1
|
Drenjančević I, Jukić I, Đambić V, Stupin A, Kozina N, Matić A, Šušnjara P, Kibel A, Biljan D, Mihaljević Z. Variability in flow-induced vasodilation mechanisms in cerebral arteries: the impact of different hyperbaric oxygen protocols. Med Gas Res 2025; 15:383-390. [PMID: 39923134 PMCID: PMC12054677 DOI: 10.4103/mgr.medgasres-d-24-00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 02/10/2025] Open
Abstract
The present study aimed to assess the mechanisms of flow-induced dilation (FID) altered by acute/intermittent hyperbaric oxygenation (HBO 2 ) in isolated middle cerebral arteries of healthy male Sprague‒Dawley rats ( n = 96) and randomized to the Ac-HBO 2 group (exposed to a single HBO 2 session, 120 minutes of 100% O 2 at 2.0 bars), the 4Dys-HBO 2 group (4 consecutive days of single HBO 2 sessions, analyzed on the fifth day), and the CTRL (untreated) group. Results demonstrated increased vascular oxidative stress and decreased vascular nitric oxide bioavailability, as measured by direct fluorescence microscopy, leading to attenuated FID in the Ac-HBO 2 group compared with the CTRL and 4Dys-HBO 2 groups. Superoxide scavenging restored FID. Moreover, the increased expression of antioxidative enzymes in the cerebral vasculature in the 4Dys-HBO 2 group indicates the ability of intermittent HBO 2 to activate antioxidative mechanisms. Importantly, the results suggest a switch or at least activation of the compensatory mechanism of FID after HBO 2 from nitric oxide-dependent to epoxygenase metabolite-mediated via TRPV4 (transient receptor potential cation channel subfamily V member 4) and potassium channels, as demonstrated by increased protein expression of KCNMB1 (potassium calcium-activated channel subfamily M regulatory beta subunit 1), TRPV4, and Kir2 (a component of the inward rectifier-type potassium channel Kir2) in the vasculature. Overall, acute HBO 2 modulates FID in cerebral vessels by increasing oxidative stress and altering the subsequent mechanisms of FID, which are mainly mediated by nitric oxide, while suppressing potassium and TRPV4 channel function/expression due to increased oxidative stress. Moreover, intermittent HBO 2 activates antioxidative mechanisms and the compensatory mechanism of FID from nitric oxide-dependent to epoxygenase metabolite-mediated mechanisms via TRPV4, KCNMB1 and Kir2.1.
Collapse
Affiliation(s)
- Ines Drenjančević
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Ivana Jukić
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Vedran Đambić
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Ana Stupin
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Nataša Kozina
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Anita Matić
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Institute for Integrative Medicine, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Petar Šušnjara
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Faculty of Kinesiology Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Aleksandar Kibel
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- International Medical Center Priora, Čepin, Croatia
| | - Darko Biljan
- Department of Infectology and Dermatovenerology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Department of Dermatology and Venereology, Osijek University Hospital, Osijek, Croatia
| | - Zrinka Mihaljević
- Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| |
Collapse
|
2
|
Tihăuan BM, Onisei T, Slootweg W, Gună D, Iliescu C, Chifiriuc MC. Cannabidiol-A friend or a foe? Eur J Pharm Sci 2025; 208:107036. [PMID: 39929375 DOI: 10.1016/j.ejps.2025.107036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/23/2025]
Abstract
Cannabidiol (CBD), one of the main actives from Cannabis sativa has been perpetually explored lately for its therapeutic effects. Its main attributes, such as anti-inflammatory and antioxidant effects, snowball into pain management, epilepsy and seizure alleviation, anxiety relief, as well as numerous other implications through the entire metabolism. However, conventional administration routes challenge its therapeutic potential, with reported poor water solubility, hepatic degradation, gastric instability and erratic bioavailability observed in oral administration. As a result, the transdermal delivery systems have emerged as a promising alternative to oral or inhaled routes, offering improved bioavailability and targeted effects. The medical use of CBD throughout Europe, UK, USA or Australia is extensive and usually represented by pharmaceutical preparations recommended after conventional treatment routs fail. The non-medical use is limited by each country's own legislation, a wider range of products being available, but the irregular regulatory landscape coupled with the growing market of cannabinoid-infused products, emphasizes the need for standardized formulations and further clinical research. The present work critically examines the transdermal administration of cannabidiol, explores the skin's potential as a route and the strategies involved in using it for systemic targeting. We highlighted key challenges and provided insights into CBD`s variable bioavailability based on different administration routes and methods, thus compiling a literature-based absorption, distribution, metabolism, and excretion (ADME) study. We also explore the role of the endocannabinoid system, its function in various medical conditions, and the therapeutic effects associated with CBD, particularly in light of the varying legislation across countries. While the breadth of potential benefits is compelling, it is essential to emphasize the ongoing nature of CBD research as individual responses to it can vary significantly.
Collapse
Affiliation(s)
- Bianca-Maria Tihăuan
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania; National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania; eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania
| | - Tatiana Onisei
- National Institute for Research and Development in Food Bioresources, Dinu Vintilă Street, No.6, 021102 Bucharest, Romania
| | - Walter Slootweg
- QB3 Research & Development, Spaarndammerstraaat 4d, 1013SV Amsterdam, Netherlands
| | - Daniel Gună
- S.C. Absolute Essential Oils Ltd. (AEO), Adunații Copăceni Village, Giurgiu County, 38 Troitei Street, 087005, Romania
| | - Ciprian Iliescu
- eBio-hub Research Centre, National University for Science and Technology Politehnica Bucharest, Bucharest, Romania; Academy of Romanian Scientists, Bucharest, Romania; National Institute for Microtechnologies, 126A Erou Iancu Nicolae Street, Voluntari 077190, Romania.
