1
|
Shehab S, Hamad MIK, Emerald BS. A novel approach to completely alleviate peripheral neuropathic pain in human patients: insights from preclinical data. Front Neuroanat 2025; 18:1523095. [PMID: 39839257 PMCID: PMC11747518 DOI: 10.3389/fnana.2024.1523095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
Neuropathic pain is a pervasive health concern worldwide, posing significant challenges to both clinicians and neuroscientists. While acute pain serves as a warning signal for potential tissue damage, neuropathic pain represents a chronic pathological condition resulting from injury or disease affecting sensory pathways of the nervous system. Neuropathic pain is characterized by long-lasting ipsilateral hyperalgesia (increased sensitivity to pain), allodynia (pain sensation in response to stimuli that are not normally painful), and spontaneous unprovoked pain. Current treatments for neuropathic pain are generally inadequate, and prevention remains elusive. In this review, we provide an overview of current treatments, their limitations, and a discussion on the potential of capsaicin and its analog, resiniferatoxin (RTX), for complete alleviation of nerve injury-induced neuropathic pain. In an animal model of neuropathic pain where the fifth lumbar (L5) spinal nerve is unilaterally ligated and cut, resulting in ipsilateral hyperalgesia, allodynia, and spontaneous pain akin to human neuropathic pain. The application of capsaicin or RTX to the adjacent uninjured L3 and L4 nerves completely alleviated and prevented mechanical and thermal hyperalgesia following the L5 nerve injury. The effects of this treatment were specific to unmyelinated fibers (responsible for pain sensation), while thick myelinated nerve fibers (responsible for touch and mechanoreceptor sensations) remained intact. Here, we propose to translate these promising preclinical results into effective therapeutic interventions in humans by direct application of capsaicin or RTX to adjacent uninjured nerves in patients who suffer from neuropathic pain due to peripheral nerve injury, following surgical interventions, diabetic neuropathy, trauma, vertebral disc herniation, nerve entrapment, ischemia, postherpetic lesion, and spinal cord injury.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | | |
Collapse
|
2
|
Shen YR, Cheng L, Zhang DF. TRPV1: A novel target for the therapy of diabetes and diabetic complications. Eur J Pharmacol 2024; 984:177021. [PMID: 39362389 DOI: 10.1016/j.ejphar.2024.177021] [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: 06/20/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Diabetes mellitus is a chronic metabolic disease characterized by abnormally elevated blood glucose levels. Type II diabetes accounts for approximately 90% of all cases. Several drugs are available for hyperglycemia treatment. However, the current therapies for managing high blood glucose do not prevent or reverse the disease progression, which may result in complications and adverse effects, including diabetic neuropathy, retinopathy, and nephropathy. Hence, developing safer and more effective methods for lowering blood glucose levels is imperative. Transient receptor potential vanilloid-1 (TRPV1) is a significant member of the transient receptor potential family. It is present in numerous body tissues and organs and performs vital physiological functions. PURPOSE This review aimed to develop new targeted TRPV1 hypoglycemic drugs by systematically summarizing the mechanism of action of the TRPV1-based signaling pathway in preventing and treating diabetes and its complications. METHODS Literature searches were performed in the PubMed, Web of Science, Google Scholar, Medline, and Scopus databases for 10 years from 2013 to 2023. The search terms included "diabetes," "TRPV1," "diabetic complications," and "capsaicin." RESULTS TRPV1 is an essential potential target for treating diabetes mellitus and its complications. It reduces hepatic glucose production and food intake and promotes thermogenesis, metabolism, and insulin secretion. Activation of TRPV1 ameliorates diabetic nephropathy, retinopathy, myocardial infarction, vascular endothelial dysfunction, gastroparesis, and bladder dysfunction. Suppression of TRPV1 improves diabetes-related osteoporosis. However, the therapeutic effects of activating or suppressing TRPV1 may vary when treating diabetic neuropathy and periodontitis. CONCLUSION This review demonstrates that TRPV1 is a potential therapeutic target for diabetes and its complications. Additionally, it provides a theoretical basis for developing new hypoglycemic drugs that target TRPV1.
Collapse
Affiliation(s)
- Yu-Rong Shen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Long Cheng
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Dong-Fang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
3
|
Pacifico P, Menichella DM. Molecular mechanisms of neuropathic pain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:279-309. [PMID: 39580215 DOI: 10.1016/bs.irn.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Peripheral neuropathic pain, which occurs after a lesion or disease affecting the peripheral somatosensory nervous system, is a complex and challenging condition to treat. This chapter will cover molecular mechanisms underlying the pathophysiology of peripheral neuropathic pain, focusing on (1) sensitization of nociceptors, (2) neuro-immune crosstalk, and (3) axonal degeneration and regeneration. The chapter will also emphasize the importance of identifying novel therapeutic targets in non-neuronal cells. A comprehensive understanding of how changes at both neuronal and non-neuronal levels contribute to peripheral neuropathic pain may significantly improve pain management and treatment options, expanding to topical application that bypass the side effects associated with systemic administration.
Collapse
Affiliation(s)
- Paola Pacifico
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Daniela M Menichella
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
4
|
Chinigò G, Ruffinatti FA, Munaron L. The potential of TRP channels as new prognostic and therapeutic targets against prostate cancer progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189226. [PMID: 39586480 DOI: 10.1016/j.bbcan.2024.189226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Prostate cancer (PCa) is the second deadliest cancer among men worldwide. Particularly critical is its development towards metastatic androgen-independent forms for which the current therapies are ineffective. Indeed, the 5-year relative survival for PCa drops dramatically to 34 % in the presence of metastases. The superfamily of Transient Receptor Potential (TRP) channels could answer the urgent request to identify new prognostic and therapeutic tools against metastatic PCa. Indeed, this class of ion channels revealed an appealing de-regulation during PCa development and its progression towards aggressive forms. Altered expression and/or functionality of several TRPs have been associated with the PCa metastatic cascade by significantly impacting tumor growth, invasiveness, and angiogenesis. In this review, we will dissect the contribution of TRP channels in such hallmarks of PCa and then discuss their applicability as new prognostic and therapeutic agents in the fight against metastatic PCa. In particular, the great potential of TRPM8, TRPV6, and TRPA1 in opening the way to new treatment perspectives will be highlighted.
Collapse
Affiliation(s)
- Giorgia Chinigò
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| | | | - Luca Munaron
- University of Turin, Department of Life Sciences and Systems Biology, via Accademia Albertina 13, 10123 Turin, Italy.
| |
Collapse
|
5
|
Miliano C, Natividad LA, Quello S, Stoolmiller M, Gregus AM, Buczynski MW, Mason BJ. The Predictive Value of Plasma Bioactive Lipids on Craving in Human Volunteers With Alcohol Use Disorder. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100368. [PMID: 39282655 PMCID: PMC11400622 DOI: 10.1016/j.bpsgos.2024.100368] [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: 03/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 09/19/2024] Open
Abstract
Background Alcohol use disorder (AUD) is a chronic relapsing disorder characterized by alcohol seeking and consumption despite negative consequences. Despite the availability of multiple treatments, patients continue to exhibit high relapse rates. Thus, biomarkers that can identify patients at risk for heightened craving are urgently needed. Mounting preclinical and clinical evidence implicates perturbations in bioactive lipid signaling in the neurobiology of craving in AUD. We hypothesize that these lipids are potential biomarkers for predicting alcohol craving in patients with AUD. Methods This study used archival deidentified clinical data and corresponding plasma specimens from 157 participants in 3 clinical studies of AUD. We evaluated plasma levels of 8 lipid species as predictors of craving in response to in vivo alcohol and affective cues during abstinence. Results Participants were 109 men and 48 women who met DSM-5 criteria for severe AUD. We found that plasma levels of 12- and 15-HETE, 12/15-lipoxygenase-produced proinflammatory lipids, and palmitoylethanolamide, an anti-inflammatory fatty acid amide hydrolase-regulated lipid metabolite, were differentially correlated with alcohol craving during abstinence, predicting higher craving independent of demographics, alcohol use history, and multiple therapeutic treatments. Conclusions Our findings highlight the promise of these lipid metabolites as biomarkers of heightened alcohol craving. The results open a novel opportunity for further research and clinical evaluation of these biomarkers to optimize existing treatments and develop new therapeutics for AUD.
Collapse
Affiliation(s)
- Cristina Miliano
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Luis A. Natividad
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas
| | - Susan Quello
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | | | - Ann M. Gregus
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Matthew W. Buczynski
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Barbara J. Mason
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| |
Collapse
|
6
|
Rahman MM, Jo YY, Kim YH, Park CK. Current insights and therapeutic strategies for targeting TRPV1 in neuropathic pain management. Life Sci 2024; 355:122954. [PMID: 39128820 DOI: 10.1016/j.lfs.2024.122954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/01/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Neuropathic pain, a common symptom of several disorders, exerts a substantial socioeconomic burden worldwide. Transient receptor potential vanilloid 1 (TRPV1), a non-selective cation channel predominantly ex-pressed in nociceptive neurons, plays a pivotal role in nociception, by detecting various endogenous and exogenous stimuli, including heat, pro-inflammatory mediators, and physical stressors. Dysregulation of TRPV1 signaling further contributes to the pathophysiology of neuropathic pain. Therefore, targeting TRPV1 is a promising strategy for developing novel analgesics with improved efficacy and safety profiles. Several pharmacological approaches to modulate TRPV1 activity, including agonists, antagonists, and biological TRPV1 RNA interference (RNAi, small interfering RNA [siRNA]) have been explored. Despite preclinical success, the clinical translation of TRPV1-targeted therapies has encountered challenges, including hyperthermia, hypothermia, pungency, and desensitization. Nevertheless, ongoing research efforts aim to refine TRPV1-targeted interventions through structural modifications, development of selective modulators, and discovery of natural, peptide-based drug candidates. Herein, we provide guidance for researchers and clinicians involved in the development of new interventions specifically targeting TRPV1 by reviewing the existing literature and highlighting current research activities. This study further discusses potential future research endeavors for enhancing the efficacy, safety, and tolerability of TRPV1 candidates, and thereby facilitates the translation of these discoveries into effective clinical interventions to alleviate neuropathic pain disorders.
Collapse
Affiliation(s)
- Md Mahbubur Rahman
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea
| | - Youn-Yi Jo
- Department of Anesthesiology and Pain Medicine, Gachon University, Gil Medical Center, Incheon 21565, Republic of Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea.
