1
|
Maltsev DI, Solotenkov MA, Mukhametshina LF, Sokolov RA, Solius GM, Jappy D, Tsopina AS, Fedotov IV, Lanin AA, Fedotov AB, Krut' VG, Ermakova YG, Moshchenko AA, Rozov A, Zheltikov AM, Podgorny OV, Belousov VV. Human TRPV1 is an efficient thermogenetic actuator for chronic neuromodulation. Cell Mol Life Sci 2024; 81:437. [PMID: 39448456 PMCID: PMC11502623 DOI: 10.1007/s00018-024-05475-x] [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: 05/12/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Thermogenetics is a promising neuromodulation technique based on the use of heat-sensitive ion channels. However, on the way to its clinical application, a number of questions have to be addressed. First, to avoid immune response in future human applications, human ion channels should be studied as thermogenetic actuators. Second, heating levels necessary to activate these channels in vivo in brain tissue should be studied and cytotoxicity of these temperatures addressed. Third, the possibility and safety of chronic neuromodulation has to be demonstrated. In this study, we present a comprehensive framework for thermogenetic neuromodulation in vivo using the thermosensitive human ion channel hTRPV1. By targeting hTRPV1 expression to excitatory neurons of the mouse brain and activating them within a non-harmful temperature range with a fiber-coupled infrared laser, we not only induced neuronal firing and stimulated locomotion in mice, but also demonstrated that thermogenetics can be employed for repeated neuromodulation without causing evident brain tissue injury. Our results lay the foundation for the use of thermogenetic neuromodulation in brain research and therapy of neuropathologies.
Collapse
Affiliation(s)
- Dmitry I Maltsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | | | - Liana F Mukhametshina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
- Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Rostislav A Sokolov
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 603022, Nizhny Novgorod, Russia
| | - Georgy M Solius
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia
| | - David Jappy
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
- Kazan Federal University, 420008, Kazan, Russia
| | | | - Ilya V Fedotov
- Lomonosov Moscow State University, 119991, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia
| | - Aleksandr A Lanin
- Lomonosov Moscow State University, 119991, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia
| | - Andrei B Fedotov
- Lomonosov Moscow State University, 119991, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia
| | - Viktoriya G Krut'
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Yulia G Ermakova
- European Molecular Biology Laboratory (EMBL), 69117, Heidelberg, Germany
| | - Aleksandr A Moshchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia
| | - Andrei Rozov
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia.
| | | | - Oleg V Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia.
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia.
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| | - Vsevolod V Belousov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997, Moscow, Russia.
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117997, Moscow, Russia.
- Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
- Life Improvement by Future Technologies (LIFT) Center, 143025, Moscow, Russia.
| |
Collapse
|
2
|
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
|
3
|
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: 6.0] [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
|
4
|
Maliszewska K, Miniewska K, Godlewski A, Gosk W, Mojsak M, Kretowski A, Ciborowski M. Changes in plasma endocannabinoids concentrations correlate with 18F-FDG PET/MR uptake in brown adipocytes in humans. Front Mol Biosci 2023; 10:1073683. [PMID: 37564131 PMCID: PMC10411954 DOI: 10.3389/fmolb.2023.1073683] [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: 10/18/2022] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction: Recent data suggest a possible role of endocannabinoids in the regulation of brown adipose tissue (BAT) activity. Those findings indicate potential treatment options for obesity. The aim of this study was to evaluate the relationship between plasma endocannabinoids concentrations and the presence of BAT in humans. Methods: The study group consisted of 25 subjects divided into two groups: BAT positive BAT(+), (n = 17, median age = 25 years) and BAT negative BAT(-), (n = 8, median age = 28 years). BAT was estimated using 18F-FDG PET/MR after 2 h of cold exposure. The level of plasma endocannabinoids was assessed at baseline, 60 min and 120 min of cold exposure. Results: In both groups, BAT(+) and BAT(-), during the cooling, we observed a decrease of the same endocannabinoids: arachidonoylethanolamide (AEA), eicosapentaenoyl ethanolamide (EPEA) and oleoyl ethanolamide (OEA) with a much more profound decline in BAT(+) subjects. Statistically significant fall of PEA (palmitoylethanolamide) and SEA (stearoylethanolamide) concentrations after 60 min (FC = 0.7, p = 0.007 and FC = 0.8, p = 0.03, respectively) and 120 min (FC = 0.81, p = 0.004, and FC = 0.9, p = 0.01, respectively) of cooling was observed only in individuals with BAT. Conclusion: We noticed the profound decline of endocannabinoids concentrations in subjects with increased 18F-FDG PET/MR uptake in BAT. Identification of a new molecules related to BAT activity may create a new target for obesity treatment.
Collapse
Affiliation(s)
- Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Miniewska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Godlewski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Wioleta Gosk
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Malgorzata Mojsak
- Independent Laboratory of Molecular Imaging, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
5
|
Rosenbaum T, Morales-Lázaro SL. Regulation of ThermoTRP Channels by PIP2 and Cholesterol. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:245-277. [PMID: 36988884 DOI: 10.1007/978-3-031-21547-6_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Transient receptor potential (TRP) ion channels are proteins that are expressed by diverse tissues and that play pivotal functions in physiology. These channels are polymodal and are activated by several stimuli. Among TRPs, some members of this family of channels respond to changes in ambient temperature and are known as thermoTRPs. These proteins respond to heat or cold in the noxious range and some of them to temperatures considered innocuous, as well as to mechanical, osmotic, and/or chemical stimuli. In addition to this already complex ability to respond to different signals, the activity of these ion channels can be fine-tuned by lipids. Two lipids well known to modulate ion channel activity are phosphatidylinositol 4,5-bisphosphate (PIP2) and cholesterol. These lipids can either influence the function of these proteins through direct interaction by binding to a site in the structure of the ion channel or through indirect mechanisms, which can include modifying membrane properties, such as curvature and rigidity, by regulating their expression or by modulating the actions of other molecules or signaling pathways that affect the physiology of ion channels. Here, we summarize the key aspects of the regulation of thermoTRP channels by PIP2 and cholesterol.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Sara L Morales-Lázaro
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
6
|
Xiao T, Sun M, Zhao C, Kang J. TRPV1: A promising therapeutic target for skin aging and inflammatory skin diseases. Front Pharmacol 2023; 14:1037925. [PMID: 36874007 PMCID: PMC9975512 DOI: 10.3389/fphar.2023.1037925] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/20/2023] [Indexed: 02/17/2023] Open
Abstract
TRPV1 is a non-selective channel receptor widely expressed in skin tissues, including keratinocytes, peripheral sensory nerve fibers and immune cells. It is activated by a variety of exogenous or endogenous inflammatory mediators, triggering neuropeptide release and neurogenic inflammatory response. Previous studies have shown that TRPV1 is closely related to the occurrence and/or development of skin aging and various chronic inflammatory skin diseases, such as psoriasis, atopic dermatitis, rosacea, herpes zoster, allergic contact dermatitis and prurigo nodularis. This review summarizes the structure of the TRPV1 channel and discusses the expression of TRPV1 in the skin as well as its role of TRPV1 in skin aging and inflammatory skin diseases.
Collapse
Affiliation(s)
- Tengfei Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Mingzhong Sun
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Chuanxiang Zhao
- Institute of Medical Genetics and Reproductive Immunity, School of Medical Science and Laboratory Medicine, Jiangsu College of Nursing, Huai'an, Jiangsu, China
| | - Jingjing Kang
- Department of Clinical Laboratory, Affiliated Hospital of Nanjing University Medical School, Yancheng First People's Hospital, Yancheng, Jiangsu, China
| |
Collapse
|
7
|
Oh J, Kwak HS, Kim MK. The influence of a carrier food on the perceived spiciness of chili pepper sauce. J SENS STUD 2022. [DOI: 10.1111/joss.12794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jungmin Oh
- SME solution Research Center, Korea Food Research Institute Wanju‐gun Jeollabuk‐do Republic of Korea
- Department of Food Science and Human Nutrition and K‐Food Research Center Jeonbuk National University Jeonju‐si Jeollabuk‐do Republic of Korea
| | - Han Sub Kwak
- Research Group of Food Processing Korea Food Research Institute Wanju‐gun Jeollabuk‐do Republic of Korea
- KFRI School University of Science and Technology Wanju‐gun Jeollabuk‐do Republic of Korea
| | - Mina K. Kim
- Department of Food Science and Human Nutrition and K‐Food Research Center Jeonbuk National University Jeonju‐si Jeollabuk‐do Republic of Korea
| |
Collapse
|
8
|
Fernández-Carvajal A, Fernández-Ballester G, Ferrer-Montiel A. TRPV1 in chronic pruritus and pain: Soft modulation as a therapeutic strategy. Front Mol Neurosci 2022; 15:930964. [PMID: 36117910 PMCID: PMC9478410 DOI: 10.3389/fnmol.2022.930964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic pain and pruritus are highly disabling pathologies that still lack appropriate therapeutic intervention. At cellular level the transduction and transmission of pain and pruritogenic signals are closely intertwined, negatively modulating each other. The molecular and cellular pathways involved are multifactorial and complex, including peripheral and central components. Peripherally, pain and itch are produced by subpopulations of specialized nociceptors that recognize and transduce algesic and pruritogenic signals. Although still under intense investigation, cumulative evidence is pointing to the thermosensory channel TRPV1 as a hub for a large number of pro-algesic and itchy agents. TRPV1 appears metabolically coupled to most neural receptors that recognize algesic and pruritic molecules. Thus, targeting TRPV1 function appears as a valuable and reasonable therapeutic strategy. In support of this tenet, capsaicin, a desensitizing TRPV1 agonist, has been shown to exhibit clinically relevant analgesic, anti-inflammatory, and anti-pruritic activities. However, potent TRPV1 antagonists have been questioned due to an hyperthermic secondary effect that prevented their clinical development. Thus, softer strategies directed to modulate peripheral TRPV1 function appear warranted to alleviate chronic pain and itch. In this regard, soft, deactivatable TRPV1 antagonists for topical or local application appear as an innovative approach for improving the distressing painful and itchy symptoms of patients suffering chronic pain or pruritus. Here, we review the data on these compounds and propose that this strategy could be used to target other peripheral therapeutic targets.