| | - Mariana-Carmen Chifiriuc
- Research Institute of the University of Bucharest-ICUB, 91-95 Spl. Independentei, 50567 Bucharest, Romania.
| |
Collapse
|
3
|
Takayama Y. Interaction between thermosensitive TRP channels and anoctamin 1. J Physiol Sci 2025; 75:100015. [PMID: 40184917 PMCID: PMC11999596 DOI: 10.1016/j.jphyss.2025.100015] [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: 07/06/2024] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Some thermosensitive transient receptor potential (TRP) channels form a protein complex with anoctamin 1 (ANO1, also called TMEM16A). TRP channels have high calcium permeability, and the calcium entering cells through TRP channel activation activates ANO1, a calcium-activated chloride channel, involved in many physiological and pathological conditions. The physiological significance of TRP channels is often mediated by their ability to activate ANO1, which controls chloride flux across the plasma membrane. This review summarizes the latest understanding on the interactions between ANO1 and thermosensitive TRP channels, including TRPV1, TRPV3, and TRPV4, which are involved in pain sensitization in primary sensory neurons, proliferation and migration of human keratinocytes, and fluid secretion such as sweat, respectively.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, Japan.
| |
Collapse
|
4
|
Parthasarathy A, Anishkin A, Xie Y, Drachuk K, Nishijma Y, Fang J, Koukouritaki SB, Wilcox DA, Zhang DX. Phosphorylation of distal C-terminal residues promotes TRPV4 channel activation in response to arachidonic acid. J Biol Chem 2025; 301:108260. [PMID: 39909371 PMCID: PMC11903807 DOI: 10.1016/j.jbc.2025.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable channel activated by diverse physical and chemical stimuli, including mechanical stress and endogenous lipid arachidonic acid (AA) and its metabolites. Phosphorylation of TRPV4 by protein kinase A (PKA) and protein kinase C (PKC) is a predominant mechanism for channel regulation, especially in the cytoplasmic domains due to their importance in protein assembly, and channelopathies. However, studies corresponding to phosphorylation sites for these kinases remain incomplete. We investigated the role of Ser-823 residue as a potential phosphorylation site in regulating TRPV4 activity and chemical agonist-induced channel activation. Using mass spectrometry, we identified a new phosphorylation site Ser-823 residue and confirmed the previously known phosphorylation site Ser-824 in the C-terminal tail. The low level of phosphorylation at Ser-823 was stimulated by PKC and to a lesser extent by PKA in human coronary artery endothelial cells (HCAECs) and human embryonic kidney 293 (HEK 293) cells. AA-induced TRPV4 activation was enhanced in the phosphomimetic S823E but was blunted in the S823A/S824A mutants, whereas the channel activation by the synthetic agonist GSK1016790A was unaffected. Further, TRPV4 activation by AA but not GSK1016790A was blunted or abolished by PKA inhibitor alone or in combination with PKC inhibitor, respectively. Using computational modeling, we refined a previously proposed structural model for TRPV4 regulation by Ser-823 and Ser-824 phosphorylation. Together, these results provide insight into how stimuli-specific TRPV4 activation is regulated by the phosphorylation of discrete residues (e.g., Ser-823 and Ser-824) in the C-terminal domains of the TRPV4 channel.
Collapse
Affiliation(s)
- Aravind Parthasarathy
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, Maryland, USA
| | - Yangjing Xie
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kostiantyn Drachuk
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yoshinori Nishijma
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Sevasti B Koukouritaki
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - David A Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - David X Zhang
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
5
|
Meng MY, Paine LW, Sagnat D, Bello I, Oldroyd S, Javid F, Harper MT, Hockley JRF, St John Smith E, Owens RM, Alric L, Buscail E, Welsh F, Vergnolle N, Bulmer DC. TRPV4 stimulates colonic afferents through mucosal release of ATP and glutamate. Br J Pharmacol 2025; 182:1324-1340. [PMID: 39626870 DOI: 10.1111/bph.17408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Abdominal pain is a leading cause of morbidity for people living with gastrointestinal disease. Whereas the transient receptor potential vanilloid 4 (TRPV4) ion channel has been implicated in the pathogenesis of abdominal pain, the relative paucity of TRPV4 expression in colon-projecting sensory neurons suggests that non-neuronal cells may contribute to TRPV4-mediated nociceptor stimulation. EXPERIMENTAL APPROACH Changes in murine colonic afferent activity were examined using ex vivo electrophysiology in tissues with the gut mucosa present or removed. ATP and glutamate release were measured by bioluminescence assays from human colon organoid cultures and mouse colon. Dorsal root ganglion sensory neuron activity was evaluated by Ca2+ imaging when cultured alone or co-cultured with colonic mucosa. KEY RESULTS Bath application of TRPV4 agonist GSK1016790A elicited a robust increase in murine colonic afferent activity, which was abolished by removing the gut mucosa. GSK1016790A promoted ATP and glutamate release from human colon organoid cultures and mouse colon. Inhibition of ATP degradation in mouse colon enhanced the afferent response to GSK1016790A. Pretreatment with purinoceptor or glutamate receptor antagonists attenuated and abolished the response to GSK1016790A when given alone or in combination, respectively. Sensory neurons co-cultured with colonic mucosal cells produced a marked increase in intracellular Ca2+ to GSK1016790A compared with neurons cultured alone. CONCLUSION AND IMPLICATIONS Our data indicate that mucosal release of ATP and glutamate is responsible for the stimulation of colonic afferents following TRPV4 activation. These findings highlight an opportunity to target the gut mucosa for the development of new visceral analgesics.