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon 21999, Republic of Korea.
| |
Collapse
|
7
|
Yin Y, Zhao P, Xu X, Zhou B, Chen J, Jiang X, Liu Y, Wu Y, Yue W, Xu H, Bu W. Piezoelectric Analgesia Blocks Cancer-Induced Bone Pain. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403979. [PMID: 39044708 DOI: 10.1002/adma.202403979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/23/2024] [Indexed: 07/25/2024]
Abstract
The manipulation of cell surface receptors' activity will open a new frontier for drug development and disease treatment. However, limited by the desensitization of drugs, effective physical intervention strategy remains challenging. Here, the controllable internalization of transient receptor potential vanilloid 1 (TRPV1) on neural cells by local piezoelectric field is reported. Single-cell-level local electric field is construct by synthesizing piezoelectric BiOIO3 nanosheets (BIONSs). Upon a mild ultrasound of 0.08 W cm-2, an electric field of 15.29 µV is generated on the surface of BIONSs, further inducing TRPV1 internalization in 5 min. The as-downregulated TRPV1 expression results in the reduction of Ca2+ signal in a spinal neuron and the inhibition of the activity of wide range dynamic neurons, therefore effectively preventing the transmission of cancer-induced bone pain (CIBP). This strategy not only charts a new course for CIBP alleviation, but also introduces a promising nanotechnology for regulating cell surface receptors, showing significant potential in neuropathological and receptor-related diseases.
Collapse
Affiliation(s)
- Yifei Yin
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Peiran Zhao
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xianyun Xu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330006, China
| | - Bangguo Zhou
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Jian Chen
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yelin Wu
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Wenwen Yue
- Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Shanghai, 200072, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200072, China
- Center of Minimally Invasive Treatment for Tumor, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| |
Collapse
|
8
|
Arcas JM, Oudaha K, González A, Fernández-Trillo J, Peralta FA, Castro-Marsal J, Poyraz S, Taberner F, Sala S, de la Peña E, Gomis A, Viana F. The ion channel TRPM8 is a direct target of the immunosuppressant rapamycin in primary sensory neurons. Br J Pharmacol 2024; 181:3192-3214. [PMID: 38741464 DOI: 10.1111/bph.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.
Collapse
Affiliation(s)
- José Miguel Arcas
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Khalid Oudaha
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Alejandro González
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Jorge Fernández-Trillo
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | | | - Júlia Castro-Marsal
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Seyma Poyraz
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Francisco Taberner
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Salvador Sala
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Elvira de la Peña
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Ana Gomis
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| | - Félix Viana
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
| |
Collapse
|
9
|
Zheng Y, Li S, Kang Y, Hu Q, Zheng Y, Wang X, Chi H, Guo K, Jiang M, Wei Z, Shao X, Xu C, Liu B, Du J, He X, Fang J, Lu Z, Jiang Y. Electroacupuncture Alleviates Streptozotocin-Induced Diabetic Neuropathic Pain via the TRPV1-Mediated CaMKII/CREB Pathway in Rats. J Mol Neurosci 2024; 74:79. [PMID: 39162890 DOI: 10.1007/s12031-024-02256-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Diabetic neuropathic pain (DNP) is a diabetic complication that causes severe pain and deeply impacts the quality of the sufferer's daily life. Currently, contemporary clinical treatments for DNP generally exhibit a deficiency in effectiveness. Electroacupuncture (EA) is recognized as a highly effective and safe treatment for DNP with few side effects. Regrettably, the processes via which EA alleviates DNP are still poorly characterized. Transient receptor potential vanilloid 1 (TRPV1) and phosphorylated calcium/calmodulin-dependent protein kinase II (p-CaMKII) are overexpressed on spinal cord dorsal horn (SCDH) in DNP rats, and co-localization is observed between them. Capsazepine, a TRPV1 antagonist, effectively reduced nociceptive hypersensitivity and downregulated the overexpression of phosphorylated CaMKIIα in rats with DNP. Conversely, the CaMKII inhibitor KN-93 did not have any impact on TRPV1. EA alleviated heightened sensitivity to pain caused by nociceptive stimuli and downregulated the level of TRPV1, p-CaMKIIα, and phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB) in DNP rats. Intrathecal injection of capsaicin, on the other hand, reversed the above effects of EA. These findings indicated that the CaMKII/CREB pathway on SCDH is located downstream of TRPV1 and is affected by TRPV1. EA alleviates DNP through the TRPV1-mediated CaMKII/CREB pathway.
Collapse
Affiliation(s)
- Yinmu Zheng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Siyi Li
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yurong Kang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qunqi Hu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Zheng
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoxiang Wang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hengyu Chi
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Keying Guo
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Minjian Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhouyuan Wei
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaomei Shao
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Xu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Boyu Liu
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junying Du
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaofen He
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | | | - Yongliang Jiang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, China.
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
10
|
Yuan Y, Zhang Q, Qiu F, Kang N, Zhang Q. Targeting TRPs in autophagy regulation and human diseases. Eur J Pharmacol 2024; 977:176681. [PMID: 38821165 DOI: 10.1016/j.ejphar.2024.176681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Transient receptor potential channels (TRPs) are widely recognized as a group of ion channels involved in various sensory perceptions, such as temperature, taste, pressure, and vision. While macroautophagy (hereafter referred to as autophagy) is primarily regulated by core machinery, the ion exchange mediated by TRPs between intracellular and extracellular compartments, as well as within organelles and the cytoplasm, plays a crucial role in autophagy regulation as an important signaling transduction mechanism. Moreover, certain TRPs can directly interact with autophagy regulatory proteins to participate in autophagy regulation. In this article, we provide an in-depth review of the current understanding of the regulatory mechanisms of autophagy, with a specific focus on TRPs. Furthermore, we highlight the potential prospects for drug development targeting TRPs in autophagy for the treatment of human diseases.
Collapse
Affiliation(s)
- Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| |
Collapse
|
11
|
Chahl LA. TRPV1 Channels in the Central Nervous System as Drug Targets. Pharmaceuticals (Basel) 2024; 17:756. [PMID: 38931423 PMCID: PMC11206835 DOI: 10.3390/ph17060756] [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: 03/30/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
TRPV1 channels are polymodal cation channels located predominantly on primary afferent neurons that are activated by inflammatory mediators, capsaicin (the active component in chili peppers), and noxious heat. TRPV1 channel antagonists are potential new analgesic agents, but their development has been hindered by the finding that they also produce loss of thermal homeostasis and response to noxious heat. Results from recent studies of the TRPV1 channel indicate that it might be possible to develop TRPV1 channel antagonists that inhibit pain without affecting noxious heat sensation. TRPV1 channels are also present in the central nervous system (CNS) and have been implicated in learning, memory, and behaviour. TRPV1 channel modulators have been proposed to have possible therapeutic potential in the treatment of neurological and psychiatric conditions. However, further understanding of the role of TRPV1 channels in the CNS is required before therapeutic advances in the treatment of neuropsychiatric conditions with TRPV1 channel modulators can be made.
Collapse
Affiliation(s)
- Loris A Chahl
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
12
|
Huang YZ, Ma JX, Bian YJ, Bai QR, Gao YH, Di SK, Lei YT, Yang H, Yang XN, Shao CY, Wang WH, Cao P, Li CZ, Zhu MX, Sun MY, Yu Y. TRPV1 analgesics disturb core body temperature via a biased allosteric mechanism involving conformations distinct from that for nociception. Neuron 2024; 112:1815-1831.e4. [PMID: 38492574 DOI: 10.1016/j.neuron.2024.02.016] [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: 09/11/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/18/2024]
Abstract
Efforts on developing transient receptor potential vanilloid 1 (TRPV1) drugs for pain management have been hampered by deleterious hypo- or hyperthermia caused by TRPV1 agonists/antagonists. Here, we compared the effects of four antagonists on TRPV1 polymodal gating and core body temperature (CBT) in Trpv1+/+, Trpv1-/-, and Trpv1T634A/T634A. Neither the effect on proton gating nor drug administration route, hair coverage, CBT rhythmic fluctuations, or inflammation had any influence on the differential actions of TRPV1 drugs on CBT. We identified the S4-S5 linker region exposed to the vanilloid pocket of TRPV1 to be critical for hyperthermia associated with certain TRPV1 antagonists. PSFL2874, a TRPV1 antagonist we discovered, is effective against inflammatory pain but devoid of binding to the S4-S5 linker and inducing CBT changes. These findings implicate that biased allosteric mechanisms exist for TRPV1 coupling to nociception and CBT regulation, opening avenues for the development of non-opioid analgesics without affecting CBT.
Collapse
Affiliation(s)
- Yi-Zhe Huang
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Xian Ma
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Jing Bian
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qin-Ru Bai
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yu-Hao Gao
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Shu-Ke Di
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yun-Tao Lei
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Yang
- Tea Research Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, Hunan, China
| | - Xiao-Na Yang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Chang-Yan Shao
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Wen-Hui Wang
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Cao
- Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chang-Zhu Li
- State Key Laboratory of Utilization of Woody Oil Resource, Hunan Academy of Forestry, Changsha 410004, Hunan, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Meng-Yang Sun
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Ye Yu
- Schools of Basic Medicine and Clinical Pharmacy and Traditional Chinese Pharmacy, and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
13
|
Cheng XL, Ruan YL, Dai JY, Fan HZ, Ling JY, Chen J, Lu WG, Gao XJ, Cao P. 8-shogaol derived from dietary ginger alleviated acute and inflammatory pain by targeting TRPV1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155500. [PMID: 38484627 DOI: 10.1016/j.phymed.2024.155500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/24/2024] [Accepted: 02/26/2024] [Indexed: 05/01/2024]
Abstract
Ginger, a well-known spice plant, has been used widely in medicinal preparations for pain relief. However, little is known about its analgesic components and the underlying mechanism. Here, we ascertained, the efficacy of ginger ingredient 8-Shogaol (8S), on inflammatory pain and tolerance induced by morphine, and probed the role of TRPV1 in its analgesic action using genetic and electrophysiology approaches. Results showed that 8S effectively reduced nociceptive behaviors of mice elicited by chemical stimuli, noxious heat as well as inflammation, and antagonized morphine analgesic tolerance independent on opioid receptor function. Genetic deletion of TRPV1 significantly abolished 8S' analgesia action. Further calcium imaging and patch-clamp recording showed that 8S could specifically activate TRPV1 in TRPV1-expressing HEK293T cells and dorsal root ganglion (DRG) neurons. The increase of [Ca2+]i in DRG was primarily mediated through TRPV1. Mutational and computation studies revealed the key binding sites for the interactions between 8S and TRPV1 included Leu515, Leu670, Ile573, Phe587, Tyr511, and Phe591. Further studies showed that TRPV1 activation evoked by 8S resulted in channel desensitization both in vitro and in vivo, as may be attributed to TRPV1 degradation or TRPV1 withdrawal from the cell surface. Collectively, this work provides the first evidence for the attractive analgesia of 8S in inflammatory pain and morphine analgesic tolerance mediated by targeting pain-sensing TRPV1 channel. 8S from dietary ginger has potential as a candidate drug for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Xiao-Lan Cheng
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yong-Lan Ruan
- Department of Neurology, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, 213003, China
| | - Jing-Ya Dai
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; Wanbei Health Vocational College, Suzhou, Anhui, 234000, China
| | - Hai-Zhen Fan
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Jin-Ying Ling
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Jiao Chen
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Wu-Guang Lu
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China
| | - Xue-Jiao Gao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China.
| | - Peng Cao
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Nanjing 210028, China; The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, 100 Minjiang Road, Quzhou, Zhejiang 324000, China.
| |
Collapse
|
14
|
Antoniazzi CTDD, Ruviaro NA, Peres DS, Rodrigues P, Viero FT, Trevisan G. Targeting TRPV4 Channels for Cancer Pain Relief. Cancers (Basel) 2024; 16:1703. [PMID: 38730655 PMCID: PMC11083562 DOI: 10.3390/cancers16091703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the unique and complex nature of cancer pain, the activation of different ion channels can be related to the initiation and maintenance of pain. The transient receptor potential vanilloid 4 (TRPV4) is a cation channel broadly expressed in sensory afferent neurons. This channel is activated by multiple stimuli to mediate pain perception associated with inflammatory and neuropathic pain. Here, we focused on summarizing the role of TRPV4 in cancer etiology and cancer-induced pain mechanisms. Many studies revealed that the administration of a TRPV4 antagonist and TRPV4 knockdown diminishes nociception in chemotherapy-induced peripheral neuropathy (CIPN). Although the evidence on TRPV4 channels' involvement in cancer pain is scarce, the expression of these receptors was reportedly enhanced in cancer-induced bone pain (CIBP), perineural, and orofacial cancer models following the inoculation of tumor cells to the bone marrow cavity, sciatic nerve, and tongue, respectively. Effective pain management is a continuous problem for patients diagnosed with cancer, and current guidelines fail to address a mechanism-based treatment. Therefore, examining new molecules with potential antinociceptive properties targeting TRPV4 modulation would be interesting. Identifying such agents could lead to the development of treatment strategies with improved pain-relieving effects and fewer adverse effects than the currently available analgesics.