Collapse
|
9
|
Capsaicin for Weight Control: “Exercise in a Pill” (or Just Another Fad)? Pharmaceuticals (Basel) 2022; 15:ph15070851. [PMID: 35890150 PMCID: PMC9316879 DOI: 10.3390/ph15070851] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Medical management of obesity represents a large unmet clinical need. Animal experiments suggest a therapeutic potential for dietary capsaicin, the pungent ingredient in hot chili peppers, to lose weight. This is an attractive theory since capsaicin has been a culinary staple for thousands of years and is generally deemed safe when consumed in hedonically acceptable, restaurant-like doses. This review critically evaluates the available experimental and clinical evidence for and against capsaicin as a weight control agent and comes to the conclusion that capsaicin is not a magic “exercise in a pill”, although there is emerging evidence that it may help restore a healthy gut microbiota.
Collapse
|
10
|
Zhang J, Yuan H, Yao X, Chen S. Endogenous ion channels expressed in human embryonic kidney (HEK-293) cells. Pflugers Arch 2022; 474:665-680. [PMID: 35567642 DOI: 10.1007/s00424-022-02700-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/25/2022] [Accepted: 04/30/2022] [Indexed: 12/21/2022]
Abstract
Mammalian expression systems, particularly the human embryonic kidney (HEK-293) cells, combined with electrophysiological studies, have greatly benefited our understanding of the function, characteristic, and regulation of various ion channels. It was previously assumed that the existence of endogenous ion channels in native HEK-293 cells could be negligible. Still, more and more ion channels are gradually reported in native HEK-293 cells, which should draw our attention. In this regard, we summarize the different ion channels that are endogenously expressed in HEK-293 cells, including voltage-gated Na+ channels, Ca2+ channels, K+ channels, Cl- channels, nonselective cation channels, TRP channels, acid-sensitive ion channels, and Piezo channels, which may complicate the recording of the heterogeneously expressed ion channels to a certain degree. We noted that the expression patterns and channel profiles varied with different studies, which may be due to the distinct originality of the cells, cell culture conditions, passage numbers, and different recording protocols. Therefore, a better knowledge of endogenous ion channels may help minimize potential problems in characterizing heterologously expressed ion channels. Based on this, it is recommended that HEK-293 cells from unknown sources should be examined before transfection for the characterization of their functional profile, especially when the expression level of exogenous ion channels does not overwhelm the endogenous ion channels largely, or the current amplitude is not significantly higher than the native currents.
Collapse
Affiliation(s)
- Jun Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Huikai Yuan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Shuo Chen
- Department of Biopharmaceutical Sciences, School of Pharmacy, Harbin Medical University at Daqing, No. 39 Xinyang Rd, High-tech District, Daqing, 163319, Heilongjiang Province, China.
| |
Collapse
|
11
|
A Comparison Study of the Effect on IBS-D Rats among Ginger-Partitioned Moxibustion, Mild Moxibustion, and Laser Moxibustion. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4296216. [PMID: 34840586 PMCID: PMC8612783 DOI: 10.1155/2021/4296216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 01/30/2023]
Abstract
Background Diarrhea-predominant irritable bowel syndrome (IBS-D) is a functional gastrointestinal disorder that severely affects patients' life. Moxibustion is believed to be an effective way to treat IBS-D. However, the therapeutic effects and the underlying mechanisms in symptom management of IBS-D by different moxibustion therapies remain unclear. Methods IBS-D model rats were divided into groups and treated with ginger-partitioned moxibustion (GPM), mild moxibustion (MM), and laser moxibustion (LM) at a temperature of 43°C, respectively. The temperature curves of acupoints were recorded during interventions. The therapeutic effects were evaluated on the basis of general condition, stool, and hematoxylin-eosin staining of the colon tissue. Moreover, the expression of transient receptor potential vanilloid 1 (TRPV1) receptors in both acupoint tissue and colon tissue was analyzed by immunohistochemistry. Results After moxibustion treatment, the symptoms were improved. The expression of TRPV1 was increased in acupoint tissue and decreased in colon tissue. GPM and MM showed a more significant influence on IBS-D rats compared with LM. The temperature profile of GPM and MM was wave-like, while LM had an almost stable temperature curve. Conclusion GPM, MM, and LM could improve the symptoms in IBS-D rats. Moxibustion might activate TRPV1 channels in the acupoint tissue and induce acupoint functions, which in turn inhibit the pathological activation state of the colon's TRPV1, followed by improvements in abdominal pain and diarrheal symptoms. LM with stable temperature might lead to the desensitization of TRPV1 receptors and the tolerance of acupoint. GPM and MM provided dynamic and repetitive thermal stimulations that perhaps induced acupoint sensitization to increase efficacy. Therefore, dynamic and repetitive thermal stimulation is recommended in the application of moxibustion.
Collapse
|
12
|
Roy P, Martinelli I, Moruzzi M, Maggi F, Amantini C, Micioni Di Bonaventura MV, Cifani C, Amenta F, Tayebati SK, Tomassoni D. Ion channels alterations in the forebrain of high-fat diet fed rats. Eur J Histochem 2021; 65:3305. [PMID: 34814650 PMCID: PMC8636841 DOI: 10.4081/ejh.2021.3305] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Evidence suggests that transient receptor potential (TRP) ion channels dysfunction significantly contributes to the physiopathology of metabolic and neurological disorders. Dysregulation in functions and expression in genes encoding the TRP channels cause several inherited diseases in humans (the so-called 'TRP channelopathies'), which affect the cardiovascular, renal, skeletal, and nervous systems. This study aimed to evaluate the expression of ion channels in the forebrain of rats with diet-induced obesity (DIO). DIO rats were studied after 17 weeks under a hypercaloric diet (high-fat diet, HFD) and were compared to the control rats with a standard diet (CHOW). To determine the systemic effects of HFD exposure, we examined food intake, fat mass content, fasting glycemia, insulin levels, cholesterol, and triglycerides. qRT-PCR, Western blot, and immunochemistry analysis were performed in the frontal cortex (FC) and hippocampus (HIP). After 17 weeks of HFD, DIO rats increased their body weight significantly compared to the CHOW rats. In DIO rats, TRPC1 and TRPC6 were upregulated in the HIP, while they were downregulated in the FC. In the case of TRPM2 expression, instead was increased both in the HIP and in the FC. These could be related to the increase of proteins and nucleic acid oxidation. TRPV1 and TRPV2 gene expression showed no differences both in the FC and HIP. In general, qRT-PCR analyses were confirmed by Western blot analysis. Immunohistochemical procedures highlighted the expression of the channels in the cell body of neurons and axons, particularly for the TRPC1 and TRPC6. The alterations of TRP channel expression could be related to the activation of glial cells or the neurodegenerative process presented in the brain of the DIO rat highlighted with post synaptic protein (PSD 95) alterations. The availability of suitable animal models may be useful for studying possible pharmacological treatments to counter obesity-induced brain injury. The identified changes in DIO rats may represent the first insight to characterize the neuronal alterations occurring in obesity. Further investigations are necessary to characterize the role of TRP channels in the regulation of synaptic plasticity and obesity-related cognitive decline.
Collapse
Affiliation(s)
- Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | - Federica Maggi
- Department of Molecular Medicine, La Sapienza University of Rome.
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino.
| | | | | | | | | | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino.
| |
Collapse
|
13
|
Kollewe A, Chubanov V, Tseung FT, Correia L, Schmidt E, Rössig A, Zierler S, Haupt A, Müller CS, Bildl W, Schulte U, Nicke A, Fakler B, Gudermann T. The molecular appearance of native TRPM7 channel complexes identified by high-resolution proteomics. eLife 2021; 10:68544. [PMID: 34766907 PMCID: PMC8616561 DOI: 10.7554/elife.68544] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/08/2021] [Indexed: 12/22/2022] Open
Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed membrane protein consisting of ion channel and protein kinase domains. TRPM7 plays a fundamental role in the cellular uptake of divalent cations such as Zn2+, Mg2+, and Ca2+, and thus shapes cellular excitability, plasticity, and metabolic activity. The molecular appearance and operation of TRPM7 channels in native tissues have remained unresolved. Here, we investigated the subunit composition of endogenous TRPM7 channels in rodent brain by multi-epitope affinity purification and high-resolution quantitative mass spectrometry (MS) analysis. We found that native TRPM7 channels are high-molecular-weight multi-protein complexes that contain the putative metal transporter proteins CNNM1-4 and a small G-protein ADP-ribosylation factor-like protein 15 (ARL15). Heterologous reconstitution experiments confirmed the formation of TRPM7/CNNM/ARL15 ternary complexes and indicated that complex formation effectively and specifically impacts TRPM7 activity. These results open up new avenues towards a mechanistic understanding of the cellular regulation and function of TRPM7 channels.
Collapse
Affiliation(s)
- Astrid Kollewe
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vladimir Chubanov
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Fong Tsuen Tseung
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Leonor Correia
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Eva Schmidt
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Anna Rössig
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Susanna Zierler
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,Institute of Pharmacology, Johannes Kepler University Linz, Linz, Austria
| | - Alexander Haupt
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Catrin Swantje Müller
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Bildl
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Uwe Schulte
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Annette Nicke
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Bernd Fakler
- Institute of Physiology II, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, Freiburg, Germany
| | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,German Center for Lung Research, Munich, Germany
| |
Collapse
|
14
|
Tobita N, Tsuneto K, Ito S, Yamamoto T. Human TRPV1 and TRPA1 are receptors for bacterial quorum sensing molecules. J Biochem 2021; 170:775-785. [PMID: 34557892 DOI: 10.1093/jb/mvab099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
In this study, we investigated the activation of TRPV1 and TRPA1 by N-acyl homoserine lactones, quorum sensing molecules produced by Gram-negative bacteria, and the inhibitory effect of TRPV1 and TRPA1 by autoinducing peptides, quorum sensing molecules produced by Gram-positive bacteria, using human embryonic kidney 293T cell lines stably expressing human TRPV1 and TRPA1, respectively. As a result, we found that some N-acyl homoserine lactones, such as N-octanoyl-L-homoserine lactone (C8-HSL), N-nonanoyl-L-homoserine lactone (C9-HSL) and N-decanoyl-L-homoserine lactone (C10-HSL) activated both TRPV1 and TRPA1. In addition, we clarified that some N-acyl homoserine lactones, for example, N-3-oxo-dodecanoyl-L-homoserine lactone (3-oxo-C12-HSL) only activated TRPV1, and N-acyl homoserine lactones having saturated short acyl chain, such as N-acetyl-L-homoserine lactone (C2-HSL) and N-butyryl-L-homoserine lactone (C4-HSL) only activated TRPA1, respectively. Furthermore, we found that an autoinducing peptide, simple linear peptide CHWPR, inhibited both TRPV1 and TRPA1, and peptide having thiolactone ring DICNAYF, thiolactone ring were formed between C3 to F7, strongly inhibited only the TRPV1. Although the specificity of TRPV1 and TRPA1 for quorum sensing molecules were different, these data suggest that both TRPV1 and TRPA1 would function as receptors for quorum sensing molecule produced by bacteria.