Collapse
Affiliation(s)
- Michelle Y Meng
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Luke W Paine
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - David Sagnat
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Ivana Bello
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Sophie Oldroyd
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Farideh Javid
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | | | | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Laurent Alric
- Internal Medicine Department of Digestive Disease, CHU Toulouse-Rangueil and Université de Toulouse, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Etienne Buscail
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Surgery, CHU Toulouse-Rangueil and Université de Toulouse, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Fraser Welsh
- BioPharmaceuticals R&D, AstraZeneca, Neuroscience, Cambridge, UK
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
- Department of Physiology and Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - David C Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
6
|
Solano AS, Lavanderos B, Metwally E, Earley S. Transient Receptor Potential Channels in Vascular Mechanotransduction. Am J Hypertens 2025; 38:151-160. [PMID: 39579078 DOI: 10.1093/ajh/hpae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 11/25/2024] Open
Abstract
Transmural pressure and shear stress are mechanical forces that profoundly affect the smooth muscle cells (SMCs) comprising the vascular wall and the endothelial cells (ECs) lining the lumen. Pressure and flow are detected by mechanosensors in these cells and translated into appropriate responses to regulate blood pressure and flow. This review focuses on the role of the transient receptor potential (TRP) superfamily of cation channels in this process. We discuss how specific members of the TRP superfamily (TRPC6, TRPM4, TRPV1, TRPV4, and TRPP1) regulate the resting membrane and intracellular Ca2+ levels in SMCs and ECs to promote changes in vascular tone in response to intraluminal pressure and shear stress. Although TRP channels participate in vascular mechanotransduction, little evidence supports their intrinsic mechanosensitivity. Therefore, we also examine the evidence exploring the force-sensitive signal transduction pathways acting upstream of vascular TRP channels. Understanding the interplay between mechanosensors, force-induced signaling cascades, and TRP channels holds promise for the development of targeted therapies for diseases caused by vascular dysfunction.
Collapse
Affiliation(s)
- Alfredo Sanchez Solano
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Boris Lavanderos
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Elsayed Metwally
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Scott Earley
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
7
|
Hook JL, Kuebler WM. CFTR as a therapeutic target for severe lung infection. Am J Physiol Lung Cell Mol Physiol 2025; 328:L229-L238. [PMID: 39772994 DOI: 10.1152/ajplung.00289.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Lung infection is one of the leading causes of morbidity and mortality worldwide. Even with appropriate antibiotic and antiviral treatment, mortality in hospitalized patients often exceeds 10%, highlighting the need for the development of new therapeutic strategies. Of late, cystic fibrosis transmembrane conductance regulator (CFTR) is-in addition to its well-established roles in the lung airway and extrapulmonary organs-increasingly recognized as a key regulator of alveolar homeostasis and defense. In the alveolar epithelium, CFTR mediates alveolar fluid secretion and liquid homeostasis; in the microvascular endothelium, CFTR maintains vascular barrier function. CFTR also contributes to alveolar immunity. Yet, in lung infection, diverse molecular mechanisms reduce CFTR abundance and otherwise impair its function, promoting alveolar inflammation, edema, and cell death. Preservation or restoration of CFTR function by CFTR modulator drugs thus presents a promising avenue to combat lung infection in a pathogen-independent manner.
Collapse
Affiliation(s)
- Jaime L Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Turovsky EA, Plotnikov EY, Simakin AV, Gudkov SV, Varlamova EG. New magnetic iron nanoparticle doped with selenium nanoparticles and the mechanisms of their cytoprotective effect on cortical cells under ischemia-like conditions. Arch Biochem Biophys 2025; 764:110241. [PMID: 39613283 DOI: 10.1016/j.abb.2024.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
Ischemic stroke is the cause of high mortality and disability Worldwide. The material costs of stroke treatment and recovery are constantly increasing, making the search for effective and more cost-effective treatment approaches an urgent task for modern biomedicine. In this study, iron nanoparticles doped with selenium nanoparticles, FeNP@SeNPs, which are three-layered structures, were created and characterized using physical methods. Fluorescence microscopy, inhibitor and PCR analyzes were used to determine the signaling pathways involved in the activation of the Ca2+ signaling system of cortical astrocytes and the protection of cells from ischemia-like conditions (oxygen-glucose deprivation and reoxygenation). In particular, when using magnetic selenium nanoparticles together with electromagnetic stimulation, an additional pathway for nanoparticle penetration into the cell is activated through the activation of TRPV4 channels and the mechanism of their endocytosis is facilitated. It has been shown that the use of magnetic selenium nanoparticles together with magnetic stimulation represents an advantage over the use of classical selenium nanoparticles, as the effective concentration of nanoparticles can be reduced many times over.
Collapse
Affiliation(s)
- Egor A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| | - Egor Y Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia; V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997, Moscow, Russia
| | - Alexander V Simakin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State, University of Nizhni Novgorod, 23 Gagarin Ave., 603950, Nizhny Novgorod, Russia
| | - Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| |
Collapse
|
9
|
Qian SH, Liu S, Wang M, Wang Q, Hu CP, Huang JH, Zhang Z. Deficiency of endothelial TRPV4 cation channels ameliorates experimental abdominal aortic aneurysm. Eur J Pharmacol 2025; 986:177150. [PMID: 39577553 DOI: 10.1016/j.ejphar.2024.177150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA), albeit usually asymptomatic, is highly lethal if ruptured. The 28-member transient receptor potential (TRP) ion channel superfamily, most of which are present in the aortic cells, is understudied in AAA. We aim to identify single TRP channel that could represent a novel therapeutic target, and dissect dysfunctional ionic signaling that drives AAA. METHODS AAA was developed in mice by perfusing porcine pancreatic elastase into the infrarenal abdominal aorta. AAA was assessed by measurement of external diameter with a digital caliper, or internal diameter with ultrasonography. Aortic pathohistology was evaluated via histological and immunohistochemical staining. The TRP channel family was analyzed in the GSE17901 dataset. TRPC6, TRPC1/4/5 and TRPC3 channels were blocked in aneurysmal mice by BI749327, Pico145 and Pyr3, respectively. Endothelial cell-selective Trpv4 knockout mice were generated and leveraged for AAA analysis. TRPV4 channel was activated indirectly by TPPU or directly opened by GSK1016790A. RESULTS RNA-seq data mining revealed altered expression profiles of Trpc3/Trpc6, Trpv4. Pharmacological block of TRPC6, TRPC1/4/5 or TRPC3 did not influence AAA, whereas selective deletion of endothelial TRPV4 protected against AAA in endothelial cell-selective Trpv4 knockout mice. Indirect activation of TRPV4 by TPPU exacerbated AAA, but TRPV4-mediated nitric oxide signaling contributed minimally to AAA. TRPV4 activation promoted endothelial cell apoptosis in a Ca2+-dependent manner, a relevant mechanism underlying AAA. CONCLUSIONS Our data underscore the pathogenic importance of Ca2+ perturbation in AAA and illuminate that endothelial TRPV4 cation channel could be harnessed for AAA treatment.