Collapse
Affiliation(s)
- Caren Tatiane de David Antoniazzi
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Náthaly Andrighetto Ruviaro
- Graduate Program in Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil;
| | - Diulle Spat Peres
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Patrícia Rodrigues
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Fernanda Tibolla Viero
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Gabriela Trevisan
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
- Graduate Program in Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil;
| |
Collapse
|
15
|
Bekauri T, Fischer S, Honn KV, Maddipati KR, Love T, Little C, Wood RW, Bonham AD, Linder MA, Yule DI, Emanuelle C, Falsetta ML. Inflammation, lipid dysregulation, and transient receptor potential cation channel subfamily V member 4 signaling perpetuate chronic vulvar pain. Pain 2024; 165:820-837. [PMID: 37889581 PMCID: PMC10949218 DOI: 10.1097/j.pain.0000000000003088] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/19/2023] [Accepted: 08/23/2023] [Indexed: 10/29/2023]
Abstract
ABSTRACT Localized provoked vulvodynia is characterized by chronic vulvar pain that disrupts every aspect of the patient's life. Pain is localized to the vulvar vestibule, a specialized ring of tissue immediately surrounding the vaginal opening involved in immune defense. In this article, we show inflammation is the critical first step necessary for the generation of pain signals in the vulva. Inflammatory stimuli alone or combined with the transient receptor potential cation channel subfamily V member 4 (TRPV4) agonist 4α-phorbol 12,13-didecanoate stimulate calcium flux into vulvar fibroblast cells. Activity is blocked by the TRPV4 antagonist HC067047, denoting specificity to TRPV4. Using lipidomics, we found pro-resolving lipids in the vulvar vestibule were dysregulated, characterized by a reduction in pro-resolving mediators and heightened production of inflammatory mediators. We demonstrate specialized pro-resolving mediators represent a potential new therapy for vulvar pain, acting on 2 key parts of the disease mechanism by limiting inflammation and acutely inhibiting TRPV4 signaling.
Collapse
Affiliation(s)
- Tamari Bekauri
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Sarah Fischer
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Kenneth V. Honn
- Pathology Department, Wayne State University, Detroit, MI, United States
- Lipidomics Core Facility and Bioactive Lipids Research Program, Wayne State University, Detroit, MI, United States
| | - Krishna Rao Maddipati
- Pathology Department, Wayne State University, Detroit, MI, United States
- Lipidomics Core Facility and Bioactive Lipids Research Program, Wayne State University, Detroit, MI, United States
| | - Tanzy Love
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Chantelle Little
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Ronald W. Wood
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - Adrienne D. Bonham
- OB/GYN Department, Oregon Health Sciences University, Portland, OR, United States
| | - Mitchell A. Linder
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
| | - David I. Yule
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| | - Chrysilla Emanuelle
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| | - Megan L. Falsetta
- OB/GYN Research Division, University of Rochester, Rochester, NY, United States
- Pharmacology and Physiology Department, University of Rochester, Rochester, NY, United States
| |
Collapse
|
16
|
An Y, Hu J, Hao H, Zhao W, Zhang X, Shao J, Wang C, Li X, Liu C, He J, Zhao Y, Zhang H, Du X. The transmembrane channel-like 6 (TMC6) in primary sensory neurons involving thermal sensation via modulating M channels. Front Pharmacol 2024; 15:1330167. [PMID: 38440182 PMCID: PMC10909837 DOI: 10.3389/fphar.2024.1330167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction: The transmembrane channel-like (TMC) protein family contains eight members, TMC1-TMC8. Among these members, only TMC1 and TMC2 have been intensively studied. They are expressed in cochlear hair cells and are crucial for auditory sensations. TMC6 and TMC8 contribute to epidermodysplasia verruciformis, and predispose individuals to human papilloma virus. However, the impact of TMC on peripheral sensation pain has not been previously investigated. Methods: RNAscope was employed to detect the distribution of TMC6 mRNA in DRG neurons. Electrophysiological recordings were conducted to investigate the effects of TMC6 on neuronal characteristics and M channel activity. Zn2+ indicators were utilized to detect the zinc concentration in DRG tissues and dissociated neurons. A series of behavioural tests were performed to assess thermal and mechanical sensation in mice under both physiological and pathological conditions. Results and Discussion: We demonstrated that TMC6 is mainly expressed in small and medium dorsal root ganglion (DRG) neurons and is involved in peripheral heat nociception. Deletion of TMC6 in DRG neurons hyperpolarizes the resting membrane potential and inhibits neuronal excitability. Additionally, the function of the M channel is enhanced in TMC6 deletion DRG neurons owing to the increased quantity of free zinc in neurons. Indeed, heat and mechanical hyperalgesia in chronic pain are alleviated in TMC6 knockout mice, particularly in the case of heat hyperalgesia. This suggests that TMC6 in the small and medium DRG neurons may be a potential target for chronic pain treatment.
Collapse
Affiliation(s)
- Yating An
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jingyi Hu
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weixin Zhao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoxue Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jicheng Shao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Caixue Wang
- The Forth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinmeng Li
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chao Liu
- The Key Laboratory of Experimental Animal, Department of Animal Care, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinsha He
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiwen Zhao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, The Hebei Collaboration Innovation Center for Mechanism, Diagnosis and Treatment of Neurological and Psychiatric Disease, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
17
|
Szallasi A. Targeting TRPV1 for Cancer Pain Relief: Can It Work? Cancers (Basel) 2024; 16:648. [PMID: 38339399 PMCID: PMC11154559 DOI: 10.3390/cancers16030648] [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: 12/01/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Chronic intractable pain affects a large proportion of cancer patients, especially those with metastatic bone disease. Blocking sensory afferents for cancer pain relief represents an attractive alternative to opioids and other drugs acting in the CNS in that sensory nerve blockers are not addictive and do not affect the mental state of the patient. A distinct subpopulation of sensory afferents expresses the capsaicin receptor TRPV1. Intrathecal resiniferatoxin, an ultrapotent capsaicin analog, ablates TRPV1-expressing nerve endings exposed to the cerebrospinal fluid, resulting in permanent analgesia in women with cervical cancer metastasis to the pelvic bone. High-dose capsaicin patches are effective pain killers in patients with chemotherapy-induced peripheral neuropathic pain. However, large gaps remain in our knowledge since the mechanisms by which cancer activates TRPV1 are essentially unknown. Most important, it is not clear whether or not sensory denervation mediated by TRPV1 agonists affects cancer progression. In a murine model of breast cancer, capsaicin desensitization was reported to accelerate progression. By contrast, desensitization mediated by resiniferatoxin was found to block melanoma growth. These observations imply that TRPV1 blockade for pain relief may be indicated for some cancers and contraindicated for others. In this review, we explore the current state of this field and compare the analgesic potential of TRPV1 antagonism and sensory afferent desensitization in cancer patients.
Collapse
Affiliation(s)
- Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
18
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
19
|
Ye W, Lui ST, Zhao Q, Wong YM, Cheng A, Sung HHY, Williams ID, Qian PY, Huang P. Novel marine natural products as effective TRPV1 channel blockers. Int J Biol Macromol 2023; 253:127136. [PMID: 37776932 DOI: 10.1016/j.ijbiomac.2023.127136] [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: 07/18/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Chronic pain management poses a formidable challenge to healthcare, exacerbated by current analgesic options' limitations and adverse effects. Transient receptor potential vanilloid 1 (TRPV1), a non-selective cation channel, has emerged as a promising target for novel analgesics. However, safety and tolerability concerns have constrained the development of TRPV1 modulators. In this study, we explored marine-derived natural products as a source of potential TRPV1 modulators using high-throughput dye-uptake assays. We identified chrexanthomycins, a family of hexacyclic xanthones, exhibited potent TRPV1 inhibitory effects, with compounds cC and cF demonstrating the most significant activity. High-resolution patch-clamp assays confirmed the direct action of these compounds on the TRPV1 channel. Furthermore, in vivo assays revealed that cC and cF effectively suppressed capsaicin-induced pain sensation in mice, comparable to the known TRPV1 inhibitor, capsazepine. Structural-activity relationship analysis highlighted the importance of specific functional groups in modulating TRPV1 activity. Our findings underscore the therapeutic potential of chrexanthomycins and pave the way for further investigations into marine-derived TRPV1 modulators for pain management.
Collapse
Affiliation(s)
- Wenkang Ye
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China; SZU-HKUST Joint Ph.D. Program in Marine Environmental Science, Shenzhen University, Shenzhen 518060, China
| | - Sin Tung Lui
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Qirui Zhao
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yuk Ming Wong
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Aifang Cheng
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China
| | - Herman H-Y Sung
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ian D Williams
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Pei-Yuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Department of Ocean Science and Hong Kong Branch of Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Pingbo Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou 511458, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Department of Chemical and Biological Engineering, Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
20
|
Zhu K, Wang L, Liao T, Li W, Zhou J, You Y, Shi J. Progress in the development of TRPV1 small-molecule antagonists: Novel Strategies for pain management. Eur J Med Chem 2023; 261:115806. [PMID: 37713804 DOI: 10.1016/j.ejmech.2023.115806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) channels are widely distributed in sensory nerve endings, the central nervous system, and other tissues, functioning as ion channel proteins responsive to thermal pain and chemical stimuli. In recent years, the TRPV1 receptor has garnered significant interest as a potential therapeutic approach for various pain-related disorders, particularly TRPV1 antagonists. The present review offers a comprehensive, systematic exploration of both first- and second-generation TRPV1 antagonists in the context of pain management. Antagonists are categorized and explicated according to their structural characteristics. Detailed examination of binding modes, structural features, and pharmacological activities, alongside a critical appraisal of the advantages and limitations inherent to typical compounds within each structural category, are undertaken. Detailed discussions of the binding modes, structural features, pharmacological activities, advantages, and limitations of typical compounds within each structural category offer valuable insights and guidance for the future research and development of safer, more effective, and more targeted TRPV1 antagonists.