Collapse
Affiliation(s)
- Naoya Tobita
- Tobacco Science Research Center, Japan Tobacco Inc., 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Kana Tsuneto
- Tobacco Science Research Center, Japan Tobacco Inc., 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Shigeaki Ito
- Scientific Product Assessment Center, Japan Tobacco Inc., 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Takeshi Yamamoto
- Tobacco Science Research Center, Japan Tobacco Inc., 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| |
Collapse
|
15
|
Chon S, Kim MK. Analysis of capsaicin and the perceived spiciness of cabbage kimchi according to different preparation methods. J SENS STUD 2021. [DOI: 10.1111/joss.12709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Seo‐yeong Chon
- Department of Food Science and Human Nutrition and Obesity Research Center Jeonbuk National University Jeonju‐si Jeollabukdo Republic of Korea
| | - Mina K. Kim
- Department of Food Science and Human Nutrition and Obesity Research Center Jeonbuk National University Jeonju‐si Jeollabukdo Republic of Korea
| |
Collapse
|
16
|
Biringer RG. A review of non-prostanoid, eicosanoid receptors: expression, characterization, regulation, and mechanism of action. J Cell Commun Signal 2021; 16:5-46. [PMID: 34173964 DOI: 10.1007/s12079-021-00630-6] [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: 05/21/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022] Open
Abstract
Eicosanoid signaling controls a wide range of biological processes from blood pressure homeostasis to inflammation and resolution thereof to the perception of pain and to cell survival itself. Disruption of normal eicosanoid signaling is implicated in numerous disease states. Eicosanoid signaling is facilitated by G-protein-coupled, eicosanoid-specific receptors and the array of associated G-proteins. This review focuses on the expression, characterization, regulation, and mechanism of action of non-prostanoid, eicosanoid receptors.
Collapse
Affiliation(s)
- Roger G Biringer
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, 5000 Lakewood Ranch Blvd, Bradenton, FL, 34211, USA.
| |
Collapse
|
17
|
Bagood MD, Isseroff RR. TRPV1: Role in Skin and Skin Diseases and Potential Target for Improving Wound Healing. Int J Mol Sci 2021; 22:ijms22116135. [PMID: 34200205 PMCID: PMC8201146 DOI: 10.3390/ijms22116135] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Skin is innervated by a multitude of sensory nerves that are important to the function of this barrier tissue in homeostasis and injury. The role of innervation and neuromediators has been previously reviewed so here we focus on the role of the transient receptor potential cation channel, subfamily V member 1 (TRPV1) in wound healing, with the intent of targeting it in treatment of non-healing wounds. TRPV1 structure and function as well as the outcomes of TRPV1-targeted therapies utilized in several diseases and tissues are summarized. In skin, keratinocytes, sebocytes, nociceptors, and several immune cells express TRPV1, making it an attractive focus area for treating wounds. Many intrinsic and extrinsic factors confound the function and targeting of TRPV1 and may lead to adverse or off-target effects. Therefore, a better understanding of what is known about the role of TRPV1 in skin and wound healing will inform future therapies to treat impaired and chronic wounds to improve healing.
Collapse
Affiliation(s)
- Michelle D. Bagood
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA 95816, USA;
| | - R. Rivkah Isseroff
- Department of Dermatology, School of Medicine, UC Davis, Sacramento, CA 95816, USA;
- Dermatology Section, VA Northern California Health Care System, Mather, CA 95655, USA
- Correspondence: ; Tel.: +1-(916)-551-2606
| |
Collapse
|
18
|
Fischer MJM, Ciotu CI, Szallasi A. The Mysteries of Capsaicin-Sensitive Afferents. Front Physiol 2020; 11:554195. [PMID: 33391007 PMCID: PMC7772409 DOI: 10.3389/fphys.2020.554195] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
A fundamental subdivision of nociceptive sensory neurons is named after their unique sensitivity to capsaicin, the pungent ingredient in hot chili peppers: these are the capsaicin-sensitive afferents. The initial excitation by capsaicin of these neurons manifested as burning pain sensation is followed by a lasting refractory state, traditionally referred to as "capsaicin desensitization," during which the previously excited neurons are unresponsive not only to capsaicin but a variety of unrelated stimuli including noxious heat. The long sought-after capsaicin receptor, now known as TRPV1 (transient receptor potential cation channel, subfamily V member 1), was cloned more than two decades ago. The substantial reduction of the inflammatory phenotype of Trpv1 knockout mice has spurred extensive efforts in the pharmaceutical industry to develop small molecule TRPV1 antagonists. However, adverse effects, most importantly hyperthermia and burn injuries, have so far prevented any compounds from progressing beyond Phase 2. There is increasing evidence that these limitations can be at least partially overcome by approaches outside of the mainstream pharmaceutical development, providing novel therapeutic options through TRPV1. Although ablation of the whole TRPV1-expressing nerve population by high dose capsaicin, or more selectively by intersectional genetics, has allowed researchers to investigate the functions of capsaicin-sensitive afferents in health and disease, several "mysteries" remain unsolved to date, including the molecular underpinnings of "capsaicin desensitization," and the exact role these nerves play in thermoregulation and heat sensation. This review tries to shed some light on these capsaicin mechanisms.
Collapse
Affiliation(s)
- Michael J. M. Fischer
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Cosmin I. Ciotu
- Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Arpad Szallasi
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Tobita N, Makino M, Fujita R, Jyotaki M, Shinohara Y, Yamamoto T. Sweet scent lactones activate hot capsaicin receptor, TRPV1. Biochem Biophys Res Commun 2020; 534:547-552. [PMID: 33239169 DOI: 10.1016/j.bbrc.2020.11.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
In this study, we investigated the activation of Transient receptor potential vanilloid subtype 1, TRPV1, by lactones, a representative flavor ingredient currently used for foods and beverages. As a result, we found that some lactones having C4 acyl chain length, γ-octalactone, δ-nonalactone and β-methyl-γ-octalactone, γ-undecalactone with C7 acyl chain length and δ-undecalactone with C6 acyl chain length activated TRPV1. TRPV1 is known as a non-selective cation channels that respond to a wide range of physical and chemical stimuli such as high temperature, protons, capsaicin and so on. Furthermore, it has been also demonstrated that activation of TRPV1 induced energy expenditure enhancement and thermogenesis, suppressed accumulation of visceral fat in mice and prevented non-alcoholic fatty acid liver. Thus, lactones that function as TRPV1 agonists are thought to be important candidates for decreasing the risks of developing a metabolic syndrome.
Collapse
Affiliation(s)
- Naoya Tobita
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Masanari Makino
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Ryujiro Fujita
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Masafumi Jyotaki
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Yuhei Shinohara
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan
| | - Takeshi Yamamoto
- Tobacco Science Research Center, Japan Tobacco Inc, 6-2 Umegaoka, Aoba, Yokohama, Kanagawa, 227-8512, Japan.
| |
Collapse
|
20
|
Lu CL, Liao CH, Lu KC, Ma MC. TRPV1 Hyperfunction Involved in Uremic Toxin Indoxyl Sulfate-Mediated Renal Tubular Damage. Int J Mol Sci 2020; 21:ijms21176212. [PMID: 32867359 PMCID: PMC7503230 DOI: 10.3390/ijms21176212] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Indoxyl sulfate (IS) is accumulated during severe renal insufficiency and known for its nephrotoxic properties. Transient receptor potential vanilloid 1 (TRPV1) is present in the kidney and acts as a renal sensor. However, the mechanism underlying IS-mediated renal tubular damage in view of TRPV1 is lacking. Here, we demonstrated that TRPV1 was expressed in tubular cells of Lilly Laboratories cell-porcine kidney 1 (LLC-PK1) and Madin-Darby canine kidney cells (MDCK). IS treatment in both cells exhibited tubular damage with increased LDH release and reduced cell viability in dose- and time-dependent manners. MDCK, however, was more vulnerable to IS. We, therefore, investigated MDCK cells to explore a more detailed mechanism. Interestingly, IS-induced tubular damage was markedly attenuated in the presence of selective TRPV1 blockers. IS showed no effect on TRPV1 expression but significantly increased arachidonate 12-lipoxygenase (ALOX12) protein, mRNA expression, and 12(S)-hydroxyeicosatetraenoic acid (12(S)-HETE) amounts in a dose-dependent manner, indicating that the ALOX12/12(S)-HETE pathway induced TRPV1 hyperfunction in IS-mediated tubulotoxicity. Blockade of ALOX12 by cinnamyl-3,4-dihydroxy-α-cyanocinnamate or baicalein attenuated the effects of IS. Since aryl hydrocarbon receptor (AhR) activation after IS binding is crucial in mediating cell death, here, we found that the AhR blockade not only ameliorated tubular damage but also attenuated ALOX12 expression and 12(S)-HETE production caused by IS. The uremic toxic adsorbent AST-120, however, showed little effect on ALOX12 and 12(S)-HETE, as well as IS-induced cell damage. These results clearly indicated that IS activated AhR and then upregulated ALOX12, and this induced endovanilloid 12(S)-HETE synthesis and contributed to TRPV1 hyperfunction in IS-treated tubular cells. Further study on TRPV1 may attenuate kidney susceptibility to the functional loss of end-stage kidney disease via IS.
Collapse
Affiliation(s)
- Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, New Taipei City 24205, Taiwan;
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Chun-Hou Liao
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- Divisions of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei City 23148, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
| | - Ming-Chieh Ma
- School of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- Correspondence:
| |
Collapse
|
21
|
Searching for Predictors of Migraine Chronification: a Pilot Study of 1911A>G Polymorphism of TRPV1 Gene in Episodic Versus Chronic Migraine. J Mol Neurosci 2020; 71:618-624. [PMID: 32827294 DOI: 10.1007/s12031-020-01683-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) receptors activated by heat and capsaicin are expressed in trigeminal nociceptive neurons and implicated in the generation of migraine pain. Genetic studies suggested that single-nucleotide polymorphism (SNP) 1911A>G (rs8065080), leading to amino acid substitution Ile585Val, in the TRPV1 gene affects functional activity of TRPV1 receptors and is involved in different pain conditions. However, this polymorphism has not been tested in migraine patients. The objective of this pilot study was to investigate genetic factors of migraine susceptibility. We evaluated frequency distribution of AA, AG, and GG variants of SNP 1911A>G in the TRPV1 gene in patients with episodic and chronic migraine compared with healthy individuals. The study included 46 patients diagnosed with migraine (27 episodic and 19 chronic) and 50 healthy individuals as a control group. DNA from peripheral blood was used to test TRPV1 SNP using allele-specific PCR combined with gel electrophoresis. The genotype frequency distribution in episodic migraine was comparable with that in controls (AA 33%, AG 56%, GG 11% and AA 34%, AG 46%, GG 20%, respectively). On the contrary, in chronic migraine, the distribution differed significantly (p < 0.05) (AA 68%, AG 32%, GG 0%). This are first indications for a distinctive genotype frequency distribution of TRPV1 1911A>G in chronic migraine patients compared with episodic migraine patients and controls. Our data confirm a different predisposition to chronic pain in migraine and give a prerequisite for a new look at the nature of chronification of migraine, proposing that the absence of GG genotype may be considered as possible risk biomarker of episodic migraine evolution to chronic form.