Collapse
MESH Headings
- Animals
- TRPV Cation Channels/genetics
- TRPV Cation Channels/metabolism
- TRPV Cation Channels/deficiency
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Mice
- Mice, Knockout
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Humans
- Calcium/metabolism
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/drug effects
- Apoptosis/drug effects
Collapse
Affiliation(s)
- She-Hua Qian
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Shuai Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qing Wang
- Department of the Interventional Radiology & Vascular Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, Hunan, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410013, Hunan, China
| | - Jun-Hao Huang
- Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, Guangzhou, 510050, Guangdong, China.
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Allerkamp HH, Bondarenko AI, Tawfik I, Kamali-Simsek N, Horvat Mercnik M, Madreiter-Sokolowski CT, Wadsack C. In vitro examination of Piezo1-TRPV4 dynamics: implications for placental endothelial function in normal and preeclamptic pregnancies. Am J Physiol Cell Physiol 2025; 328:C227-C244. [PMID: 39652778 DOI: 10.1152/ajpcell.00794.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 01/11/2025]
Abstract
Mechanosensation is essential for endothelial cell (EC) function, which is compromised in early-onset preeclampsia (EPE), impacting offspring health. The ion channels Piezo-type mechanosensitive ion channel component 1 (Piezo1) and transient receptor potential cation channel subfamily V member 4 (TRPV4) are coregulated mechanosensors in ECs. Current evidence suggests that both channels could mediate aberrant placental endothelial function in EPE. Using isolated fetoplacental ECs (fpECs) from early control (EC) and EPE pregnancies, we show functional coexpression of both channels and that Ca2+ influx and membrane depolarization in response to chemical channel activation is reduced in EPE fpECs. Downstream of channel activation, Piezo1 alone can induce phosphorylation of endothelial nitric oxide synthase (eNOS) in fpECs, while combined activation of Piezo1 and TRPV4 only affects eNOS phosphorylation in EPE fpECs. Additionally, combined activation reduces the barrier integrity of fpECs and has a stronger effect on EPE fpECs. This implies altered Piezo1-TRPV4 coregulation in EPE. Mechanistically, we suggest this to be driven by changes in the arachidonic acid metabolism in EPE fpECs as identified by RNA sequencing. Targeting of Piezo1 and TRPV4 might hold potential for EPE treatment options in the future.NEW & NOTEWORTHY This study shows Piezo-type mechanosensitive ion channel component 1 (Piezo1) and transient receptor potential cation channel subfamily V member 4 (TRPV4) coexpression and functionality within primary human fetoplacental endothelial cells (fpECs), mediating nitric oxide (NO) production and barrier integrity. In early-onset preeclampsia (EPE), fpEC channel functionality and coregulation are impaired, affecting Ca2+ signaling and endothelial barrier function. Combined channel activation significantly reduces endothelial barrier integrity and increases NO production in EPE. Changes in arachidonic acid metabolism are suggested as a key underlying factor mediating impaired channel functionality in EPE fpECs.
Collapse
Affiliation(s)
- Hanna H Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | | - Ines Tawfik
- Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | | | | | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
11
|
Inoue K, Takenouchi S, Kida M, Kashio M, Tominaga M, Murata T. Transient receptor potential vanilloid 4 gene-deficiency attenuates the inhibitory effect of 5,6-dihydroxy-8Z,11Z,14Z,17Z-eicosatetraenoic acid on vascular permeability in mice. J Pharmacol Sci 2025; 157:35-38. [PMID: 39706643 DOI: 10.1016/j.jphs.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/23/2024] Open
Abstract
We investigated whether an anti-inflammatory lipid metabolite named 5,6-DiHETE reduces vascular permeability by inhibiting TRPV4 channels in vivo. In wild-type (WT) mice, histamine-induced dye extravasation was reduced by pre-administration of 5,6-DiHETE. In TRPV4-deficient mice, extravasation and histamine-induced edema were already reduced, and 5,6-DiHETE had no additional effect. In isolated WT aortas, 5,6-DiHETE attenuated acetylcholine-induced relaxation, but this effect was absent in TRPV4-deficient aortas. These findings suggest that 5,6-DiHETE reduces vascular permeability by inhibiting TRPV4 activity.
