036 DOI: 10.3109/10253890.2013.794336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Restraint stress modulates pain and inflammation. The present study was designed to evaluate the effect of acute restraint stress on inflammatory pain induced by subcutaneous injection of bee venom (BV). First, we investigated the effect of 1 h restraint on the spontaneous paw-flinching reflex (SPFR), decrease in paw withdrawal mechanical threshold (PWMT) and increase in paw volume (PV) of the injected paw induced by BV. SPFR was measured immediately after BV injection, and PWMT and PV were measured 2 h before BV and 2-8 h after BV. The results showed that acute restraint inhibited significantly the SPFR but failed to affect mechanical hyperalgesia. In contrast, stress enhanced significantly inflammatory swelling of the injected paw. In a second series of experiments, the effects of pretreatment with capsaicin locally applied to the sciatic nerve, systemic 6-hydroxydopamine (6-OHDA), and systemic naloxone were examined on the antinociception and proinflammation produced by acute restraint stress. Local capsaicin pretreatment inhibited BV-induced nociception and inflammatory edema, and had additive effects with stress on nociception but reduced stress enhancement of edema. Systemic 6-OHDA treatment attenuated the proinflammatory effect of stress, but did not affect the antinociceptive effect. Systemic naloxone pretreatment eliminated the antinociceptive effect of stress, but did not affect proinflammation. Taken together, our data indicate that acute restraint stress contributes to antinociception via activating an endogenous opioid system, while sympathetic postganglionic fibers may contribute to enhanced inflammation in the BV pain model.
Collapse
Affiliation(s)
- Hui-Sheng Chen
- Department of Neurology, General Hospital of Shen-Yang Military Region, Shen Yang 110840, China.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Lack of transient receptor potential vanilloid 1 channel modulates the development of neurogenic bladder dysfunction induced by cross-sensitization in afferent pathways. J Neuroinflammation 2013; 10:3. [PMID: 23305398 PMCID: PMC3556132 DOI: 10.1186/1742-2094-10-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 12/21/2012] [Indexed: 12/30/2022] Open
Abstract
Background Bladder pain of unknown etiology has been associated with co-morbid conditions and functional abnormalities in neighboring pelvic organs. Mechanisms underlying pain co-morbidities include cross-sensitization, which occurs predominantly via convergent neural pathways connecting distinct pelvic organs. Our previous results showed that colonic inflammation caused detrusor instability via activation of transient receptor potential vanilloid 1 (TRPV1) signaling pathways, therefore, we aimed to determine whether neurogenic bladder dysfunction can develop in the absence of TRPV1 receptors. Methods Adult male C57BL/6 wild-type (WT) and TRPV1−/− (knockout) mice were used in this study. Colonic inflammation was induced by intracolonic trinitrobenzene sulfonic acid (TNBS). The effects of transient colitis on abdominal sensitivity and function of the urinary bladder were evaluated by cystometry, contractility and relaxation of detrusor smooth muscle (DSM) in vitro to various stimuli, gene and protein expression of voltage-gated sodium channels in bladder sensory neurons, and pelvic responses to mechanical stimulation. Results Knockout of TRPV1 gene did not eliminate the development of cross-sensitization between the colon and urinary bladder. However, TRPV1−/− mice had prolonged intermicturition interval and increased number of non-voiding contractions at baseline followed by reduced urodynamic responses during active colitis. Contractility of DSM was up-regulated in response to KCl in TRPV1−/− mice with inflamed colon. Application of Rho-kinase inhibitor caused relaxation of DSM in WT but not in TRPV1−/− mice during colonic inflammation. TRPV1−/− mice demonstrated blunted effects of TNBS-induced colitis on expression and function of voltage-gated sodium channels in bladder sensory neurons, and delayed development of abdominal hypersensitivity upon colon-bladder cross-talk in genetically modified animals. Conclusions The lack of TRPV1 receptors does not eliminate the development of cross-sensitization in the pelvis. However, the function of the urinary bladder significantly differs between WT and TRPV−/− mice especially upon development of colon-bladder cross-sensitization induced by transient colitis. Our results suggest that TRPV1 pathways may participate in the development of chronic pelvic pain co-morbidities in humans.
Collapse
|
48
|
Kitagawa Y, Tamai I, Hamada Y, Usui K, Wada M, Sakata M, Matsushita M. The Orally Administered Selective TRPV1 Antagonist, JTS-653, Attenuates Chronic Pain Refractory to Non-steroidal Anti-inflammatory Drugs in Rats and Mice Including Post-herpetic Pain. J Pharmacol Sci 2013; 122:128-37. [DOI: 10.1254/jphs.12276fp] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
49
|
|
50
|
Formaldehyde up-regulates TRPV1 through MAPK and PI3K signaling pathways in a rat model of bone cancer pain. Neurosci Bull 2012; 28:165-72. [PMID: 22466127 DOI: 10.1007/s12264-012-1211-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE Our previous study showed that tumor tissue-derived formaldehyde at low concentrations plays an important role in bone cancer pain through activating transient receptor potential vanilloid subfamily member 1 (TRPV1). The present study further explored whether this tumor tissue-derived endogenous formaldehyde regulates TRPV1 expression in a rat model of bone cancer pain, and if so, what the possible signal pathways are during the development of this type of pain. METHODS A rat model of bone cancer pain was established by injecting living MRMT-1 tumor cells into the tibia. The formaldehyde levels were determined by high performance liquid chromatography, and the expression of TRPV1 was examined with Western blot and RT-PCR. In primary cultured dorsal root ganglion (DRG) neurons, the expression of TRPV1 was assessed after treatment with 100 µmol/L formaldehyde with or without pre-addition of PD98059 [an inhibitor for extracellular signal-regulated kinase], SB203580 (a p38 inhibitor), SP600125 [an inhibitor for c-Jun N-terminal kinase], BIM [a protein kinase C (PKC) inhibitor] or LY294002 [a phosphatidylinositol 3-kinase (PI3K) inhibitor]. RESULTS In the rat model of bone cancer pain, formaldehyde concentration increased in blood plasma, bone marrow and the spinal cord. TRPV1 protein expression was also increased in the DRG. In primary cultured DRG neurons, 100 μmol/L formaldehyde significantly increased the TRPV1 expression level. Pre-incubation with PD98059, SB203580, SP600125 or LY294002, but not BIM, inhibited the formaldehyde-induced increase of TRPV1 expression. CONCLUSION Formaldehyde at a very low concentration up-regulates TRPV1 expression through mitogen-activated protein kinase and PI3K, but not PKC, signaling pathways. These results further support our previous finding that TRPV1 in peripheral afferents plays a role in bone cancer pain.
Collapse
|