Collapse
|
22
|
Chu Y, Cohen BE, Chuang HH. A single TRPV1 amino acid controls species sensitivity to capsaicin. Sci Rep 2020; 10:8038. [PMID: 32415171 PMCID: PMC7229161 DOI: 10.1038/s41598-020-64584-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 04/11/2020] [Indexed: 11/09/2022] Open
Abstract
Chili peppers produce capsaicin (a vanilloid) that activates the transient receptor potential cation channel subfamily V member 1 (TRPV1) on sensory neurons to alter their membrane potential and induce pain. To identify residues responsible for differential TRPV1 capsaicin sensitivity among species, we used intracellular Ca2+ imaging to characterize chimeras composed of capsaicin-sensitive rat TRPV1 (rTRPV1) and capsaicin-insensitive chicken TRPV1 (cTRPV1) exposed to a series of capsaicinoids. We found that chimeras containing rat E570-V686 swapped into chicken receptors displayed capsaicin sensitivity, and that simply changing the alanine at position 578 in the S4-S5 helix of the chicken receptor to a glutamic acid was sufficient to endow it with capsaicin sensitivity in the micromolar range. Moreover, introduction of lysine, glutamine or proline at residue A578 also elicited capsaicin sensitivity in cTRPV1. Similarly, replacing corresponding rTRPV1 residue E570 with lysine or glutamine retained capsaicin sensitivity. The hydrophilic capsaicin analog Cap-EA activated a cTRPV1-A578E mutant, suggesting that A578 may participate in vanilloid binding. The hydrophilic vanilloid agonist zingerone did not activate any A578 mutants with capsaicin sensitivity, suggesting that the vanilloid group alone is not sufficient for receptor activation. Our study demonstrates that a subtle modification of TRPV1 in different species globally alters capsaicin responses.
Collapse
Affiliation(s)
- Ying Chu
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan.
| | - Bruce E Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Huai-Hu Chuang
- Institute of Molecular Biology, Academia Sinica, Taipei, 11529, Taiwan
| |
Collapse
|
23
|
Dong Y, Yin Y, Vu S, Yang F, Yarov-Yarovoy V, Tian Y, Zheng J. A distinct structural mechanism underlies TRPV1 activation by piperine. Biochem Biophys Res Commun 2019; 516:365-372. [PMID: 31213294 DOI: 10.1016/j.bbrc.2019.06.039] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
Piperine, the principle pungent compound in black peppers, is known to activate the capsaicin receptor TRPV1 ion channel. How piperine interacts with the channel protein, however, remains unclear. Here we show that piperine binds to the same ligand-binding pocket as capsaicin but in different poses. There was no detectable detrimental effect when T551 and E571, two major sites known to form hydrogen bond with capsaicin, were mutated to a hydrophobic amino acid. Computational structural modeling suggested that piperine makes interactions with multiple amino acids within the ligand binding pocket, including T671 on the pore-forming S6 segment. Mutations of this residue could substantially reduce or even eliminate piperine-induced activation, confirming that T671 is an important site. Our results suggest that the bound piperine may directly interact with the pore-forming S6 segment to induce channel opening. These findings help to explain why piperine is a weak agonist, and may guide future efforts to develop novel pharmaceutical reagents targeting TRPV1.
Collapse
Affiliation(s)
- Yawen Dong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, Shandong, China
| | - Yue Yin
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, Shandong, China
| | - Simon Vu
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, 95616, USA
| | - Fan Yang
- Department of Biophysics and Kidney Disease Center, First Affiliated Hospital, Institute of Neuroscience, National Health Commission and Chinese Academy of Medical Sciences Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, 95616, USA
| | - Yuhua Tian
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, Shandong, China.
| | - Jie Zheng
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, 95616, USA.
| |
Collapse
|
24
|
Kasimova MA, Yazici AT, Yudin Y, Granata D, Klein ML, Rohacs T, Carnevale V. A hypothetical molecular mechanism for TRPV1 activation that invokes rotation of an S6 asparagine. J Gen Physiol 2018; 150:1554-1566. [PMID: 30333107 PMCID: PMC6219692 DOI: 10.1085/jgp.201812124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/13/2018] [Accepted: 09/26/2018] [Indexed: 12/13/2022] Open
Abstract
TRPV1 channels comprise four subunits containing six transmembrane segments (S1–S6) that surround a central pore. Kasimova et al. hypothesize that channel opening involves rotation of an S6 asparagine residue toward the pore, as well as associated pore hydration and external cavity dehydration. The transient receptor potential channel vanilloid type 1 (TRPV1) is activated by a variety of endogenous and exogenous stimuli and is involved in nociception and body temperature regulation. Although the structure of TRPV1 has been experimentally determined in both the closed and open states, very little is known about its activation mechanism. In particular, the conformational changes that occur in the pore domain and result in ionic conduction have not yet been identified. Here we suggest a hypothetical molecular mechanism for TRPV1 activation, which involves rotation of a conserved asparagine in S6 from a position facing the S4–S5 linker toward the pore. This rotation is associated with hydration of the pore and dehydration of the four peripheral cavities located between each S6 and S4–S5 linker. In light of our hypothesis, we perform bioinformatics analyses of TRP and other evolutionary related ion channels, evaluate newly available structures, and reexamine previously reported water accessibility and mutagenesis experiments. These analyses provide several independent lines of evidence to support our hypothesis. Finally, we show that our proposed molecular mechanism is compatible with the prevailing theory that the selectivity filter acts as a secondary gate in TRPV1.
Collapse
Affiliation(s)
- Marina A Kasimova
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA
| | - Aysenur Torun Yazici
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ
| | - Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ
| | - Daniele Granata
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA
| | - Michael L Klein
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA
| |
Collapse
|
25
|
Barbero R, Vercelli C, Cuniberti B, Della Valle MF, Martano M, Re G. Expression of functional TRPV1 receptor in primary culture of canine keratinocytes. J Vet Pharmacol Ther 2018; 41:795-804. [PMID: 30043987 DOI: 10.1111/jvp.12694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/18/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
The interest for the endovanilloid system and for transient receptor potential vanilloid 1 (TRPV1) is continuously increasing, due to their involvement in inflammation, nociception and pruritus. Even if TRPV1 enrolment was highlighted in both physiological and pathological conditions, some aspects remain unclear, mostly in veterinary medicine. This study aimed to verify the expression and functionality of TRPV1 in canine keratinocytes to investigate in vitro the role of TRPV1 in these cells that are involved in different cutaneous pathologies. Keratinocytes primary cultures were isolated from bioptical samples and cultivated. Binding assay (using 3 [H]-resiniferatoxin), displacement assay (in the presence of 1.2 nM 3 [H]-resiniferatoxin) and functional assays (in the presence of 1 μCi/45 Ca2+ ) with vanilloid agonists and antagonists, specifically addressed to TRPV1 receptor, were performed. Binding assay demonstrated the presence of measurable concentrations of TRPV1 (Bmax = 1,240 ± 120 fmol/mg protein; Kd = 0.01 ± 0.004 nM). Displacement assay highlighted the highest affinity for resiniferatoxin (RTX) and 5-iodo-resiniferatoxin (5-I-RTX), among agonists and antagonists, respectively. The same compounds results as the most potent in the functional assays. This study demonstrated the identification and the characterization of TRPV1 receptor in primary canine keratinocytes cultures. The results are promising for a clinical use, but further in vivo investigations are required.
Collapse
Affiliation(s)
- Raffaella Barbero
- SC of Serology, Istituto Zooprofilattico Sperimentale Piemonte Liguria e Valle d'Aosta, Turin, Italy
| | - Cristina Vercelli
- Department of Veterinary Sciences of Turin, University of Turin, Turin, Italy
| | - Barbara Cuniberti
- Royal Dick School of Veterinary Medicine, The University of Edinburg, Edinburg, Ireland
| | | | - Marina Martano
- Department of Veterinary Sciences of Turin, University of Turin, Turin, Italy
| | - Giovanni Re
- Department of Veterinary Sciences of Turin, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Merighi A. The histology, physiology, neurochemistry and circuitry of the substantia gelatinosa Rolandi (lamina II) in mammalian spinal cord. Prog Neurobiol 2018; 169:91-134. [PMID: 29981393 DOI: 10.1016/j.pneurobio.2018.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 06/07/2018] [Accepted: 06/30/2018] [Indexed: 02/06/2023]
Abstract
The substantia gelatinosa Rolandi (SGR) was first described about two centuries ago. In the following decades an enormous amount of information has permitted us to understand - at least in part - its role in the initial processing of pain and itch. Here, I will first provide a comprehensive picture of the histology, physiology, and neurochemistry of the normal SGR. Then, I will analytically discuss the SGR circuits that have been directly demonstrated or deductively envisaged in the course of the intensive research on this area of the spinal cord, with particular emphasis on the pathways connecting the primary afferent fibers and the intrinsic neurons. The perspective existence of neurochemically-defined sets of primary afferent neurons giving rise to these circuits will be also discussed, with the proposition that a cross-talk between different subsets of peptidergic fibers may be the structural and functional substrate of additional gating mechanisms in SGR. Finally, I highlight the role played by slow acting high molecular weight modulators in these gating mechanisms.