Collapse
Affiliation(s)
- Kotoha Inoue
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shinya Takenouchi
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Misato Kida
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Makiko Kashio
- Division of Cell Signaling, National Institute for Physiological Sciences, Aichi, Japan; Department of Cell Physiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, Aichi, Japan; Thermal Biology Research Group, Nagoya Advanced Research and Development Center, Nagoya City University, Aichi, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan; Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan; Food and Animal Systemics, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
12
|
Kashio M. Thermo-TRP regulation by endogenous factors and its physiological function at core body temperature. Physiol Rep 2025; 13:e70164. [PMID: 39793986 PMCID: PMC11723785 DOI: 10.14814/phy2.70164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/01/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025] Open
Abstract
Transient receptor potential (TRP) channels with temperature sensitivities (thermo-TRPs) are involved in various physiological processes. Thermo-TRPs that detect temperature changes in peripheral sensory neurons possess indispensable functions in thermosensation, eliciting defensive behavior against noxious temperatures and driving autonomic/behavioral thermoregulatory responses to maintain body temperature in mammals. Moreover, most thermo-TRPs are functionally expressed in cells and tissues where the temperature is maintained at a constant core body temperature. To perform physiological functions, the activity of each thermo-TRP channel must be regulated by endogenous mechanisms at body temperature. Dysregulation of this process can lead to various diseases. This review highlights the endogenous factors regulating thermo-TRP activity and physiological functions at constant core body temperature.
Collapse
Affiliation(s)
- Makiko Kashio
- Department of Cell PhysiologyKumamoto UniversityKumamotoJapan
| |
Collapse
|
13
|
Pham DL, Cox K, Ko ML, Ko GYP. Peptide Lv and Angiogenesis: A Newly Discovered Angiogenic Peptide. Biomedicines 2024; 12:2851. [PMID: 39767758 PMCID: PMC11672992 DOI: 10.3390/biomedicines12122851] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Peptide Lv is a small endogenous secretory peptide with ~40 amino acids and is highly conserved among certain several species. While it was first discovered that it augments L-type voltage-gated calcium channels (LTCCs) in neurons, thus it was named peptide "Lv", it can bind to vascular endothelial growth factor receptor 2 (VEGFR2) and has VEGF-like activities, including eliciting vasodilation and promoting angiogenesis. Not only does peptide Lv augment LTCCs in neurons and cardiomyocytes, but it also promotes the expression of intermediate-conductance KCa channels (KCa3.1) in vascular endothelial cells. Peptide Lv is upregulated in the retinas of patients with early proliferative diabetic retinopathy, a disease involving pathological angiogenesis. This review will provide an overview of peptide Lv, its known bioactivities in vitro and in vivo, and its clinical relevance, with a focus on its role in angiogenesis. As there is more about peptide Lv to be explored, this article serves as a foundation for possible future developments of peptide Lv-related therapeutics to treat or prevent diseases.
Collapse
Affiliation(s)
- Dylan L. Pham
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Kelsey Cox
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Michael L. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Biology, Division of Natural and Physical Sciences, Blinn College, Bryan, TX 77802, USA
| | - Gladys Y.-P. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
14
|
Hernández-Vega AM, García-Villegas R, Rosenbaum T. Roles for TRPV4 in disease: A discussion of possible mechanisms. Cell Calcium 2024; 124:102972. [PMID: 39609180 DOI: 10.1016/j.ceca.2024.102972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is a ubiquitously expressed Ca2+-permeable ion channel that controls intracellular calcium ([Ca2+]i) homeostasis in various types of cells. The physiological roles for TRPV4 are tissue specific and the mechanisms behind this specificity remain mostly unclarified. It is noteworthy that mutations in the TRPV4 channel have been associated to a broad spectrum of congenital diseases, with most of these mutations mainly resulting in gain-of-function. Mutations have been identified in human patients showing a variety of phenotypes and symptoms, mostly related to skeletal and neuromuscular disorders. Since TRPV4 is so widely expressed throughout the body, it comes as no surprise that the literature is growing in evidence linking this protein to malfunction in systems other than the skeletal and neuromuscular. In this review, we summarize the expression patterns of TRPV4 in several tissues and highlight findings of recent studies that address critical structural and functional features of this channel, particularly focusing on its interactions and signaling pathways related to Ca2+ entry. Moreover, we discuss the roles of TRPV4 mutations in some diseases and pinpoint some of the mechanisms underlying pathological states where TRPV4's malfunction is prominent.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México, 07360, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
15
|
Sánchez-Hernández R, Benítez-Angeles M, Hernández-Vega AM, Rosenbaum T. Recent advances on the structure and the function relationships of the TRPV4 ion channel. Channels (Austin) 2024; 18:2313323. [PMID: 38354101 PMCID: PMC10868539 DOI: 10.1080/19336950.2024.2313323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.
Collapse
Affiliation(s)
- Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Ana M. Hernández-Vega
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
16
|
Shibasaki K. TRPV4 activation by core body temperature has multimodal functions in the central nervous system. J Physiol Sci 2024; 74:55. [PMID: 39578735 PMCID: PMC11583650 DOI: 10.1186/s12576-024-00948-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: 06/30/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Brain temperature is strictly regulated by various endogenous mechanisms and significantly contributes to brain function in homeothermic animals, making it an important factor for health. Thermosensitive transient receptor potential (TRP) channels convert temperature information into electrical signals through cation influx. In particular, TRPV4 is involved in the regulation of brain function. TRPV4, constitutively active in neurons through its activation by brain temperature, increases neuronal firing. TRPV4KO mice have electroencephalogram abnormalities, resulting in depression-like and social behavioral abnormalities. This basic function of TRPV4, as a translator of brain temperature information, has been implicated in several diseases, including epilepsy and stress-induced depression. In addition to its neuronal functions, TRPV4 has many key functions in glia and vasculature that depend on brain temperature and contribute to brain activity. In this review, I summarize the importance of TRPV4 activities in relation to brain temperature and focus on how hyperthermia-induced TRPV4 dysfunction exacerbates brain diseases.