Collapse
Affiliation(s)
- Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lin Wang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - TingTing Liao
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yaodong You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
21
|
Shehab S, Javed H, Johnson AM, Tariq S, Kumar CA, Emerald BS. Unveiling the mechanisms of neuropathic pain suppression: perineural resiniferatoxin targets Trpv1 and beyond. Front Neuroanat 2023; 17:1306180. [PMID: 38099210 PMCID: PMC10720729 DOI: 10.3389/fnana.2023.1306180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
Neuropathic pain arises from damage or disorders affecting the somatosensory system. In rats, L5 nerve injury induces thermal and mechanical hypersensitivity/hyperalgesia. Recently, we demonstrated that applying resiniferatoxin (RTX) directly on uninjured L3 and L4 nerves alleviated thermal and mechanical hypersensitivity resulting from L5 nerve injury. Herein, using immunohistochemistry, Western blot, and qRT-PCR techniques, we reveal that perineural application of RTX (0.002%) on the L4 nerve substantially downregulated the expression of its receptor (Trpv1) and three different voltage-gated ion channels (Nav1.9, Kv4.3, and Cav2.2). These channels are found primarily in small-sized neurons and show significant colocalization with Trpv1 in the dorsal root ganglion (DRG). However, RTX treatment did not affect the expression of Kv1.1, Piezo2 (found in large-sized neurons without colocalization with Trpv1), and Kir4.1 (localized in satellite cells) in the ipsilateral DRGs. Furthermore, RTX application on L3 and L4 nerves reduced the activation of c-fos in the spinal neurons induced by heat stimulation. Subsequently, we investigated whether applying RTX to the L3 and L4 nerves 3 weeks before the L5 nerve injury could prevent the onset of neuropathic pain. Both 0.002 and 0.004% concentrations of RTX produced significant analgesic effects, while complete prevention of thermal and mechanical hypersensitivity required a concentration of 0.008%. Importantly, this preventive effect on neuropathic manifestations was not associated with nerve degeneration, as microscopic examination revealed no morphological changes. Overall, this study underscores the mechanisms and the significance of perineural RTX treatment applied to adjacent uninjured nerves in entirely preventing nerve injury-induced neuropathic pain in humans and animals.
Collapse
Affiliation(s)
- Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | | | | | | | | |
Collapse
|
22
|
Zhang RY, Zhu BF, Zhao JG, Zhao L, Wang LK. Electroacupuncture Stimulation Alleviates Inflammatory Pain in Male Rats by Suppressing Oxidative Stress. Physiol Res 2023; 72:657-667. [PMID: 38015764 PMCID: PMC10751055 DOI: 10.33549/physiolres.934965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/13/2023] [Indexed: 01/05/2024] Open
Abstract
In the present study, we focused on whether the analgesic effect of Electroacupuncture (EA) is related to the regulation of oxidative stress. We established a chronic inflammatory pain model in male rats by a single injection of complete Freund's adjuvant (CFA) and then treated the animals with daily EA stimulation at the site of "zusanli". The analgesic effect of EA was evaluated by measuring the paw withdrawal threshold (PWT) when rats received mechanical and thermal pain stimulation. The levels of inflammation-related molecules and oxidative stress-related markers in the spinal cord were measured by western blotting or ELISA kits. EA stimulation and antioxidants effectively increased the PWT in CFA rats. Co-treatment of CFA rats with the ROS donor t-butyl hydroperoxide (t-BOOH) further decreased the PWT and weakened the analgesic effect of EA. EA treatment inhibited inflammation and oxidative stress, as shown by decreased levels of tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), IL-6, and MDA and increased activity of SOD and catalase. Moreover, EA reduced the expression of p-p38, p-ERK, and p-p65 and simultaneously downregulated the expression of TRPV1 and TRPV4 in CFA rats. In an in vitro study, direct stimulation with t-BOOH to the C6 cells increased the production of TNF-alpha, IL-1beta, IL-6, activated p38, ERK, and p65 and up-regulated the expression of TRPV1 and TRPV4, and these effects could be prevented by the ROS scavenger PBN. Taken together, our data indicate that the inhibition of oxidative stress and the generation of ROS contribute to the analgesic effect of EA in male CFA rats.
Collapse
Affiliation(s)
- R Y Zhang
- Department of Pain Management, Anhui Medical University, Hospital of Hefei, Hefei, People's Republic of China.
| | | | | | | | | |
Collapse
|
23
|
Adhya P, Vaidya B, Sharma SS. BTD: A TRPC5 activator ameliorates mechanical allodynia in diabetic peripheral neuropathic rats by modulating TRPC5-CAMKII-ERK pathway. Neurochem Int 2023; 170:105609. [PMID: 37673218 DOI: 10.1016/j.neuint.2023.105609] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/02/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Mechanical allodynia is a serious complication of painful diabetic neuropathy (PDN) with limited treatment options. The transient receptor potential canonical 5 (TRPC5) channel is a promising target in pain; however, its role in painful diabetic neuropathy has not yet been elucidated. In this study, we have investigated the role of TRPC5 channels using BTD [N-{3-(adamantan-2-yloxy)-propyl}-3-(6-methyl-1,1-dioxo-2H-1λ6,2,4-benzothiadiazin-3-yl)-propanamide)],a potent TRPC5 activator and HC070, as TRPC5 channel inhibitor in rat model of PDN. In this study, streptozotocin was used to induce diabetes in male Sprague-Dawley rats. The alterations in mechanical and thermal pain thresholds, nerve functional deficits in diabetic animals were assessed by various behavioral and functional parameters.TRPC5 involvement was investigated by treating neuropathic rats with BTD, TRPC5 channel activator (1 and 3 mg/kg, i.p. for 14 days) and HC070, a TRPC5 channel inhibitor (1 and 3 mg/kg). BTD and HC070 effects in pain reduction were assessed by western blotting, estimating oxidative stress and inflammatory markers in the lumbar spinal cord. BTD treatment (3 mg/kg, i.p.) once daily for 14 days ameliorated mechanical allodynia but not thermal hyposensation or nerve functional deficit in diabetic neuropathic rats. BTD treatment down-regulated TRPC5 expression by increasing the activity of protein kinase C. It also subsequently down-regulated the downstream pain markers (CAMKII, ERK) in the spinal cord. Additionally, a decrease in inflammatory cytokines (TNF-α, IL-6) also demonstrated BTD's potent anti-inflammatory properties in reducing mechanical allodynia. On the other hand, HC070 did not exert any beneficial effects on behavioural and nerve functional parameters. The study concludes that BTD ameliorated mechanical allodynia in a rat model of painful diabetic neuropathy not only through modulation of the TRPC5-CAMKII-ERK pathway but also through its anti-inflammatory and anti-apoptotic properties. Overall, BTD is a promising therapeutic molecule in the treatment of mechanical allodynia in painful diabetic neuropathy.
Collapse
Affiliation(s)
- Pratik Adhya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), 160 062, Punjab, India
| | - Bhupesh Vaidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), 160 062, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar (Mohali), 160 062, Punjab, India.
| |
Collapse
|
24
|
Zhu C, Yang Y, Song Y, Guo J, Yu G, Tang J, Tang Z. Mechanisms involved in the antinociceptive and anti-inflammatory effects of xanthotoxin. Eur J Neurosci 2023; 58:3605-3617. [PMID: 37671643 DOI: 10.1111/ejn.16119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/04/2023] [Accepted: 07/26/2023] [Indexed: 09/07/2023]
Abstract
Xanthotoxin (XAT) is a natural furanocoumarin clinically used in the treatment of skin diseases such as vitiligo and psoriasis. Recent studies have also investigated its effects on anti-inflammatory, anti-cognitive dysfunction, and anti-amnesia as a guideline for clinic application. However, little is known about its effects on pain relief. Here, we tested the analgesic effects of XAT in serious acute pain and chronic pain models. For acute pain, we used hot-, capsaicin- and formalin-induced paw licking. Nociceptive threshold was measured by mechanical stimuli with von Frey filaments. For chronic pain, we injected complete Freund's adjuvant (CFA) into the mice's plantar surface of the hind paw to induce inflammatory pain. Heat and mechanical hyperalgesia were evaluated by radiant heat and von Frey filament tests, respectively. To investigate the mechanisms underlying the analgesic effect of XAT, we used calcium imaging and western blot to assess transient receptor potential vanilloid 1 (TRPV1) activity and expression in isolated L4-L6 dorsal root ganglion (DRG) neurons. Haematoxylin and eosin (HE) staining, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA) were used to examine immune cell recruitment and proinflammatory factor release from skin tissue from paw injection sites. Our results demonstrated that XAT not only reduced acute pain behaviors generated by hot, capsaicin, and formalin but also attenuated CFA-induced heat and mechanical hyperalgesia. The analgesic activity of XAT may be achieved by controlling peripheral inflammation, lowering immune cell infiltration at the site of inflammatory tissue, reducing inflammatory factor production, and therefore inhibiting TRPV1 channel sensitization and expression.
Collapse
Affiliation(s)
- Chan Zhu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yan Yang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yizhi Song
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Guo
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Guang Yu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Juanjuan Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zongxiang Tang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory of Chinese Medicine for Prevention and Treatment of Neurological Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Landrum O, Marcondes L, Egharevba T, Gritsenko K. Painful diabetic peripheral neuropathy of the feet: integrating prescription-strength capsaicin into office procedures. Pain Manag 2023; 13:613-626. [PMID: 37750226 DOI: 10.2217/pmt-2023-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
Prescription-strength (8%) capsaicin topical system is a US FDA-approved treatment for painful diabetic peripheral neuropathy of the feet. A 30 min application of the capsaicin 8% topical system can provide sustained (up to 3 months) local pain relief by desensitizing and reducing TRPV1-expressing cutaneous fibers. Capsaicin is not absorbed systemically; despite associated application-site discomfort, capsaicin 8% topical system is well tolerated, with no known drug interactions or contraindications, and could offer clinical advantages over oral options. Capsaicin 8% topical system are not for patient self-administration and require incorporation into office procedures, with the added benefit of treatment compliance. This article reviews existing literature and provides comprehensive, practical information regarding the integration of capsaicin 8% topical systems into office procedures.