Collapse
Affiliation(s)
- Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095 Grugliasco (TO), Italy.
| |
Collapse
|
27
|
Ramírez-Barrantes R, Córdova C, Gatica S, Rodriguez B, Lozano C, Marchant I, Echeverria C, Simon F, Olivero P. Transient Receptor Potential Vanilloid 1 Expression Mediates Capsaicin-Induced Cell Death. Front Physiol 2018; 9:682. [PMID: 29922176 PMCID: PMC5996173 DOI: 10.3389/fphys.2018.00682] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022] Open
Abstract
The transient receptor potential (TRP) ion channel family consists of a broad variety of non-selective cation channels that integrate environmental physicochemical signals for dynamic homeostatic control. Involved in a variety of cellular physiological processes, TRP channels are fundamental to the control of the cell life cycle. TRP channels from the vanilloid (TRPV) family have been directly implicated in cell death. TRPV1 is activated by pain-inducing stimuli, including inflammatory endovanilloids and pungent exovanilloids, such as capsaicin (CAP). TRPV1 activation by high doses of CAP (>10 μM) leads to necrosis, but also exhibits apoptotic characteristics. However, CAP dose-response studies are lacking in order to determine whether CAP-induced cell death occurs preferentially via necrosis or apoptosis. In addition, it is not known whether cytosolic Ca2+ and mitochondrial dysfunction participates in CAP-induced TRPV1-mediated cell death. By using TRPV1-transfected HeLa cells, we investigated the underlying mechanisms involved in CAP-induced TRPV1-mediated cell death, the dependence of CAP dose, and the participation of mitochondrial dysfunction and cytosolic Ca2+ increase. Together, our results contribute to elucidate the pathophysiological steps that follow after TRPV1 stimulation with CAP. Low concentrations of CAP (1 μM) induce cell death by a mechanism involving a TRPV1-mediated rapid and transient intracellular Ca2+ increase that stimulates plasma membrane depolarization, thereby compromising plasma membrane integrity and ultimately leading to cell death. Meanwhile, higher doses of CAP induce cell death via a TRPV1-independent mechanism, involving a slow and persistent intracellular Ca2+ increase that induces mitochondrial dysfunction, plasma membrane depolarization, plasma membrane loss of integrity, and ultimately, cell death.
Collapse
Affiliation(s)
- Ricardo Ramírez-Barrantes
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Córdova
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Sebastian Gatica
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Belén Rodriguez
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlo Lozano
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Ivanny Marchant
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Cesar Echeverria
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Pablo Olivero
- Laboratorio de Estructura y Función Celular, Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
28
|
Kasimova MA, Yazici A, Yudin Y, Granata D, Klein ML, Rohacs T, Carnevale V. Ion Channel Sensing: Are Fluctuations the Crux of the Matter? J Phys Chem Lett 2018; 9:1260-1264. [PMID: 29439562 PMCID: PMC6310152 DOI: 10.1021/acs.jpclett.7b03396] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The nonselective cation channel TRPV1 is responsible for transducing noxious stimuli into action potentials propagating through peripheral nerves. It is activated by temperatures greater than 43 °C, while remaining completely nonconductive at temperatures lower than this threshold. The origin of this sharp response, which makes TRPV1 a biological temperature sensor, is not understood. Here we used molecular dynamics simulations and free energy calculations to characterize the molecular determinants of the transition between nonconductive and conductive states. We found that hydration of the pore and thus ion permeation depends critically on the polar character of its molecular surface: in this narrow hydrophobic enclosure, the motion of a polar side-chain is sufficient to stabilize either the dry or wet state. The conformation of this side-chain is in turn coupled to the hydration state of four peripheral cavities, which undergo a dewetting transition at the activation temperature.
Collapse
Affiliation(s)
- Marina A. Kasimova
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122
| | - Aysenur Yazici
- Department of Pharmacology, Physiology and Neuroscience, Rutgers–New Jersey, Medical School, Newark, NJ 07103
| | - Yevgen Yudin
- Department of Pharmacology, Physiology and Neuroscience, Rutgers–New Jersey, Medical School, Newark, NJ 07103
| | - Daniele Granata
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122
| | - Michael L. Klein
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122
| | - Tibor Rohacs
- Department of Pharmacology, Physiology and Neuroscience, Rutgers–New Jersey, Medical School, Newark, NJ 07103
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, Temple University, Philadelphia, PA 19122
| |
Collapse
|
29
|
Nazıroğlu M, Braidy N. Thermo-Sensitive TRP Channels: Novel Targets for Treating Chemotherapy-Induced Peripheral Pain. Front Physiol 2017; 8:1040. [PMID: 29326595 PMCID: PMC5733463 DOI: 10.3389/fphys.2017.01040] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 11/29/2017] [Indexed: 12/19/2022] Open
Abstract
Abnormal Ca2+ channel physiology, expression levels, and hypersensitivity to heat have been implicated in several pain states following treatment with chemotherapeutic agents. As members of the Ca2+ permeable transient receptor potential (TRP), five of the channels (TRPV1-4 and TRPM2) are activated by different heat temperatures, and two of the channels (TRPA1 and TRPM8) are activated by cold temperature. Accumulating evidences indicates that antagonists of TRPA1 and TRPM8 may protect against cisplatin, oxaliplatin, and paclitaxel-induced mitochondrial oxidative stress, inflammation, cold allodynia, and hyperalgesia. TRPV1 was responsible from the cisplatin-induced heat hyperalgesia and mechanical allodynia in the sensory neurons. TRPA1, TRPM8, and TRPV2 protein expression levels were mostly increased in the dorsal root (DRG) and trigeminal ganglia by these treatments. There is a debate on direct or oxaliplatin-induced oxidative cold stress dependent TRPA1 and TRPV4 activation in the DRG. Involvement of molecular pathways such as cysteine groups, glutathione metabolism, anandamide, cAMP, lipopolysaccharide, proteinase-activated receptor 2, and mitogen-activated protein kinase were also indicated in the oxaliplatin and paclitaxel-induced cold allodynia. In this review, we summarized results of five temperature-regulated TRP channels (TRPA1, TRPM8, TRPV1, TRPV2, and TRPV4) as novel targets for treating chemotherapy-induced peripheral pain
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
30
|
Ambrus L, Kelemen B, Szabó T, Bíró T, Tóth BI. Human podocytes express functional thermosensitive TRPV channels. Br J Pharmacol 2017; 174:4493-4507. [PMID: 28945920 DOI: 10.1111/bph.14052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 08/16/2017] [Accepted: 09/10/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Heat-sensitive transient receptor potential vanilloid (TRPV) channels are expressed in various epithelial tissues regulating, among else, barrier functions. Their expression is well established in the distal nephron; however, we have no data about their presence in podocytes. As podocytes are indispensable in the formation of the glomerular filtration barrier, we investigated the presence and function of Ca2+ -permeable TRPV1-4 channels in human podocyte cultures. EXPERIMENTAL APPROACH Expression of TRPV1-4 channels was investigated at protein (immunocytochemistry, Western blot) and mRNA (Q-PCR) level in a conditionally immortalized human podocyte cell line. Channel function was assessed by measuring intracellular Ca2+ concentration using Flou-4 Ca2+ -indicator dye and patch clamp electrophysiology upon applying various activators and inhibitors. KEY RESULTS Thermosensitive TRP channels were expressed in podocytes. The TRPV1-specific agonists capsaicin and resiniferatoxin did not affect the intracellular Ca2+ concentration. Cannabidiol, an activator of TRPV2 and TRPV4 channels, induced moderate Ca2+ -influxes, inhibited by both tranilast and HC067047, blockers of TRPV2 and TRPV4 channels respectively. The TRPV4-specific agonists GSK1016790A and 4α-phorbol 12,13-didecanoate induced robust Ca2+ -signals which were abolished by HC067047. Non-specific agonists of TRPV3 channels induced marked Ca2+ transients. However, TRPV3 channel blockers, ruthenium red and isopentenyl diphosphate only partly inhibited the responses and TRPV3 silencing was ineffective suggesting remarkable off-target effects of the compounds. CONCLUSION AND IMPLICATIONS Our results indicate the functional presence of TRPV4 and other thermosensitive TRPV channels in human podocytes and raise the possibility of their involvement in the regulation of glomerular filtration barrier.
Collapse
Affiliation(s)
- Lídia Ambrus
- DE-MTA 'Lendület' Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Balázs Kelemen
- DE-MTA 'Lendület' Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Department of Pediatrics, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Tamás Bíró
- DE-MTA 'Lendület' Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary.,Department of Immunology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Balázs István Tóth
- DE-MTA 'Lendület' Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
31
|
Forstenpointner J, Förster M, May D, Hofschulte F, Cascorbi I, Wasner G, Gierthmühlen J, Baron R. Short Report: TRPV1-polymorphism 1911 A>G alters capsaicin-induced sensory changes in healthy subjects. PLoS One 2017; 12:e0183322. [PMID: 28817717 PMCID: PMC5560710 DOI: 10.1371/journal.pone.0183322] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/02/2017] [Indexed: 11/22/2022] Open
Abstract
Background C-fibers express transient receptor potential (TRP) channels. These high-voltage gated channels function as integrators of different physical stresses (e.g. heat, protons, ATP). Additionally channel activation can be induced by capsaicin. Topically applied, capsaicin elicits burning pain, heat and mechanical hyperalgesia and serves as a human surrogate model for pain. It was suggested that the TRPV1-variant rs8065080 (1911A>G) plays a pivotal role in patients with neuropathic pain syndromes. We investigated the effect of this TRPV1-SNP on thermal sensitivity and superficial skin perfusion in 25 healthy subjects. Methods and findings Nine subjects being homozygous TRPV1 wild type (AA), 8 heterozygous (AG) and 8 homozygous variant (GG) carriers were selected out of a pool of genotyped healthy individuals. Under physiological conditions (no capsaicin application), there was no statistical significant difference in thermal thresholds or skin perfusion between carriers of different TRPV1 1199A>G genotypes. However, intra-individual calculations (Δ% pre vs. post capsaicin) revealed (1) less warm-detection in AA/AG (-82.1%) compared to GG (-13.1%) and (2) a gain of heat pain sensitivity in AA/AG (+22.2%) compared to GG carriers (+15.6%) after adjustment for perfusion measurements ((1)p = 0.009, (2)p = 0.021). Conclusion Presence of homozygous variant TRPV1 genotype (GG) demonstrated less capsaicin-induced warm hypoesthesia in warm-detection and less capsaicin-induced heat pain sensitivity suggesting an altered channel function. This demonstrates not only the functional influence of TRPV1 rs8065080 polymorphism itself; it further more underpins the relevance of genotyping-based approaches in both patients and surrogate models of neuropathic pain in healthy volunteers.