Collapse
Affiliation(s)
- Koji Shibasaki
- Laboratory of Neurochemistry, Department of Nutrition Science, University of Nagasaki, Nagasaki, 851-2195, Japan.
| |
Collapse
|
17
|
Liu D, Mao M, Liu W, Xie L, Zhong X, Cao W, Chen L. The Role of the TRPV4 Channel in Intestinal Physiology and Pathology. J Inflamm Res 2024; 17:9307-9317. [PMID: 39588136 PMCID: PMC11587805 DOI: 10.2147/jir.s483350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024] Open
Abstract
The transient receptor potential vanilloid 4 channel (TRPV4) is an important member of the TRP superfamily of cation channels. The channel can be activated by different physical and chemical stimuli, such as heat, osmotic, and mechanical stress. It regulates the release of nociceptive peptides (substance P and calcitonin gene-related peptide), and mediates neurogenic inflammation, which indicates the involvement of TRPV4 as a nociceptor. Previous studies show that TRPV4 regulates the contraction of intestinal smooth muscle, mucosal barrier permeability, intestinal ion transport, activation of submucosal enteric neurons, and generation of immune cells. TRPV4 is involved in various pathophysiological activities, and altered TRPV4 expression has been detected in some intestinal diseases (IBD, IBS, intestinal tumors, etc). Evidence indicates that TRPV4 plays a noxious role in intestinal barrier function when the intestine is damaged. This review focuses on the role of the TRPV4 channel in the physiological and pathological functions of the intestine, and evaluates the potential clinical significance to target TRPV4 channel in the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Mingli Mao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Wenjia Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Lihua Xie
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Xiaolin Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| |
Collapse
|
18
|
Mutafova-Yambolieva VN. Mechanosensitive release of ATP in the urinary bladder mucosa. Purinergic Signal 2024:10.1007/s11302-024-10063-6. [PMID: 39541058 DOI: 10.1007/s11302-024-10063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
The urinary bladder mucosa (urothelium and suburothelium/lamina propria) functions as a barrier between the content of the urine and the underlying bladder tissue. The bladder mucosa is also a mechanosensitive tissue that releases signaling molecules that affect functions of cells in the bladder wall interconnecting the mucosa with the detrusor muscle and the CNS. Adenosine 5'-triphosphate (ATP) is a primary mechanotransduction signal that is released from cells in the bladder mucosa in response to bladder wall distention and activates cell membrane-localized P2X and P2Y purine receptors on urothelial cells, sensory and efferent neurons, interstitial cells, and detrusor smooth muscle cells. The amounts of ATP at active receptor sites depend significantly on the amounts of extracellularly released ATP. Spontaneous and distention-induced release of ATP appear to be under differential control. This review is focused on mechanisms underlying urothelial release of ATP in response to mechanical stimulation. First, we present a brief overview of studies that report mechanosensitive ATP release in bladder cells or tissues. Then, we discuss experimental evidence for mechanosensitive release of urothelial ATP by vesicular and non-vesicular mechanisms and roles of the stretch-activated channels PIEZO channels, transient receptor potential vanilloid type 4, and pannexin 1. This is followed by brief discussion of possible involvement of calcium homeostasis modulator 1, acid-sensing channels, and connexins in the release of urothelial ATP. We conclude with brief discussion of limitations of current research and of needs for further studies to increase our understanding of mechanotransduction in the bladder wall and of purinergic regulation of bladder function.
Collapse
|
19
|
Guo Y, Wei X, Pei J, Yang H, Zheng XL. Dissecting the role of cannabinoids in vascular health and disease. J Cell Physiol 2024; 239:e31373. [PMID: 38988064 DOI: 10.1002/jcp.31373] [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: 02/29/2024] [Revised: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Cannabis, often recognized as the most widely used illegal psychoactive substance globally, has seen a shift in its legal status in several countries and regions for both recreational and medicinal uses. This change has brought to light new evidence linking cannabis consumption to various vascular conditions. Specifically, there is an association between cannabis use and atherosclerosis, along with conditions such as arteritis, reversible vasospasm, and incidents of aortic aneurysm or dissection. Recent research has started to reveal the mechanisms connecting cannabinoid compounds to atherosclerosis development. It is well known that the primary biological roles of cannabinoids operate through the activation of cannabinoid receptor types 1 and 2. Manipulation of the endocannabinoid system, either genetically or pharmacologically, is emerging as a promising approach to address metabolic dysfunctions related to obesity. Additionally, numerous studies have demonstrated the vasorelaxant properties and potential atheroprotective benefits of cannabinoids. In preclinical trials, cannabidiol is being explored as a treatment option for monocrotaline-induced pulmonary arterial hypertension. Although existing literature suggests a direct role of cannabinoids in the pathogenesis of atherosclerosis, the correlation between cannabinoids and other vascular diseases was only reported in some case series or observational studies, and its role and precise mechanisms remain unclear. Therefore, it is necessary to summarize and update previously published studies. This review article aims to summarize the latest clinical and experimental research findings on the relationship between cannabis use and vascular diseases. It also seeks to shed light on the potential mechanisms underlying these associations, offering a comprehensive view of current knowledge in this evolving field of study.
Collapse
Affiliation(s)
- Yanan Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xiaoyun Wei
- Department of Cardiology, The Fifth School of Clinical Medicine of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Junyu Pei
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Haibo Yang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [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: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
21
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
22
|
Orfali R, AlFaiz A, Alanazi M, Alabdulsalam R, Alharbi M, Alromaih Y, Dallak I, Alrahal M, Alwatban A, Saud R. TRPV4 Channel Modulators as Potential Drug Candidates for Cystic Fibrosis. Int J Mol Sci 2024; 25:10551. [PMID: 39408877 PMCID: PMC11476765 DOI: 10.3390/ijms251910551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in defective chloride ion channels. This leads to thick, dehydrated mucus that severely disrupts mucociliary clearance in the respiratory system and triggers infection that eventually is the cause of death of CF patients. Current therapeutic strategies primarily focus on restoring CFTR function, blocking epithelial sodium channels to prevent mucus dehydration, or directly targeting mucus to reduce its viscosity. Among the ion channels expressed in ciliated bronchial epithelial cells, the transient receptor potential vanilloid 4 (TRPV4) channel emerges as a significant channel in CF pathogenesis. Activation of TRPV4 channels affects the regulation of airway surface liquid by modulating sodium absorption and intracellular calcium levels, which indirectly influences CFTR activity. TRPV4 is also involved in the regulatory volume decrease (RVD) process and enhances inflammatory responses in CF patients. Here, we combine current findings on TRPV4 channel modulation as a promising therapeutic approach for CF. Although limited studies have directly explored TRPV4 in CF, emerging evidence indicates that TRPV4 activation can significantly impact key pathological processes in the disease. Further investigation into TRPV4 modulators could lead to innovative treatments that alleviate severe respiratory complications and improve outcomes for CF patients.