Collapse
Affiliation(s)
- Orlando Landrum
- Regenerative Medicine & Interventional Pain Specialist, Cutting Edge Integrative Pain Centers, 3060 Windsor Cir, Elkhart, IN 46514, USA
| | - Lizandra Marcondes
- Averitas Pharma, Inc., Morristown, 360 Mt Kemble Ave, Morristown, NJ 07960, USA
| | - Toni Egharevba
- Averitas Pharma, Inc., Morristown, 360 Mt Kemble Ave, Morristown, NJ 07960, USA
| | - Karina Gritsenko
- Montefiore Medical Center, New 111 E 210th St, Bronx, NY 10467, USA
| |
Collapse
|
26
|
Crichton M, Marshall S, Marx W, Isenring E, Lohning A. Therapeutic health effects of ginger (Zingiber officinale): updated narrative review exploring the mechanisms of action. Nutr Rev 2023; 81:1213-1224. [PMID: 36688554 DOI: 10.1093/nutrit/nuac115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Ginger (Zingiber officinale) has been investigated for its potentially therapeutic effect on a range of chronic conditions and symptoms in humans. However, a simplified and easily understandable examination of the mechanisms behind these effects is lacking and, in turn, hinders interpretation and translation to practice, and contributes to overall clinical heterogeneity confounding the results. Therefore, drawing on data from nonhuman trials, the objective for this narrative review was to comprehensively describe the current knowledge on the proposed mechanisms of action of ginger on conferring therapeutic health effects in humans. Mechanistic studies support the findings from human clinical trials that ginger may assist in improving symptoms and biomarkers of pain, metabolic chronic disease, and gastrointestinal conditions. Bioactive ginger compounds reduce inflammation, which contributes to pain; promote vasodilation, which lowers blood pressure; obstruct cholesterol production, which regulates blood lipid profile; translocate glucose transporter type 4 molecules to plasma membranes to assist in glycemic control; stimulate fatty acid breakdown to aid weight management; and inhibit serotonin, muscarinic, and histaminergic receptor activation to reduce nausea and vomiting. Additional human trials are required to confirm the antimicrobial, neuroprotective, antineoplastic, and liver- and kidney-protecting effects of ginger. Interpretation of the mechanisms of action will help clinicians and researchers better understand how and for whom ginger may render therapeutic effects and highlight priority areas for future research.
Collapse
Affiliation(s)
- Megan Crichton
- Faculty of Health Science & Medicine, Bond University Nutrition and Dietetics Research Group, Bond University, Robina, Queensland, Australia
- Cancer and Palliative Care Outcomes Centre, Centre for Healthcare Transformation, School of Nursing, Faculty of Health, Kelvin Grove, Queensland, Australia
| | - Skye Marshall
- Faculty of Health Science & Medicine, Bond University Nutrition and Dietetics Research Group, Bond University, Robina, Queensland, Australia
- Department of Science, Nutrition Research Australia, Sydney, New South Wales, Australia
| | - Wolfgang Marx
- Faculty of Health Science & Medicine, Bond University Nutrition and Dietetics Research Group, Bond University, Robina, Queensland, Australia
- Impact (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, Deakin University, Geelong, Australia
| | - Elizabeth Isenring
- Faculty of Health Science & Medicine, Bond University Nutrition and Dietetics Research Group, Bond University, Robina, Queensland, Australia
| | - Anna Lohning
- Faculty of Health Science & Medicine, Bond University Nutrition and Dietetics Research Group, Bond University, Robina, Queensland, Australia
| |
Collapse
|
27
|
Liu Y, Lyu Y, Zhu L, Wang H. Role of TRP Channels in Liver-Related Diseases. Int J Mol Sci 2023; 24:12509. [PMID: 37569884 PMCID: PMC10420300 DOI: 10.3390/ijms241512509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The liver plays a crucial role in preserving the homeostasis of an entire organism by metabolizing both endogenous and exogenous substances, a process that relies on the harmonious interactions of hepatocytes, hepatic stellate cells (HSCs), Kupffer cells (KCs), and vascular endothelial cells (ECs). The disruption of the liver's normal structure and function by diverse pathogenic factors imposes a significant healthcare burden. At present, most of the treatments for liver disease are palliative in nature, rather than curative or restorative. Transient receptor potential (TRP) channels, which are extensively expressed in the liver, play a crucial role in regulating intracellular cation concentration and serve as the origin or intermediary stage of certain signaling pathways that contribute to liver diseases. This review provides an overview of recent developments in liver disease research, as well as an examination of the expression and function of TRP channels in various liver cell types. Furthermore, we elucidate the molecular mechanism by which TRP channels mediate liver injury, liver fibrosis, and hepatocellular carcinoma (HCC). Ultimately, the present discourse delves into the current state of research and extant issues pertaining to the targeting of TRP channels in the treatment of liver diseases and other ailments. Despite the numerous obstacles encountered, TRP channels persist as an extremely important target for forthcoming clinical interventions aimed at treating liver diseases.
Collapse
Affiliation(s)
- Yusheng Liu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| | - Yihan Lyu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| | - Lijuan Zhu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China; (Y.L.); (Y.L.)
| |
Collapse
|
28
|
Falsetta ML, Maddipati KR, Honn KV. Inflammation, lipids, and pain in vulvar disease. Pharmacol Ther 2023; 248:108467. [PMID: 37285943 PMCID: PMC10527276 DOI: 10.1016/j.pharmthera.2023.108467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Localized provoked vulvodynia (LPV) affects ∼14 million people in the US (9% of women), destroying lives and relationships. LPV is characterized by chronic pain (>3 months) upon touch to the vulvar vestibule, which surrounds the vaginal opening. Many patients go months or years without a diagnosis. Once diagnosed, the treatments available only manage the symptoms of disease and do not correct the underlying problem. We have focused on elucidating the underlying mechanisms of chronic vulvar pain to speed diagnosis and improve intervention and management. We determined the inflammatory response to microorganisms, even members of the resident microflora, sets off a chain of events that culminates in chronic pain. This agrees with findings from several other groups, which show inflammation is altered in the painful vestibule. The vestibule of patients is acutely sensitive to inflammatory stimuli to the point of being deleterious. Rather than protect against vaginal infection, it causes heightened inflammation that does not resolve, which coincides with alterations in lipid metabolism that favor production of proinflammatory lipids and not pro-resolving lipids. Lipid dysbiosis in turn triggers pain signaling through the transient receptor potential vanilloid subtype 4 receptor (TRPV4). Treatment with specialized pro-resolving mediators (SPMs) that foster resolution reduces inflammation in fibroblasts and mice and vulvar sensitivity in mice. SPMs, specifically maresin 1, act on more than one part of the vulvodynia mechanism by limiting inflammation and acutely inhibiting TRPV4 signaling. Therefore, SPMs or other agents that target inflammation and/or TRPV4 signaling could prove effective as new vulvodynia therapies.
Collapse
Affiliation(s)
- Megan L Falsetta
- University of Rochester, OB/GYN Research Division, Rochester, NY, United States of America; University of Rochester, Pharmacology and Physiology Department, Rochester, NY, United States of America.
| | - Krishna Rao Maddipati
- Wayne State University, Pathology Department, Detroit, MI, United States of America; Wayne State University, Lipidomics Core Facility and Bioactive Lipids Research Program, Detroit, MI, United States of America
| | - Kenneth V Honn
- Wayne State University, Pathology Department, Detroit, MI, United States of America; Wayne State University, Lipidomics Core Facility and Bioactive Lipids Research Program, Detroit, MI, United States of America
| |
Collapse
|
29
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, 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
|
30
|
Erin N, Szallasi A. Carcinogenesis and Metastasis: Focus on TRPV1-Positive Neurons and Immune Cells. Biomolecules 2023; 13:983. [PMID: 37371563 DOI: 10.3390/biom13060983] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Both sensory neurons and immune cells, albeit at markedly different levels, express the vanilloid (capsaicin) receptor, Transient Receptor Potential, Vanilloid-1 (TRPV1). Activation of TRPV1 channels in sensory afferent nerve fibers induces local effector functions by releasing neuropeptides (most notably, substance P) which, in turn, trigger neurogenic inflammation. There is good evidence that chronic activation or inactivation of this inflammatory pathway can modify tumor growth and metastasis. TRPV1 expression was also demonstrated in a variety of mammalian immune cells, including lymphocytes, dendritic cells, macrophages and neutrophils. Therefore, the effects of TRPV1 agonists and antagonists may vary depending on the prominent cell type(s) activated and/or inhibited. Therefore, a comprehensive understanding of TRPV1 activity on immune cells and nerve endings in distinct locations is necessary to predict the outcome of therapies targeting TRPV1 channels. Here, we review the neuro-immune modulation of cancer growth and metastasis, with focus on the consequences of TRPV1 activation in nerve fibers and immune cells. Lastly, the potential use of TRPV1 modulators in cancer therapy is discussed.
Collapse
Affiliation(s)
- Nuray Erin
- Department of Medical Pharmacology, School of Medicine, Akdeniz University, Antalya 07070, Turkey
- Immuno-Pharmacology and Immuno-Oncology Unit, School of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| |
Collapse
|
31
|
Zholos AV, Dryn DO, Melnyk MI. General anaesthesia-related complications of gut motility with a focus on cholinergic mechanisms, TRP channels and visceral pain. Front Physiol 2023; 14:1174655. [PMID: 37275228 PMCID: PMC10232893 DOI: 10.3389/fphys.2023.1174655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
General anesthesia produces multiple side effects. Notably, it temporarily impairs gastrointestinal motility following surgery and causes the so-called postoperative ileus (POI), a multifactorial and complex condition that develops secondary to neuromuscular failure and mainly affects the small intestine. There are currently limited medication options for POI, reflecting a lack of comprehensive understanding of the mechanisms involved in this complex condition. Notably, although acetylcholine is one of the major neurotransmitters initiating excitation-contraction coupling in the gut, cholinergic stimulation by prokinetic drugs is not very efficient in case of POI. Acetylcholine when released from excitatory motoneurones of the enteric nervous system binds to and activates M2 and M3 types of muscarinic receptors in smooth muscle myocytes. Downstream of these G protein-coupled receptors, muscarinic cation TRPC4 channels act as the major focal point of receptor-mediated signal integration, causing membrane depolarisation accompanied by action potential discharge and calcium influx via L-type Ca2+ channels for myocyte contraction. We have recently found that both inhalation (isoflurane) and intravenous (ketamine) anesthetics significantly inhibit this muscarinic cation current (termed mI CAT) in ileal myocytes, even when G proteins are activated directly by intracellular GTPγS, i.e., bypassing muscarinic receptors. Here we aim to summarize Transient Receptor Potential channels and calcium signalling-related aspects of the cholinergic mechanisms in the gut and visceral pain, discuss exactly how these may be negatively impacted by general anaesthetics, while proposing the receptor-operated TRPC4 channel as a novel molecular target for the treatment of POI.