Collapse
Affiliation(s)
- Julia Forstenpointner
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- * E-mail:
| | - Matti Förster
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Department of Neurology, Neurophysiology, Neuropsychology and Stroke Unit, Bogenhausen Hospital STKM, Munich, Germany
| | - Denisa May
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel Germany
| | - Friederike Hofschulte
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ingolf Cascorbi
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel Germany
| | - Gunnar Wasner
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Neurological Clinic, Kiel, Germany
| | - Janne Gierthmühlen
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
32
|
Bonior J, Warzecha Z, Ceranowicz P, Gajdosz R, Pierzchalski P, Kot M, Leja-Szpak A, Nawrot-Porąbka K, Link-Lenczowski P, Pędziwiatr M, Olszanecki R, Bartuś K, Trąbka R, Kuśnierz-Cabala B, Dembiński A, Jaworek J. Capsaicin-Sensitive Sensory Nerves Are Necessary for the Protective Effect of Ghrelin in Cerulein-Induced Acute Pancreatitis in Rats. Int J Mol Sci 2017; 18:E1402. [PMID: 28665321 PMCID: PMC5535895 DOI: 10.3390/ijms18071402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/25/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
Ghrelin was shown to exhibit protective and therapeutic effect in the gut. Aim of the study was to investigate the role of sensory nerves (SN) in the protective effect of ghrelin in acute pancreatitis (AP). Studies were performed on male Wistar rats or isolated pancreatic acinar cells. After capsaicin deactivation of sensory nerves (CDSN) or treatment with saline, rats were pretreated intraperitoneally with ghrelin or saline. In those rats, AP was induced by cerulein or pancreases were used for isolation of pancreatic acinar cells. Pancreatic acinar cells were incubated in cerulein-free or cerulein containing solution. In rats with intact SN, pretreatment with ghrelin led to a reversal of the cerulein-induced increase in pancreatic weight, plasma activity of lipase and plasma concentration of tumor necrosis factor-α (TNF-α). These effects were associated with an increase in plasma interleukin-4 concentration and reduction in histological signs of pancreatic damage. CDSN tended to increase the severity of AP and abolished the protective effect of ghrelin. Exposure of pancreatic acinar cells to cerulein led to increase in cellular expression of mRNA for TNF-α and cellular synthesis of this cytokine. Pretreatment with ghrelin reduced this alteration, but this effect was only observed in acinar cells obtained from rats with intact SN. Moreover, CDSN inhibited the cerulein- and ghrelin-induced increase in gene expression and synthesis of heat shock protein 70 (HSP70) in those cells. Ghrelin exhibits the protective effect in cerulein-induced AP on the organ and pancreatic acinar cell level. Sensory nerves ablation abolishes this effect.
Collapse
Affiliation(s)
- Joanna Bonior
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka St., 31-531 Krakow, Poland.
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka St., 31-531 Krakow, Poland.
| | - Ryszard Gajdosz
- Department of Emergency Medical Care, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Piotr Pierzchalski
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Michalina Kot
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Anna Leja-Szpak
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Katarzyna Nawrot-Porąbka
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Paweł Link-Lenczowski
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| | - Michał Pędziwiatr
- 2nd Department of Surgery, Faculty of Medicine, Jagiellonian University Medical College, 21 Kopernika St., 31-501 Krakow, Poland.
| | - Rafał Olszanecki
- Department of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka St., 31-531 Krakow, Poland.
| | - Krzysztof Bartuś
- Department of Cardiovascular Surgery and Transplantology, Faculty of Medicine, Jagiellonian University, JP II Hospital, 80 Prądnicka St., 31-202 Krakow, Poland.
| | - Rafał Trąbka
- Department of Rehabilitation, Faculty of Health Sciences, Jagiellonian University Medical College, 3 Koło Strzelnicy St., 30-219 Krakow, Poland.
| | - Beata Kuśnierz-Cabala
- Department of Diagnostics, Chair of Clinical Biochemistry, Faculty of Medicine Jagiellonian University Medical College, 15 A Kopernika St., 31-501 Krakow, Poland.
| | - Artur Dembiński
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka St., 31-531 Krakow, Poland.
| | - Jolanta Jaworek
- Department of Medical Physiology, Faculty of Health Sciences, Jagiellonian University Medical College, 12 Michałowskiego St., 31-126 Krakow, Poland.
| |
Collapse
|
33
|
Enhanced gap junction intercellular communication inhibits catabolic and pro-inflammatory responses in tenocytes against heat stress. J Cell Commun Signal 2017; 11:369-380. [PMID: 28601938 DOI: 10.1007/s12079-017-0397-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/28/2017] [Indexed: 12/17/2022] Open
Abstract
Elevation of tendon core temperature during severe activity is well known. However, its effects on tenocyte function have not been studied in detail. The present study tested a hypothesis that heat stimulation upregulates tenocyte catabolism, which can be modulated by the inhibition or the enhancement of gap junction intercellular communication (GJIC). Tenocytes isolated from rabbit Achilles tendons were subjected to heat stimulation at 37 °C, 41 °C or 43 °C for 30 min, and changes in cell viability, gene expressions and GJIC were examined. It was found that GJIC exhibited no changes by the stimulation even at 43 °C, but cell viability was decreased and catabolic and proinflammatory gene expressions were upregulated. Inhibition of GJIC demonstrated further upregulated catabolic and proinflammatory gene expressions. In contrast, enhanced GJIC, resulting from forced upregulation of connexin 43 gene, counteracted the heat-induced upregulation of catabolic and proinflammatory genes. These findings suggest that the temperature rise in tendon core could upregulate catabolic and proinflammatory activities, potentially leading to the onset of tendinopathy, and such upregulations could be suppressed by the enhancement of GJIC. Therefore, to prevent tendon injury at an early stage from becoming chronic injury, tendon core temperature and GJIC could be targets for post-activity treatments.
Collapse
|
34
|
Morales-Lázaro SL, Rosenbaum T. Multiple Mechanisms of Regulation of Transient Receptor Potential Ion Channels by Cholesterol. CURRENT TOPICS IN MEMBRANES 2017; 80:139-161. [DOI: 10.1016/bs.ctm.2017.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Aghazadeh Tabrizi M, Baraldi PG, Baraldi S, Gessi S, Merighi S, Borea PA. Medicinal Chemistry, Pharmacology, and Clinical Implications of TRPV1 Receptor Antagonists. Med Res Rev 2016; 37:936-983. [PMID: 27976413 DOI: 10.1002/med.21427] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/28/2022]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is an ion channel expressed on sensory neurons triggering an influx of cations. TRPV1 receptors function as homotetramers responsive to heat, proinflammatory substances, lipoxygenase products, resiniferatoxin, endocannabinoids, protons, and peptide toxins. Its phosphorylation increases sensitivity to both chemical and thermal stimuli, while desensitization involves a calcium-dependent mechanism resulting in receptor dephosphorylation. TRPV1 functions as a sensor of noxious stimuli and may represent a target to avoid pain and injury. TRPV1 activation has been associated to chronic inflammatory pain and peripheral neuropathy. Its expression is also detected in nonneuronal areas such as bladder, lungs, and cochlea where TRPV1 activation is responsible for pathology development of cystitis, asthma, and hearing loss. This review offers a comprehensive overview about TRPV1 receptor in the pathophysiology of chronic pain, epilepsy, cough, bladder disorders, diabetes, obesity, and hearing loss, highlighting how drug development targeting this channel could have a clinical therapeutic potential. Furthermore, it summarizes the advances of medicinal chemistry research leading to the identification of highly selective TRPV1 antagonists and their analysis of structure-activity relationships (SARs) focusing on new strategies to target this channel.
Collapse
Affiliation(s)
- Mojgan Aghazadeh Tabrizi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Giovanni Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Baraldi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Gessi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Stefania Merighi
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Pier Andrea Borea
- Section of Pharmacology, Department of Medical Sciences, University of Ferrara, 44121, Ferrara, Italy
| |
Collapse
|
36
|
Spatial temperature gradients guide axonal outgrowth. Sci Rep 2016; 6:29876. [PMID: 27460512 PMCID: PMC4962095 DOI: 10.1038/srep29876] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/24/2016] [Indexed: 02/06/2023] Open
Abstract
Formation of neural networks during development and regeneration after injury depends on accuracy of axonal pathfinding, which is primarily believed to be influenced by chemical cues. Recently, there is growing evidence that physical cues can play crucial role in axonal guidance. However, detailed mechanism involved in such guidance cues is lacking. By using weakly-focused near-infrared continuous wave (CW) laser microbeam in the path of an advancing axon, we discovered that the beam acts as a repulsive guidance cue. Here, we report that this highly-effective at-a-distance guidance is the result of a temperature field produced by the near-infrared laser light absorption. Since light absorption by extracellular medium increases when the laser wavelength was red shifted, the threshold laser power for reliable guidance was significantly lower in the near-infrared as compared to the visible spectrum. The spatial temperature gradient caused by the near-infrared laser beam at-a-distance was found to activate temperature-sensitive membrane receptors, resulting in an influx of calcium. The repulsive guidance effect was significantly reduced when extracellular calcium was depleted or in the presence of TRPV1-antagonist. Further, direct heating using micro-heater confirmed that the axonal guidance is caused by shallow temperature-gradient, eliminating the role of any non-photothermal effects.
Collapse
|
37
|
Witte DG, Cassar SC, Masters JN, Esbenshade T, Hancock AA. Use of a Fluorescent Imaging Plate Reader-Based Calcium Assay to Assess Pharmacological Differences between the Human and Rat Vanilloid Receptor. ACTA ACUST UNITED AC 2016; 7:466-75. [PMID: 14599363 DOI: 10.1177/108705702237679] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cloned vanilloid receptor 1 (VR1) is a ligand-gated calcium channel that is believed to be the capsaicin-activated vanilloid receptor found in native tissues, based on similarities regarding molecular mass, tissue distribution, and electrophysiological properties. Using a Fluorescent Imaging Plate Reader (FLIPR), along with Fluo-3 to signal intracellular calcium levels ([Ca++]i), rat VR1 (rVR1) and a human orthologue (hVR1) were pharmacologically characterized with various VR1 ligands. HEK-293 cells, stably expressing rVR1 or hVR1, exhibited dose-dependent increases in [Ca++]i when challenged with capsaicin (EC50s ≅ 10 nM). Responses to capsaicin were blocked by the VR1 antagonist capsazepine and were dependent on VR1 expression. Potencies for 10 structurally diverse VR1 agonists revealed rVR1 potencies highly correlated to that of hVR1 ( R2 = 0.973). However, a subset of agonists (tinyatoxin, gingerol, and zingerone) was approximately 10-fold more potent for rVR1 compared to hVR1. Schild analysis for blockade of capsaicin-induced responses by capsazepine was consistent with competitive antagonism, whereas ruthenium red displayed noncompetitive antagonism. Compared to rVR1, hVR1 was more sensitive to blockade by both antagonists. For both rVR1 and hVR1, time-response waveforms elicited by resiniferatoxin increased more gradually compared to other agonists. Tinyatoxin also displayed slow responses with hVR1 but showed rapid responses with rVR1. Thus, FLIPR technology can be used to readily reveal differences between rVR1 and hVR1 pharmacology with respect to potencies, efficacies, and kinetics for several VR1 ligands.