Collapse
Affiliation(s)
- Razan Orfali
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Ali AlFaiz
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Madhawi Alanazi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Rahaf Alabdulsalam
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Meaad Alharbi
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Yara Alromaih
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Ismail Dallak
- King Abdulaziz Medical City, Jeddah 9515, Saudi Arabia
| | - Marah Alrahal
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 12231, Saudi Arabia (M.A.)
| | - Abdulaziz Alwatban
- College of Medicine, Imam Mohammad Ibn Saud Islamic University, Riyadh 13317, Saudi Arabia
| | - Reem Saud
- General Education Department, Dar Al-Hikmah University, Jeddah 22246, Saudi Arabia
| |
Collapse
|
23
|
Amoakon JP, Lee J, Liyanage P, Arora K, Karlstaedt A, Mylavarapu G, Amin R, Naren AP. Defective CFTR modulates mechanosensitive channels TRPV4 and PIEZO1 and drives endothelial barrier failure. iScience 2024; 27:110703. [PMID: 39252977 PMCID: PMC11382128 DOI: 10.1016/j.isci.2024.110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by a mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Despite reports of CFTR expression on endothelial cells, pulmonary vascular perturbations, and perfusion deficits in CF patients, the mechanism of pulmonary vascular disease in CF remains unclear. Here, our pilot study of 40 CF patients reveals a loss of small pulmonary blood vessels in patients with severe lung disease. Using a vessel-on-a-chip model, we establish a shear-stress-dependent mechanism of endothelial barrier failure in CF involving TRPV4, a mechanosensitive channel. Furthermore, we demonstrate that CFTR deficiency downregulates the function of PIEZO1, another mechanosensitive channel involved in angiogenesis and wound repair, and exacerbates loss of small pulmonary blood vessel. We also show that CFTR directly interacts with PIEZO1 and enhances its function. Our study identifies key cellular targets to mitigate loss of small pulmonary blood vessels in CF.
Collapse
Affiliation(s)
- Jean-Pierre Amoakon
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jesun Lee
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pramodha Liyanage
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kavisha Arora
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anja Karlstaedt
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Goutham Mylavarapu
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Raouf Amin
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anjaparavanda P Naren
- Department of Systems Biology and Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Pulmonary Medicine and Critical Care, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
24
|
Sakaguchi R, Takahashi N, Yoshida T, Ogawa N, Ueda Y, Hamano S, Yamaguchi K, Sawamura S, Yamamoto S, Hara Y, Kawamoto T, Suzuki R, Nakao A, Mori MX, Furukawa T, Shimizu S, Inoue R, Mori Y. Dynamic remodeling of TRPC5 channel-caveolin-1-eNOS protein assembly potentiates the positive feedback interaction between Ca 2+ and NO signals. J Biol Chem 2024; 300:107705. [PMID: 39178948 PMCID: PMC11420454 DOI: 10.1016/j.jbc.2024.107705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/26/2024] Open
Abstract
The cell signaling molecules nitric oxide (NO) and Ca2+ regulate diverse biological processes through their closely coordinated activities directed by signaling protein complexes. However, it remains unclear how dynamically the multicomponent protein assemblies behave within the signaling complexes upon the interplay between NO and Ca2+ signals. Here we demonstrate that TRPC5 channels activated by the stimulation of G-protein-coupled ATP receptors mediate Ca2+ influx, that triggers NO production from endothelial NO synthase (eNOS), inducing secondary activation of TRPC5 via cysteine S-nitrosylation and eNOS in vascular endothelial cells. Mutations in the caveolin-1-binding domains of TRPC5 disrupt its association with caveolin-1 and impair Ca2+ influx and NO production, suggesting that caveolin-1 serves primarily as the scaffold for TRPC5 and eNOS to assemble into the signal complex. Interestingly, during ATP receptor activation, eNOS is dissociated from caveolin-1 and in turn directly associates with TRPC5, which accumulates at the plasma membrane dependently on Ca2+ influx and calmodulin. This protein reassembly likely results in a relief of eNOS from the inhibitory action of caveolin-1 and an enhanced TRPC5 S-nitrosylation by eNOS localized in the proximity, thereby facilitating the secondary activation of Ca2+ influx and NO production. In isolated rat aorta, vasodilation induced by acetylcholine was significantly suppressed by the TRPC5 inhibitor AC1903. Thus, our study provides evidence that dynamic remodeling of the protein assemblies among TRPC5, eNOS, caveolin-1, and calmodulin determines the ensemble of Ca2+ mobilization and NO production in vascular endothelial cells.