Collapse
Affiliation(s)
- Alexander V. Zholos
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Dariia O. Dryn
- O.O. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Mariia I. Melnyk
- ESC “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
- O.O. Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
32
|
Barthó L, Szallasi A. János Szolcsányi and capsaicin research - from the early years to his legacy. Temperature (Austin) 2023; 10:3-8. [PMID: 37187835 PMCID: PMC10177740 DOI: 10.1080/23328940.2023.2179820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Affiliation(s)
- Lóránd Barthó
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Pécs, Hungary,
| | - Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
33
|
He S, Zambelli VO, Sinharoy P, Brabenec L, Bian Y, Rwere F, Hell RC, Stein Neto B, Hung B, Yu X, Zhao M, Luo Z, Wu C, Xu L, Svensson KJ, McAllister SL, Stary CM, Wagner NM, Zhang Y, Gross ER. A human TRPV1 genetic variant within the channel gating domain regulates pain sensitivity in rodents. J Clin Invest 2023; 133:163735. [PMID: 36472910 PMCID: PMC9888391 DOI: 10.1172/jci163735] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Pain signals are relayed to the brain via a nociceptive system, and in rare cases, this nociceptive system contains genetic variants that can limit the pain response. Here, we questioned whether a human transient receptor potential vanilloid 1 (TRPV1) missense variant causes a resistance to noxious stimuli and, further, whether we could target this region with a cell-permeable peptide as a pain therapeutic. Initially using a computational approach, we identified a human K710N TRPV1 missense variant in an otherwise highly conserved region of mammalian TRPV1. After generating a TRPV1K710N-knockin mouse using CRISPR/Cas9, we discovered that the K710N variant reduced capsaicin-induced calcium influx in dorsal root ganglion neurons. The TRPV1K710N rodents also had less acute behavioral responses to noxious chemical stimuli and less hypersensitivity to nerve injury, while their response to noxious heat remained intact. Furthermore, blocking this K710 region in WT rodents using a cell-penetrating peptide limited acute behavioral responses to noxious stimuli and returned pain hypersensitivity induced by nerve injury to baseline levels. These findings identify K710 TRPV1 as a discrete site that is crucial for the control of nociception and provide insights into how to leverage rare genetic variants in humans to uncover fresh strategies for developing pain therapeutics.
Collapse
Affiliation(s)
- Shufang He
- Department of Anesthesiology and Perioperative Medicine, the Second Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China.,Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Vanessa O. Zambelli
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA.,Laboratory of Pain and Signaling, Butantan Institute, Sāo Paulo, Brazil
| | - Pritam Sinharoy
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Laura Brabenec
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Yang Bian
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Freeborn Rwere
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Rafaela C.R. Hell
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Beatriz Stein Neto
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA.,Laboratory of Pain and Signaling, Butantan Institute, Sāo Paulo, Brazil
| | - Barbara Hung
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Xuan Yu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Meng Zhao
- Department of Pathology, ,Stanford Diabetes Research Center, and,Stanford Cardiovascular Institute, School of Medicine, Stanford University, Stanford, California, USA
| | - Zhaofei Luo
- Department of Anesthesiology and Perioperative Medicine, the Second Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Chao Wu
- Department of Anesthesiology and Perioperative Medicine, the Second Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Lijun Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Katrin J. Svensson
- Department of Pathology, ,Stanford Diabetes Research Center, and,Stanford Cardiovascular Institute, School of Medicine, Stanford University, Stanford, California, USA
| | - Stacy L. McAllister
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA.,Department of Obstetrics and Gynecology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Creed M. Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA
| | - Nana-Maria Wagner
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, the Second Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
| | - Eric R. Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA.,Stanford Diabetes Research Center, and,Stanford Cardiovascular Institute, School of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
34
|
Gao M, Zhang Y, Wang B, Guo N, Shao L, Zhai W, Jiang L, Wang Q, Qian H, Yan L. Novel dual-target μ‑opioid and TRPV1 ligands as potential pharmacotherapeutics for pain management. Bioorg Chem 2023; 131:106335. [PMID: 36603243 DOI: 10.1016/j.bioorg.2022.106335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Currently, the development of effective analgesic drugs with few side effects remains a great challenge. Studies have suggested that multi-target drug treatments show high efficacy and reduced side effects compared to single-target drug therapies. In this work, we designed and synthesized two series of novel MOR/TRPV1 dual active ligands in which the phenylpiperidine group or the N-phenyl-N-(piperidin-4-yl) propionamide group as the MOR pharmacophore was fused to the benzylpiperazinyl urea-based TRPV1 pharmacophore. In particular, compound 5a exhibited promising dual pharmacological activity for MOR (EC50 = 53.7 nM) and TRPV1 (IC50 = 32.9 nM) in vitro. In formalin tests, compound 5a showed potent, dose-dependent in vivo analgesic activity in both the 1st and 2nd phases. Gratifyingly, compound 5a did not cause the side effects of hyperthermia and analgesic tolerance. Consistent with its in vitro activity, compound 5a also simultaneously agonized MOR and antagonized TRPV1 in vivo. Further studies on compound 5a showed acceptable pharmacokinetic properties and brain permeability. Furthermore, molecular docking studies showed that compound 5a tightly bound to the active pockets of hMOR and hTRPV1, respectively. Overall, this work shows the promise in discovering new analgesic treatments through the strategy of simultaneously targeting MOR and TRPV1 with a single molecule.
Collapse
Affiliation(s)
- Mengkang Gao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China; State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Yang Zhang
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China; Department of Life Sciences, Changzhi University, 73 East Chengbei Street, Changzhi, Shanxi 046011, China
| | - Bingxin Wang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Ning Guo
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Lulian Shao
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Weibin Zhai
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Lei Jiang
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China
| | - Qiang Wang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, 182 Minyuan road, Wuhan, Hubei 430074, China
| | - Hai Qian
- State Key Laboratory of Natural Medicines, Center of Drug Discovery, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China.
| | - Lin Yan
- School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, China.
| |
Collapse
|
35
|
Zhang J, Zhou J, Yu Y, Cai Y, Li Z, Lu Y, Zhao J. Sesamin Induces the Transdifferentiation of Type II Alveolar Epithelial Cells via AnnexinA1 and TRPV1. Lung 2023; 201:65-77. [PMID: 36735045 DOI: 10.1007/s00408-023-00598-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023]
Abstract
PURPOSE Acute lung injury (ALI) with high rates of morbidity is often accompanied by the apoptosis in the type I alveolar epithelial cells (ATIs). Thus, the transdifferentiation of type II alveolar epithelial cells (ATIIs) into ATIs is crucial for the maintenance of alveolar epithelial functions. We aimed to elucidate the role of sesamin in the transdifferentiation of ATIIs to ATIs and the involvement of the TRPV1/AKT pathway. METHODS In vivo, the mouse model of ALI was simulated by intraperitoneal and intratracheal injections of lipopolysaccharide (LPS), respectively. The protective effects of sesamin on ALI were investigated using the survival rate, lung/body weight ratio, histological analysis of lung with HE staining, and mRNA levels of inflammatory factors. Western blot analysis and immunofluorescence detection of ATIs marker AQP5 were used to evaluate the protective effect of sesamin on ATIs. Western blot, EdU, and qPCR analyses were applied to detect changes in apoptosis, proliferation, and transdifferentiation markers of ATII A549 cell lines. Small interfering RNA (siRNA) was used to detect the involvement and relationships between the sesamin receptors (ANXA1 and TRPV1) and the AKT pathway in transdifferentiation. RESULTS Sesamin (200 mg/kg) significantly improved LPS-induced ALI and inhibited LPS-induced ATIs reduction. A low concentration of sesamin (20 μM) promoted the transdifferentiation of ATIIs to ATIs. Both ANXA1 and TRPV1 were involved in sesamin-promoted transdifferentiation, while the P-AKT (S473) level was down-regulated by TRPV1 siRNA. CONCLUSION Sesamin may promote transdifferentiation of ATII to ATI to ultimately rescue ALI, with TRPV1/AKT pathway involved in this transdifferentiation. This study revealed a novel role of sesamin in promoting the transdifferentiation of ATIIs to ATIs, providing experimental supports for the potential targets of ALI therapy.
Collapse
Affiliation(s)
- Jiaojiao Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Jinrun Zhou
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Yifan Yu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Yuqing Cai
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Zhiliang Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Yao Lu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China
| | - Jing Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, 266237, People's Republic of China.
| |
Collapse
|
36
|
Katz B, Zaguri R, Edvardson S, Maayan C, Elpeleg O, Lev S, Davidson E, Peters M, Kfir-Erenfeld S, Berger E, Ghazalin S, Binshtok AM, Minke B. Nociception and pain in humans lacking a functional TRPV1 channel. J Clin Invest 2023; 133:153558. [PMID: 36454632 PMCID: PMC9888381 DOI: 10.1172/jci153558] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUNDChronic pain is a debilitating illness with currently limited therapy, in part due to difficulties in translating treatments derived from animal models to patients. The transient receptor potential vanilloid 1 (TRPV1) channel is associated with noxious heat detection and inflammatory pain, and reports of adverse effects in human trials have hindered extensive efforts in the clinical development of TRPV1 antagonists as novel pain relievers.METHODSWe examined 2 affected individuals (A1 and A2) carrying a homozygous missense mutation in TRPV1, rendering the channel nonfunctional. Biochemical and functional assays were used to analyze the mutant channel. To identify possible phenotypes of the affected individuals, we performed psychophysical and medical examinations.RESULTSWe demonstrated that diverse TRPV1 activators, acting at different sites of the channel protein, were unable to open the cloned mutant channel. This finding was not a consequence of impairment in the expression, cellular trafficking, or assembly of protein subunits. The affected individuals were insensitive to application of capsaicin to the mouth and skin and did not demonstrate aversive behavior toward capsaicin. Furthermore, quantitative sensory testing of A1 revealed an elevated heat-pain threshold but also, surprisingly, an elevated cold-pain threshold and extensive neurogenic inflammatory, flare, and pain responses following application of the TRPA1 channel activator mustard oil.CONCLUSIONOur study provides direct evidence in humans for pain-related functional changes linked to TRPV1, which is a prime target in the development of pain relievers.FUNDINGSupported by the Israel Science Foundation (368/19); Teva's National Network of Excellence in Neuroscience grant (no. 0394886) and Teva's National Network of Excellence in Neuroscience postdoctoral fellowship.
Collapse
Affiliation(s)
- Ben Katz
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Rachel Zaguri
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Simon Edvardson
- Pediatric Neurology Unit, Pediatric Department, Hadassah University Hospital, Mount Scopus, Jerusalem, Israel
| | - Channa Maayan
- Pediatric Neurology Unit, Pediatric Department, Hadassah University Hospital, Mount Scopus, Jerusalem, Israel
| | | | - Shaya Lev
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Elyad Davidson
- Pain Relief Unit, Department of Anesthesiology and Critical Care Medicine, and
| | - Maximilian Peters
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Shlomit Kfir-Erenfeld
- Department of Bone Marrow Transplantation and Cancer Immunology, Hadassah University Hospital, Ein Kerem, Jerusalem, Israel
| | - Esther Berger
- Department of Pathology, E. Wolfson Medical Center, Holon, Israel
| | - Shifa Ghazalin
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Alexander M. Binshtok
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| | - Baruch Minke
- Department of Medical Neurobiology, Faculty of Medicine and the Edmond and Lily Safra Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem, Israel
| |
Collapse
|
37
|
Chen W, Xu Q, Ma X, Mo J, Lin G, He G, Chu Z, Li J. Synthesis and biological evaluation of N-(benzene sulfonyl)acetamide derivatives as anti-inflammatory and analgesic agents with COX-2/5-LOX/TRPV1 multifunctional inhibitory activity. Bioorg Med Chem Lett 2023; 80:129101. [PMID: 36481449 DOI: 10.1016/j.bmcl.2022.129101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
In this study, a series of structurally novel N-(benzene sulfonyl) acetamide derivatives were designed, synthesized, and biologically evaluated as COX-2/5-LOX/TRPV1 multitarget inhibitors for anti-inflammatory and analgesic therapy. Among them, 9a and 9b displayed favorable COX-2 (9a IC50 = 0.011 μM, 9b IC50 = 0.023 μM), 5-LOX (9a IC50 = 0.046 μM, 9b IC50 = 0.31 μM) and TRPV1 (9a IC50 = 0.008 μM, 9b IC50 = 0.14 μM) inhibitory activities. The pharmacokinetic (PK) study of 9a in SD rats at the dosage of 10 mg/kg demonstrated a high oral exposure, an acceptable clearance and a favorable bioavailability (Cmax = 5807.18 ± 2657.83 ng/mL, CL = 3.24 ± 1.47 mL/min/kg, F = 96.8 %). Further in vivo efficacy studies illustrated that 9a was capable of ameliorating formalin-induced pain and inhibiting capsaicin-induced ear edema.