Collapse
Affiliation(s)
- David G Witte
- Global Pharmaceutical Research and Development, Neuroscience Research, Abbott Laboratories, Abbott Park, IL, USA.
| | | | | | | | | |
Collapse
|
38
|
Shin YH, Kim JM, Park K. The Effect of Capsaicin on Salivary Gland Dysfunction. Molecules 2016; 21:molecules21070835. [PMID: 27347918 PMCID: PMC6274068 DOI: 10.3390/molecules21070835] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 12/28/2022] Open
Abstract
Capsaicin (trans-8-methyl-N-vanilyl-6-nonenamide) is a unique alkaloid isolated from hot chili peppers of the capsicum family. Capsaicin is an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), which is expressed in nociceptive sensory neurons and a range of secretory epithelia, including salivary glands. Capsaicin has analgesic and anti-inflammatory properties in sensory neurons. Recently, increasing evidence has indicated that capsaicin also affects saliva secretion and inflammation in salivary glands. Applying capsaicin increases salivary secretion in human and animal models. Capsaicin appears to increase salivation mainly by modulating the paracellular pathway in salivary glands. Capsaicin activates TRPV1, which modulates the permeability of tight junctions (TJ) by regulating the expression and function of putative intercellular adhesion molecules in an ERK (extracelluar signal-regulated kinase) -dependent manner. Capsaicin also improved dysfunction in transplanted salivary glands. Aside from the secretory effects of capsaicin, it has anti-inflammatory effects in salivary glands. The anti-inflammatory effect of capsaicin is, however, not mediated by TRPV1, but by inhibition of the NF-κB pathway. In conclusion, capsaicin might be a potential drug for alleviating dry mouth symptoms and inflammation of salivary glands.
Collapse
Affiliation(s)
- Yong-Hwan Shin
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea.
| | - Jin Man Kim
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea.
| | - Kyungpyo Park
- Department of Physiology, School of Dentistry, Seoul National University and Dental Research Institute, Seoul 110-749, Korea.
| |
Collapse
|
39
|
Capsaicin, Nociception and Pain. Molecules 2016; 21:molecules21060797. [PMID: 27322240 PMCID: PMC6273518 DOI: 10.3390/molecules21060797] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/06/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
Capsaicin, the pungent ingredient of the hot chili pepper, is known to act on the transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1). TRPV1 is involved in somatic and visceral peripheral inflammation, in the modulation of nociceptive inputs to spinal cord and brain stem centers, as well as the integration of diverse painful stimuli. In this review, we first describe the chemical and pharmacological properties of capsaicin and its derivatives in relation to their analgesic properties. We then consider the biochemical and functional characteristics of TRPV1, focusing on its distribution and biological effects within the somatosensory and viscerosensory nociceptive systems. Finally, we discuss the use of capsaicin as an agonist of TRPV1 to model acute inflammation in slices and other ex vivo preparations.
Collapse
|
40
|
Rational design and validation of a vanilloid-sensitive TRPV2 ion channel. Proc Natl Acad Sci U S A 2016; 113:E3657-66. [PMID: 27298359 DOI: 10.1073/pnas.1604180113] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Vanilloids activation of TRPV1 represents an excellent model system of ligand-gated ion channels. Recent studies using cryo-electron microcopy (cryo-EM), computational analysis, and functional quantification revealed the location of capsaicin-binding site and critical residues mediating ligand-binding and channel activation. Based on these new findings, here we have successfully introduced high-affinity binding of capsaicin and resiniferatoxin to the vanilloid-insensitive TRPV2 channel, using a rationally designed minimal set of four point mutations (F467S-S498F-L505T-Q525E, termed TRPV2_Quad). We found that binding of resiniferatoxin activates TRPV2_Quad but the ligand-induced open state is relatively unstable, whereas binding of capsaicin to TRPV2_Quad antagonizes resiniferatoxin-induced activation likely through competition for the same binding sites. Using Rosetta-based molecular docking, we observed a common structural mechanism underlying vanilloids activation of TRPV1 and TRPV2_Quad, where the ligand serves as molecular "glue" that bridges the S4-S5 linker to the S1-S4 domain to open these channels. Our analysis revealed that capsaicin failed to activate TRPV2_Quad likely due to structural constraints preventing such bridge formation. These results not only validate our current working model for capsaicin activation of TRPV1 but also should help guide the design of drug candidate compounds for this important pain sensor.
Collapse
|
41
|
Vasconcelos LHC, Souza ILL, Pinheiro LS, Silva BA. Ion Channels in Obesity: Pathophysiology and Potential Therapeutic Targets. Front Pharmacol 2016; 7:58. [PMID: 27065858 PMCID: PMC4811910 DOI: 10.3389/fphar.2016.00058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/29/2016] [Indexed: 01/29/2023] Open
Abstract
Obesity is a multifactorial disease related to metabolic disorders and associated with genetic determinants. Currently, ion channels activity has been linked to many of these disorders, in addition to the central regulation of food intake, energetic balance, hormone release and response, as well as the adipocyte cell proliferation. Therefore, the objective of this work is to review the current knowledge about the influence of ion channels in obesity development. This review used different sources of literature (Google Scholar, PubMed, Scopus, and Web of Science) to assess the role of ion channels in the pathophysiology of obesity. Ion channels present diverse key functions, such as the maintenance of physiological homeostasis and cell proliferation. Cell biology and pharmacological experimental evidences demonstrate that proliferating cells exhibit ion channel expression, conductance, and electrical properties different from the resting cells. Thereby, a large variety of ion channels has been identified in the pathogenesis of obesity such as potassium, sodium, calcium and chloride channels, nicotinic acetylcholine receptor and transient receptor potential channels. The fundamental involvement of these channels on the generation of obesity leads to the progress in the knowledge about the mechanisms responsible for the obesity pathophysiology, consequently emerging as new targets for pharmacological modulation.
Collapse
Affiliation(s)
- Luiz H C Vasconcelos
- Laboratório de Farmacologia Funcional Prof. George Thomas, Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Iara L L Souza
- Laboratório de Farmacologia Funcional Prof. George Thomas, Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Lílian S Pinheiro
- Laboratório de Farmacologia Funcional Prof. George Thomas, Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Bagnólia A Silva
- Laboratório de Farmacologia Funcional Prof. George Thomas, Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da ParaíbaJoão Pessoa, Brazil; Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da ParaíbaJoão Pessoa, Brazil
| |
Collapse
|
42
|
Sanchez AM, Cioffi R, Viganò P, Candiani M, Verde R, Piscitelli F, Di Marzo V, Garavaglia E, Panina-Bordignon P. Elevated Systemic Levels of Endocannabinoids and Related Mediators Across the Menstrual Cycle in Women With Endometriosis. Reprod Sci 2016; 23:1071-9. [PMID: 26887427 DOI: 10.1177/1933719116630414] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cannabinoids and modulators of the endocannabinoid system affect specific mechanisms that are critical to the establishment and development of endometriosis. The aim of this study was to measure the systemic levels of endocannabinoids and related mediators in women with and without endometriosis and to investigate whether such levels correlated with endometriosis-associated pain. Plasma and endometrial biopsies were obtained from women with a laparoscopic diagnosis of endometriosis (n = 27) and no endometrial pathology (n = 29). Plasma levels of endocannabinoids (N-arachidonoylethanolamine [AEA] and 2-arachidonoylglycerol [2-AG]) and related mediators (N-oleoylethanolamine [OEA] and N-palmitoylethanolamine [PEA]), messenger RNA expression of some of their receptors (cannabinoid receptor type 1 [CB1], CB2, transient receptor potential vanilloid type [TRPV1]), and the enzymes involved in the synthesis (N-acyl-phosphatidylethanolamine-hydrolyzing phospholipase D [NAPE-PLD]) and degradation (fatty acid amide hydrolase 1 [FAAH]) of AEA, OEA, and PEA were evaluated in endometrial stromal cells. The systemic levels of AEA, 2-AG, and OEA were elevated in endometriosis in the secretory phase compared to controls. The expression of CB1 was higher in secretory phase endometrial stromal cells of controls versus endometriosis. Similar expression levels of CB2, TRPV1, NAPE-PLD, and FAAH were detected in controls and endometriosis. Patients with moderate-to-severe dysmenorrhea and dyspareunia showed higher AEA and PEA levels than those with low-to-moderate pain symptoms, respectively. The association of increased circulating AEA and 2-AG with decreased local CB1 expression in endometriosis suggests a negative feedback loop regulation, which may impair the capability of these mediators to control pain. These preliminary data suggest that the pharmacological manipulation of the action or levels of these mediators may offer an alternative option for the management of endometriosis-associated pain.
Collapse
Affiliation(s)
- Ana Maria Sanchez
- Division of Genetics and Cell Biology, Reproductive Sciences Laboratory, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Raffaella Cioffi
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Paola Viganò
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Massimo Candiani
- Department of Obstetrics and Gynecology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Roberta Verde
- Endocannabinoid Research Group, CNR, Pozzuoli, Italy
| | | | | | | | - Paola Panina-Bordignon
- Division of Genetics and Cell Biology, Reproductive Sciences Laboratory, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
43
|
Ferreira LGB, Faria RX. TRPing on the pore phenomenon: what do we know about transient receptor potential ion channel-related pore dilation up to now? J Bioenerg Biomembr 2016; 48:1-12. [PMID: 26728159 DOI: 10.1007/s10863-015-9634-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 12/02/2015] [Indexed: 01/06/2023]
Abstract
Ion channels allow for rapid ion diffusion through the plasma membrane. In some conditions, ion channels induce changes in the critical plasma membrane permeability that permit 900-Da solutes to enter cells. This process is known as the pore phenomenon. Some transient receptor potential (TRP) channel subtypes have been highlighted such as the P2X7 receptor, plasma membrane VDAC-1 channel, and pannexin hemichannels. The TRP ion channels are considered multimodal transducers that respond to several kinds of stimuli. In addition, many TRP channel subtypes are involved in physiological and pathophysiological processes such as inflammation, pain, and cancer. The TRPA1, TRPM8, and TRPV1-4 subtypes have been shown to promote large-molecular-weight solute uptake, including impermeable fluorescent dyes, QX-314 hydrophilic lidocaine derivative, gabapentin, and antineoplastic drugs. This review discusses the current knowledge of TRP-associated pores and encourages scientists to study their features and explore them as novel therapeutic tools.