Collapse
Affiliation(s)
- Wenli Chen
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China
| | - Qinlong Xu
- Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China
| | - Xiaodong Ma
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiajia Mo
- Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China
| | - Gaofeng Lin
- Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China
| | - Guangwei He
- Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China
| | - Zhaoxing Chu
- Hefei Industrial Pharmaceutical Institute Co., Ltd., Hefei, Anhui 230061, China.
| | - Jiaming Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
38
|
Zhao C, MacKinnon R. Structural and functional analyses of a GPCR-inhibited ion channel TRPM3. Neuron 2023; 111:81-91.e7. [PMID: 36283409 DOI: 10.1016/j.neuron.2022.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/03/2022] [Accepted: 09/29/2022] [Indexed: 12/24/2022]
Abstract
G-protein coupled receptors (GPCRs) govern the physiological response to stimuli by modulating the activity of downstream effectors, including ion channels. TRPM3 is an ion channel inhibited by GPCRs through direct interaction with G protein (Gβγ) released upon their activation. This GPCR-TRPM3 signaling pathway contributes to the analgesic effect of morphine. Here, we characterized Gβγ inhibition of TRPM3 using electrophysiology and single particle cryo-electron microscopy (cryo-EM). From electrophysiology, we obtained a half inhibition constant (IC50) of ∼240 nM. Using cryo-EM, we determined structures of mouse TRPM3 expressed in human cells with and without Gβγ and with and without PIP2, a lipid required for TRPM3 activity, at resolutions of 2.7-4.7 Å. Gβγ-TRPM3 interfaces vary depending on PIP2 occupancy; however, in all cases, Gβγ appears loosely attached to TRPM3. The IC50 in electrophysiology experiments raises the possibility that additional unknown factors may stabilize the TRPM3-Gβγ complex.
Collapse
Affiliation(s)
- Chen Zhao
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, the Rockefeller University, New York, NY 10065, United States
| | - Roderick MacKinnon
- Laboratory of Molecular Neurobiology and Biophysics, Howard Hughes Medical Institute, the Rockefeller University, New York, NY 10065, United States.
| |
Collapse
|
39
|
Wang S, Wang W, Ye X. Bibliometric Analysis of Global Research on Transient Receptor Potential Vanilloid 1 in the Field of Pain. J Pain Res 2023; 16:1517-1532. [PMID: 37193358 PMCID: PMC10182809 DOI: 10.2147/jpr.s407384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
Background Transient Receptor Potential Vanilloid 1 (TRPV1) is a heat-activated cation channel modulated by inflammatory mediators, which is closely related to pain and serves as a potential analgesic target. However, the bibliometric analyses summarizing TRPV1 in the field of pain are scarce. This study aims to summarize the current status of TRPV1 in pain and the potential research direction. Methods Articles regarding TRPV1 in the pain field between 2013 and 2022 were extracted from the Web of Science core collection database on 31 December 2022. Scientometric software (VOSviewer and CiteSpace 6.1.R6) were used to perform bibliometric analysis. This study provided data on the trend of the annual outputs, countries/regions, institutions, journals, authors, co-cited references and keywords. Results A total of 2462 publications related to TRPV1 in the field of pain were extracted from 2013 to 2022, which were written by 12,005 authors of 2304 institutions, 68 countries/regions in 686 journals, with 48,723 citations totally. The number of publications has grown rapidly over the past 10 years. Most publications were from the USA and China; the Seoul Natl Univ was the most active institution; Tominaga M published the most papers and Caterina MJ was the most productive co-cited author; The top-contributing journal was Pain; The most cited references was the article authored by Julius D. "Neuropathic pain", "inflammatory pain", "visceral pain" and "migraine" were the most common types of pain in this field. The mechanism of TRPV1 in pain was one of the main research directions. Conclusion This study presented an overview of the major research directions of TRPV1 in the pain field by bibliometric methods over the past decade. The results could reveal the research trends and the hotspots in the field and provide helpful information for clinical treatments of pain.
Collapse
Affiliation(s)
- Sisi Wang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Wen Wang
- Department of Preventive Treatment Center, Quzhou Hospital of Traditional Chinese Medicine, Quzhou, Zhejiang, People’s Republic of China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
- Correspondence: Xiangming Ye, Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, 158 Shangtang Road, Gongshu District, Hangzhou City, Zhejiang, People’s Republic of China, Tel +86 571 87692748, Email
| |
Collapse
|
40
|
Padín JF, Maroto M, Entrena JM, Egea J, Montell E, Vergés J, López MG, Cobos EJ, García AG. Small Synthetic Hyaluronan Disaccharide BIS014 Mitigates Neuropathic Pain in Mice. THE JOURNAL OF PAIN 2023; 24:68-83. [PMID: 36087908 DOI: 10.1016/j.jpain.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 07/31/2022] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a challenging condition to treat, as the need for new drugs to treat NP is an unmet goal. We investigated the analgesic potential of a new sulfated disaccharide compound, named BIS014. Oral administration (p.o.) of this compound induced ameliorative effects in formalin-induced nociception and capsaicin-induced secondary mechanical hypersensitivity in mice, but also after partial sciatic nerve transection (spared nerve injury), chemotherapy (paclitaxel)-induced NP, and diabetic neuropathy induced by streptozotocin. Importantly, BIS014, at doses active on neuropathic hypersensitivity (60 mg/kg/p.o.), did not alter exploratory activity or motor coordination (in the rotarod test), unlike a standard dose of gabapentin (40 mg/kg/p.o.) which although inducing antiallodynic effects on the NP models, it also markedly decreased exploration and motor coordination. In docking and molecular dynamic simulation studies, BIS014 interacted with TRPV1, a receptor involved in pain transmission where it behaved as a partial agonist. Additionally, similar to capsaicin, BIS014 increased cytosolic Ca2+ concentration ([Ca2+]c) in neuroblastoma cells expressing TRPV1 receptors; these elevations were blocked by ruthenium red. BIS014 did not block capsaicin-elicited [Ca2+]c transients, but inhibited the increase in the firing rate of action potentials in bradykinin-sensitized dorsal root ganglion neurons stimulated with capsaicin. Perspective: We report that the oral administration of a new sulfated disaccharide compound, named BIS014, decreases neuropathic pain from diverse etiology in mice. Unlike the comparator gabapentin, BIS014 does not induce sedation. Thus, BIS014 has the potential to become a new efficacious non-sedative oral medication for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Departamento de Ciencias Médicas (Farmacología), Facultad de Medicina, Universidad de Castilla-La Mancha, Ciudad Real, Spain.
| | - Marcos Maroto
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain.
| | - José Manuel Entrena
- Unidad de Análisis de Comportamiento Animal, Centro de Instrumentación Científica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain.
| | - Javier Egea
- Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Eulàlia Montell
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Josep Vergés
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Manuela G López
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Enrique J Cobos
- Departamento de Farmacología e Instituto de Neurociencias, Facultad de Medicina, Universidad de Granada e Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| | - Antonio G García
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| |
Collapse
|
41
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
42
|
Jennings EM, Sullivan LC, Jamshidi RJ, LoCoco PM, Smith HR, Chavera TS, Berg KA, Clarke WP. Age-related changes in peripheral nociceptor function. Neuropharmacology 2022; 216:109187. [PMID: 35835212 DOI: 10.1016/j.neuropharm.2022.109187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 01/05/2023]
Abstract
Pain and pain management in the elderly population is a significant social and medical problem. Pain sensation is a complex phenomenon that typically involves activation of peripheral pain-sensing neurons (nociceptors) which send signals to the spinal cord and brain that are interpreted as pain, an unpleasant sensory experience. In this work, young (4-5 months) and aged (26-27 months) Fischer 344 x Brown Norway (F344xBN) rats were examined for nociceptor sensitivity to activation by thermal (cold and heat) and mechanical stimulation following treatment with inflammatory mediators and activators of transient receptor potential (TRP) channels. Unlike other senses that decrease in sensitivity with age, sensitivity of hindpaw nociceptors to thermal and mechanical stimulation was not different between young and aged F344xBN rats. Intraplantar injection of bradykinin (BK) produced greater thermal and mechanical allodynia in aged versus young rats, whereas only mechanical allodynia was greater in aged rats following injection of prostaglandin E2 (PGE2). Intraplantar injection of TRP channel activators, capsaicin (TRPV1), mustard oil (TRPA1) and menthol (TRPM8) each resulted in greater mechanical allodynia in aged versus young rats and capsaicin-induced heat allodynia was also greater in aged rats. A treatment-induced allodynia that was greater in young rats was never observed. The anti-allodynic effects of intraplantar injection of kappa and delta opioid receptor agonists, salvinorin-A and D-Pen2,D-Pen5]enkephalin (DPDPE), respectively, were greater in aged than young rats, whereas mu opioid receptor agonists, [D-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) and morphine, were not effective in aged rats. Consistent with these observations, in primary cultures of peripheral sensory neurons, inhibition of cAMP signaling in response to delta and kappa receptor agonists was greater in cultures derived from aged rats. By contrast, mu receptor agonists did not inhibit cAMP signaling in aged rats. Thus, age-related changes in nociceptors generally favor increased pain signaling in aged versus young rats, suggesting that changes in nociceptor sensitivity may play a role in the increased incidence of pain in the elderly population. These results also suggest that development of peripherally-restricted kappa or delta opioid receptor agonists may provide safer and effective pain relief for the elderly.
Collapse
Affiliation(s)
- Elaine M Jennings
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Laura C Sullivan
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Raehannah J Jamshidi
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Peter M LoCoco
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Hudson R Smith
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Teresa S Chavera
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - Kelly A Berg
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA
| | - William P Clarke
- Department of Pharmacology, The University of Texas Health San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
43
|
Selezneva A, Gibb AJ, Willis D. The contribution of ion channels to shaping macrophage behaviour. Front Pharmacol 2022; 13:970234. [PMID: 36160429 PMCID: PMC9490177 DOI: 10.3389/fphar.2022.970234] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
The expanding roles of macrophages in physiological and pathophysiological mechanisms now include normal tissue homeostasis, tissue repair and regeneration, including neuronal tissue; initiation, progression, and resolution of the inflammatory response and a diverse array of anti-microbial activities. Two hallmarks of macrophage activity which appear to be fundamental to their diverse cellular functionalities are cellular plasticity and phenotypic heterogeneity. Macrophage plasticity allows these cells to take on a broad spectrum of differing cellular phenotypes in response to local and possibly previous encountered environmental signals. Cellular plasticity also contributes to tissue- and stimulus-dependent macrophage heterogeneity, which manifests itself as different macrophage phenotypes being found at different tissue locations and/or after different cell stimuli. Together, plasticity and heterogeneity align macrophage phenotypes to their required local cellular functions and prevent inappropriate activation of the cell, which could lead to pathology. To execute the appropriate function, which must be regulated at the qualitative, quantitative, spatial and temporal levels, macrophages constantly monitor intracellular and extracellular parameters to initiate and control the appropriate cell signaling cascades. The sensors and signaling mechanisms which control macrophages are the focus of a considerable amount of research. Ion channels regulate the flow of ions between cellular membranes and are critical to cell signaling mechanisms in a variety of cellular functions. It is therefore surprising that the role of ion channels in the macrophage biology has been relatively overlooked. In this review we provide a summary of ion channel research in macrophages. We begin by giving a narrative-based explanation of the membrane potential and its importance in cell biology. We then report on research implicating different ion channel families in macrophage functions. Finally, we highlight some areas of ion channel research in macrophages which need to be addressed, future possible developments in this field and therapeutic potential.
Collapse
|
44
|
Abstract
Transient receptor potential (TRP) ion channels are sophisticated signaling machines that detect a wide variety of environmental and physiological signals. Every cell in the body expresses one or more members of the extended TRP channel family, which consists of over 30 subtypes, each likely possessing distinct pharmacological, biophysical, and/or structural attributes. While the function of some TRP subtypes remains enigmatic, those involved in sensory signaling are perhaps best characterized and have served as models for understanding how these excitatory ion channels serve as polymodal signal integrators. With the recent resolution revolution in cryo-electron microscopy, these and other TRP channel subtypes are now yielding their secrets to detailed atomic analysis, which is beginning to reveal structural underpinnings of stimulus detection and gating, ion permeation, and allosteric mechanisms governing signal integration. These insights are providing a framework for designing and evaluating modality-specific pharmacological agents for treating sensory and other TRP channel-associated disorders.
Collapse
Affiliation(s)
- Melinda M Diver
- Department of Physiology, University of California, San Francisco, California, USA;
- Current affiliation: Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - John V Lin King
- Department of Physiology, University of California, San Francisco, California, USA;
- Current affiliation: Department of Biology, Stanford University, Palo Alto, California, USA
| | - David Julius
- Department of Physiology, University of California, San Francisco, California, USA;
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA;
- Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| |
Collapse
|
45
|
Añibarro-Ortega M, Pinela J, Alexopoulos A, Petropoulos SA, Ferreira ICFR, Barros L. The powerful Solanaceae: Food and nutraceutical applications in a sustainable world. ADVANCES IN FOOD AND NUTRITION RESEARCH 2022; 100:131-172. [PMID: 35659351 DOI: 10.1016/bs.afnr.2022.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The Solanaceae family is considered one of the most important families among plant species because, on one hand encompasses many staple food crops of the human diet while, on the other hand, it includes species rich in powerful secondary metabolites that could be valorized in medicine or drug formulation as well as nutraceuticals and food supplements. The main genera are Solanum, Capsicum, Physalis, and Lycium which comprise several important cultivated crops (e.g., tomato, pepper, eggplant, tomatillo, and goji berry), as well as genera notable for species with several pharmaceutical properties (e.g., Datura, Nicotiana, Atropa, Mandragora, etc.). This chapter discusses the nutritional value of the most important Solanaceae species commonly used for their edible fruit, as well as those used in the development of functional foods, food supplements, and nutraceuticals due to their bioactive constituents. The toxic and poisonous effects are also discussed aiming to highlight possible detrimental consequences due to irrational use. Finally, considering the high amount of waste and by-products generated through the value chain of the main crops, the sustainable management practices implemented so far are presented with the aim to increase the added-value of these crops.
Collapse
Affiliation(s)
- Mikel Añibarro-Ortega
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal
| | - José Pinela
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal.
| | - Alexios Alexopoulos
- Laboratory of Agronomy, Department of Agriculture, University of the Peloponnese, Kalamata, Messinia, Greece
| | - Spyridon A Petropoulos
- Department of Agriculture, Crop Production and Rural Environment, University of Thessaly, Volos, Greece
| | - Isabel C F R Ferreira
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal
| | - Lillian Barros
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Bragança, Portugal.
| |
Collapse
|
46
|
Activation of TRPV1 by capsaicin-loaded CaCO3 nanoparticle for tumor-specific therapy. Biomaterials 2022; 284:121520. [DOI: 10.1016/j.biomaterials.2022.121520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/21/2022] [Accepted: 04/09/2022] [Indexed: 01/07/2023]
|
47
|
Liu Y, Lyu Y, Wang H. TRP Channels as Molecular Targets to Relieve Endocrine-Related Diseases. Front Mol Biosci 2022; 9:895814. [PMID: 35573736 PMCID: PMC9095829 DOI: 10.3389/fmolb.2022.895814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 12/03/2022] Open
Abstract
Transient receptor potential (TRP) channels are polymodal channels capable of sensing environmental stimuli, which are widely expressed on the plasma membrane of cells and play an essential role in the physiological or pathological processes of cells as sensors. TRPs often form functional homo- or heterotetramers that act as cation channels to flow Na+ and Ca2+, change membrane potential and [Ca2+]i (cytosolic [Ca2+]), and change protein expression levels, channel attributes, and regulatory factors. Under normal circumstances, various TRP channels respond to intracellular and extracellular stimuli such as temperature, pH, osmotic pressure, chemicals, cytokines, and cell damage and depletion of Ca2+ reserves. As cation transport channels and physical and chemical stimulation receptors, TRPs play an important role in regulating secretion, interfering with cell proliferation, and affecting neural activity in these glands and their adenocarcinoma cells. Many studies have proved that TRPs are widely distributed in the pancreas, adrenal gland, and other glands. This article reviews the specific regulatory mechanisms of various TRP channels in some common glands (pancreas, salivary gland, lacrimal gland, adrenal gland, mammary gland, gallbladder, and sweat gland).
Collapse
|
48
|
Zhang Y, Miao Q, Shi S, Hao H, Li X, Pu Z, Yang Y, An H, Zhang W, Kong Y, Pang X, Gu C, Gamper N, Wu Y, Zhang H, Du X. Protein disulfide isomerase modulation of TRPV1 controls heat hyperalgesia in chronic pain. Cell Rep 2022; 39:110625. [PMID: 35385753 DOI: 10.1016/j.celrep.2022.110625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/14/2022] [Accepted: 03/15/2022] [Indexed: 12/21/2022] Open
Abstract
Protein disulfide isomerase (PDI) plays a key role in maintaining cellular homeostasis by mediating protein folding via catalyzing disulfide bond formation, breakage, and rearrangement in the endoplasmic reticulum. Increasing evidence suggests that PDI can be a potential treatment target for several diseases. However, the function of PDI in the peripheral sensory nervous system is unclear. Here we report the expression and secretion of PDI from primary sensory neurons is upregulated in inflammatory and neuropathic pain models. Deletion of PDI in nociceptive DRG neurons results in a reduction in inflammatory and neuropathic heat hyperalgesia. We demonstrate that secreted PDI activates TRPV1 channels through oxidative modification of extracellular cysteines of the channel, indicating that PDI acts as an unconventional positive modulator of TRPV1. These findings suggest that PDI in primary sensory neurons plays an important role in development of heat hyperalgesia and can be a potential therapeutic target for chronic pain.
Collapse
Affiliation(s)
- Yongxue Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Pharmacy, The First Hospital of Handan, Handan, Hebei, China
| | - Qi Miao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sai Shi
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Han Hao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinmeng Li
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zeyao Pu
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yakun Yang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin, Hebei, China
| | - Wei Zhang
- Department of Spinal Surgery of the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Youzhen Kong
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xu Pang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cunyang Gu
- Department of Pathology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China; Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, UK
| | - Yi Wu
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China.
| | - Hailin Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Xiaona Du
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
49
|
Kalpachidou T, Malsch P, Qi Y, Mair N, Geley S, Quarta S, Kummer KK, Kress M. Genetic and functional evidence for gp130/IL6ST-induced transient receptor potential ankyrin 1 upregulation in uninjured but not injured neurons in a mouse model of neuropathic pain. Pain 2022; 163:579-589. [PMID: 34252913 PMCID: PMC8832546 DOI: 10.1097/j.pain.0000000000002402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Peripheral nerve injuries result in pronounced alterations in dorsal root ganglia, which can lead to the development of neuropathic pain. Although the polymodal mechanosensitive transient receptor potential ankyrin 1 (TRPA1) ion channel is emerging as a relevant target for potential analgesic therapies, preclinical studies do not provide unequivocal mechanistic insight into its relevance for neuropathic pain pathogenesis. By using a transgenic mouse model with a conditional depletion of the interleukin-6 (IL-6) signal transducer gp130 in Nav1.8 expressing neurons (SNS-gp130-/-), we provide a mechanistic regulatory link between IL-6/gp130 and TRPA1 in the spared nerve injury (SNI) model. Spared nerve injury mice developed profound mechanical hypersensitivity as indicated by decreased withdrawal thresholds in the von Frey behavioral test in vivo, as well as a significant increase in mechanosensitivity of unmyelinated nociceptive primary afferents in ex vivo skin-nerve recordings. In contrast to wild type and control gp130fl/fl animals, SNS-gp130-/- mice did not develop mechanical hypersensitivity after SNI and exhibited low levels of Trpa1 mRNA in sensory neurons, which were partially restored by adenoviral gp130 re-expression in vitro. Importantly, uninjured but not injured neurons developed increased responsiveness to the TRPA1 agonist cinnamaldehyde, and neurons derived from SNS-gp130-/- mice after SNI were significantly less responsive to cinnamaldehyde. Our study shows for the first time that TRPA1 upregulation is attributed specifically to uninjured neurons in the SNI model, and this depended on the IL-6 signal transducer gp130. We provide a solution to the enigma of TRPA1 regulation after nerve injury and stress its significance as an important target for neuropathic pain disorders.
Collapse
Affiliation(s)
- Theodora Kalpachidou
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Malsch
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Yanmei Qi
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Norbert Mair
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephan Geley
- Institute of Pathophysiology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Serena Quarta
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai K. Kummer
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
50
|
Qiao Y, Zhang Y, Qiao Z, He W, Chen Y, Song D, Wang G, Guo N, Shao L, Tian Z, Wang Q, Yan L, Qian H. Discovery of (S)–N-(3-isopropylphenyl)-2-(5-phenylthiazol-2-yl)pyrrolidine-1-carboxamide as potent and brain-penetrant TRPV1 antagonist. Eur J Med Chem 2022; 233:114191. [DOI: 10.1016/j.ejmech.2022.114191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/20/2022] [Accepted: 02/06/2022] [Indexed: 11/30/2022]
|