Collapse
Affiliation(s)
- L G B Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900, Rio de Janeiro, Brazil.
| | - R X Faria
- Laboratory of Cellular Communication, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n° 4365, Manguinhos, CEP 21045-900, Rio de Janeiro, Brazil
| |
Collapse
|
44
|
Photosensitivity of neurons enabled by cell-targeted gold nanoparticles. Neuron 2015; 86:207-17. [PMID: 25772189 DOI: 10.1016/j.neuron.2015.02.033] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/18/2015] [Accepted: 02/14/2015] [Indexed: 01/05/2023]
Abstract
Unmodified neurons can be directly stimulated with light to produce action potentials, but such techniques have lacked localization of the delivered light energy. Here we show that gold nanoparticles can be conjugated to high-avidity ligands for a variety of cellular targets. Once bound to a neuron, these particles transduce millisecond pulses of light into heat, which changes membrane capacitance, depolarizing the cell and eliciting action potentials. Compared to non-functionalized nanoparticles, ligand-conjugated nanoparticles highly resist convective washout and enable photothermal stimulation with lower delivered energy and resulting temperature increase. Ligands targeting three different membrane proteins were tested; all showed similar activity and washout resistance. This suggests that many types of ligands can be bound to nanoparticles, preserving ligand and nanoparticle function, and that many different cell phenotypes can be targeted by appropriate choice of ligand. The findings have applications as an alternative to optogenetics and potentially for therapies involving neuronal photostimulation.
Collapse
|
45
|
Hsu E, Murphy S, Chang D, Cohen SP. Expert opinion on emerging drugs: chronic low back pain. Expert Opin Emerg Drugs 2014; 20:103-27. [DOI: 10.1517/14728214.2015.993379] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
46
|
Poblete H, Oyarzún I, Olivero P, Comer J, Zuñiga M, Sepulveda RV, Báez-Nieto D, González Leon C, González-Nilo F, Latorre R. Molecular determinants of phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) binding to transient receptor potential V1 (TRPV1) channels. J Biol Chem 2014; 290:2086-98. [PMID: 25425643 DOI: 10.1074/jbc.m114.613620] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) has been recognized as an important activator of certain transient receptor potential (TRP) channels. More specifically, TRPV1 is a pain receptor activated by a wide range of stimuli. However, whether or not PI(4,5)P2 is a TRPV1 agonist remains open to debate. Utilizing a combined approach of mutagenesis and molecular modeling, we identified a PI(4,5)P2 binding site located between the TRP box and the S4-S5 linker. At this site, PI(4,5)P2 interacts with the amino acid residues Arg-575 and Arg-579 in the S4-S5 linker and with Lys-694 in the TRP box. We confirmed that PI(4,5)P2 behaves as a channel agonist and found that Arg-575, Arg-579, and Lys-694 mutations to alanine reduce PI(4,5)P2 binding affinity. Additionally, in silico mutations R575A, R579A, and K694A showed that the reduction in binding affinity results from the delocalization of PI(4,5)P2 in the binding pocket. Molecular dynamics simulations indicate that PI(4,5)P2 binding induces conformational rearrangements of the structure formed by S6 and the TRP domain, which cause an opening of the lower TRPV1 channel gate.
Collapse
Affiliation(s)
- Horacio Poblete
- From the Center for Bioinformatics and Molecular Simulation, Universidad de Talca, 2 Norte 685, Talca-Chile
| | - Ingrid Oyarzún
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Pablo Olivero
- Escuela de Medicina, Universidad de Valparaíso, Hontaneda 2664, Valparaíso, Chile
| | - Jeffrey Comer
- Institute of Computational Comparative Medicine, Department of Anatomy and Physiology, Kansas State University, P-200 Mosier Hall, Manhattan, Kansas 66506-5802
| | - Matías Zuñiga
- Doctorado Fisicoquímica Molecular, Universidad Andrés Bello, Ave, Republica 275, Santiago, Chile
| | - Romina V Sepulveda
- Doctorado en Biotecnología, Universidad Andrés Bello, Av. Republica 217, Santiago, Chile, and Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. República 239, Santiago, Chile
| | - David Báez-Nieto
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Carlos González Leon
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile
| | - Fernando González-Nilo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile, Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Av. República 239, Santiago, Chile
| | - Ramón Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2366103, Chile,
| |
Collapse
|
47
|
Aguiar D, Moreira F, Terzian A, Fogaça M, Lisboa S, Wotjak C, Guimaraes F. Modulation of defensive behavior by Transient Receptor Potential Vanilloid Type-1 (TRPV1) Channels. Neurosci Biobehav Rev 2014; 46 Pt 3:418-28. [DOI: 10.1016/j.neubiorev.2014.03.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/04/2014] [Accepted: 03/18/2014] [Indexed: 12/20/2022]
|
48
|
Carlin KP, Wu G, Patel A, Crumley G, Ilyin VI. Phenylarsine oxide as a redox modulator of transient receptor potential vanilloid type 1 channel function. J Neurosci Res 2014; 93:309-20. [PMID: 25250537 DOI: 10.1002/jnr.23479] [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: 06/06/2014] [Revised: 07/24/2014] [Accepted: 07/26/2014] [Indexed: 11/09/2022]
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) channels are capable of detecting and integrating noxious stimuli and play an important role in nociceptor activation and sensitization. It has been demonstrated that oxidizing agents are capable of positively modulating (sensitizing) the TRPV1 channel. The present study investigates the ability of the thiol-oxidizing agent phenylarsine oxide (PAO) to modulate TRPV1 currents under voltage-clamp conditions. We assessed the ability of PAO to modulate both proton- and capsaicin-activated currents mediated by recombinant human TRPV1 channels as well as native rat and human TRPV1 channels in dorsal root ganglion (DRG) neurons. Experiments with other oxidizing and reducing agents having various membrane-permeating properties supported the intracellular oxidizing mechanism of PAO modulation. The PAO modulation of proton-activated currents was consistent across the cell types studied, with an increase in current across the proton concentrations studied. PAO modulation of the capsaicin-activated current in hTRPV1/Chinese hamster ovary cells consisted of potentiation of the current elicited with low capsaicin concentrations and inhibition of the current at higher concentrations. This same effect was seen with these recombinant cells in calcium imaging experiments and with native TRPV1 channels in rat DRG neurons. Contrary to this, currents in human DRG neurons were potentiated at all capsaicin concentrations tested after PAO treatment. These results could indicate important differences in the reduction-oxidation modulation of human TRPV1 channels in a native cellular environment.
Collapse
Affiliation(s)
- Kevin P Carlin
- Discovery Research, Purdue Pharma LP, Cranbury, New Jersey
| | | | | | | | | |
Collapse
|
49
|
Nash MS, Verkuyl JM, Bhalay G. TRPV1 Antagonism: From Research to Clinic. ION CHANNEL DRUG DISCOVERY 2014. [DOI: 10.1039/9781849735087-00186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The capsaicin receptor, TRPV1, has been one of the most extensively studied molecules in sensory research. Its contribution to the sensation of pain in numerous pre-clinical inflammatory and neuropathic paradigms has been well-established and expression analysis suggests a potential role clinically in pain and bladder conditions. The field has now reached an exciting point in time with the development of a number of high quality TRPV1 antagonist drug candidates and the release of clinical data. What has become apparent from this work is that inhibition of TRPV1 function brings with it the potential liabilities of increased body temperature and altered thermal perception. However, there is cause for optimism because it appears that not all antagonists have the same properties and compounds can be identified that lack significant on-target side-effects whilst retaining efficacy, at least pre-clinically. What is perhaps now more critical to address is the question of how effective the analgesia provided by a TRPV1 antagonist will be. Although tantalizing clinical data showing effects on experimentally-induced pain or pain following molar extraction have been reported, no clear efficacy in a chronic pain condition has yet been demonstrated making it difficult to perform an accurate risk-benefit analysis for TRPV1 antagonists. Here we provide an overview of some of the most advanced clinical candidates and discuss the approaches being taken to avoid the now well established on-target effects of TRPV1 antagonists.
Collapse
Affiliation(s)
- Mark S. Nash
- Novartis Institutes for Biomedical Research Forum 1, Novartis Campus CH - 4056 Basel Switzerland
| | - J. Martin Verkuyl
- Novartis Institutes for Biomedical Research Wimblehurst Road Horsham, West Sussex RH12 5AB UK
| | - Gurdip Bhalay
- Novartis Institutes for Biomedical Research Wimblehurst Road Horsham, West Sussex RH12 5AB UK
| |
Collapse
|
50
|
Abstract
TRPV1 is a well-characterised channel expressed by a subset of peripheral sensory neurons involved in pain sensation and also at a number of other neuronal and non-neuronal sites in the mammalian body. Functionally, TRPV1 acts as a sensor for noxious heat (greater than ~42 °C). It can also be activated by some endogenous lipid-derived molecules, acidic solutions (pH < 6.5) and some pungent chemicals and food ingredients such as capsaicin, as well as by toxins such as resiniferatoxin and vanillotoxins. Structurally, TRPV1 subunits have six transmembrane (TM) domains with intracellular N- (containing 6 ankyrin-like repeats) and C-termini and a pore region between TM5 and TM6 containing sites that are important for channel activation and ion selectivity. The N- and C- termini have residues and regions that are sites for phosphorylation/dephosphorylation and PI(4,5)P2 binding, which regulate TRPV1 sensitivity and membrane insertion. The channel has several interacting proteins, some of which (e.g. AKAP79/150) are important for TRPV1 phosphorylation. Four TRPV1 subunits form a non-selective, outwardly rectifying ion channel permeable to monovalent and divalent cations with a single-channel conductance of 50-100 pS. TRPV1 channel kinetics reveal multiple open and closed states, and several models for channel activation by voltage, ligand binding and temperature have been proposed. Studies with TRPV1 agonists and antagonists and Trpv1 (-/-) mice have suggested a role for TRPV1 in pain, thermoregulation and osmoregulation, as well as in cough and overactive bladder. TRPV1 antagonists have advanced to clinical trials where findings of drug-induced hyperthermia and loss of heat sensitivity have raised questions about the viability of this therapeutic approach.
Collapse
|