Collapse
Affiliation(s)
- Reiko Sakaguchi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Nobuaki Takahashi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan
| | - Takashi Yoshida
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Nozomi Ogawa
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yoshifumi Ueda
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Satoshi Hamano
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kaori Yamaguchi
- Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Seishiro Sawamura
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Shinichiro Yamamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Yuji Hara
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Department of Integrative Physiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tomoya Kawamoto
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Ryosuke Suzuki
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Akito Nakao
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Masayuki X Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Laboratory of Biomaterials and Chemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Tetsushi Furukawa
- Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shunichi Shimizu
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Tokyo, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University, Fukuoka, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan; Advanced Biomedical Engineering Research Unit, Kyoto University, Kyoto, Japan.
| |
Collapse
|
25
|
Baumann JM, Yarishkin O, Lakk M, De Ieso ML, Rudzitis CN, Kuhn M, Tseng YT, Stamer WD, Križaj D. TRPV4 and chloride channels mediate volume sensing in trabecular meshwork cells. Am J Physiol Cell Physiol 2024; 327:C403-C414. [PMID: 38881423 PMCID: PMC11427009 DOI: 10.1152/ajpcell.00295.2024] [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: 05/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Aqueous humor drainage from the anterior eye determines intraocular pressure (IOP) under homeostatic and pathological conditions. Swelling of the trabecular meshwork (TM) alters its flow resistance but the mechanisms that sense and transduce osmotic gradients remain poorly understood. We investigated TM osmotransduction and its role in calcium and chloride homeostasis using molecular analyses, optical imaging, and electrophysiology. Anisosmotic conditions elicited proportional changes in TM cell volume, with swelling, but not shrinking, evoking elevations in intracellular calcium concentration [Ca2+]TM. Hypotonicity-evoked calcium signals were sensitive to HC067047, a selective blocker of TRPV4 channels, whereas the agonist GSK1016790A promoted swelling under isotonic conditions. TRPV4 inhibition partially suppressed hypotonicity-induced volume increases and reduced the magnitude of the swelling-induced membrane current, with a substantial fraction of the swelling-evoked current abrogated by Cl- channel antagonists 4,4'-diisothiocyanato-2,2'-stilbenedisulfonic acid (DIDS) and niflumic acid. The transcriptome of volume-sensing chloride channel candidates in primary human was dominated by ANO6 transcripts, with moderate expression of ANO3, ANO7, and ANO10 transcripts and low expression of LTTRC genes that encode constituents of the volume-activated anion channel. Imposition of 190 mosM but not 285 mosM hypotonic gradients increased conventional outflow in mouse eyes. TRPV4-mediated cation influx thus works with Cl- efflux to sense and respond to osmotic stress, potentially contributing to pathological swelling, calcium overload, and intracellular signaling that could exacerbate functional disturbances in inflammatory disease and glaucoma.NEW & NOTEWORTHY Intraocular pressure is dynamically regulated by the flow of aqueous humor through paracellular passages within the trabecular meshwork (TM). This study shows hypotonic gradients that expand the TM cell volume and reduce the outflow facility in mouse eyes. The swelling-induced current consists of TRPV4 and chloride components, with TRPV4 as a driver of swelling-induced calcium signaling. TRPV4 inhibition reduced swelling, suggesting a novel treatment for trabeculitis and glaucoma.
Collapse
Affiliation(s)
- Jackson M Baumann
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah, United States
- Department of Bioengineering, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Oleg Yarishkin
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - Michael L De Ieso
- Duke Eye Center, Duke University, Durham, North Carolina, United States
| | | | - Megan Kuhn
- Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - Yun Ting Tseng
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah, United States
| | - W Daniel Stamer
- Duke Eye Center, Duke University, Durham, North Carolina, United States
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, Utah, United States
- Department of Bioengineering, University of Utah School of Medicine, Salt Lake City, Utah, United States
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
26
|
Hernández-Morales M, Morales-Weil K, Han SM, Han V, Tran T, Benner EJ, Pegram K, Meanor J, Miller EW, Kramer RH, Liu C. Electrophysiological Mechanisms and Validation of Ferritin-Based Magnetogenetics for Remote Control of Neurons. J Neurosci 2024; 44:e1717232024. [PMID: 38777598 PMCID: PMC11270515 DOI: 10.1523/jneurosci.1717-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Magnetogenetics was developed to remotely control genetically targeted neurons. A variant of magnetogenetics uses magnetic fields to activate transient receptor potential vanilloid (TRPV) channels when coupled with ferritin. Stimulation with static or RF magnetic fields of neurons expressing these channels induces Ca2+ transients and modulates behavior. However, the validity of ferritin-based magnetogenetics has been questioned due to controversies surrounding the underlying mechanisms and deficits in reproducibility. Here, we validated the magnetogenetic approach Ferritin-iron Redistribution to Ion Channels (FeRIC) using electrophysiological (Ephys) and imaging techniques. Previously, interference from RF stimulation rendered patch-clamp recordings inaccessible for magnetogenetics. We solved this limitation for FeRIC, and we studied the bioelectrical properties of neurons expressing TRPV4 (nonselective cation channel) and transmembrane member 16A (TMEM16A; chloride-permeable channel) coupled to ferritin (FeRIC channels) under RF stimulation. We used cultured neurons obtained from the rat hippocampus of either sex. We show that RF decreases the membrane resistance (Rm) and depolarizes the membrane potential in neurons expressing TRPV4FeRIC RF does not directly trigger action potential firing but increases the neuronal basal spiking frequency. In neurons expressing TMEM16AFeRIC, RF decreases the Rm, hyperpolarizes the membrane potential, and decreases the spiking frequency. Additionally, we corroborated the previously described biochemical mechanism responsible for RF-induced activation of ferritin-coupled ion channels. We solved an enduring problem for ferritin-based magnetogenetics, obtaining direct Ephys evidence of RF-induced activation of ferritin-coupled ion channels. We found that RF does not yield instantaneous changes in neuronal membrane potentials. Instead, RF produces responses that are long-lasting and moderate, but effective in controlling the bioelectrical properties of neurons.
Collapse
Affiliation(s)
- Miriam Hernández-Morales
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Koyam Morales-Weil
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Sang Min Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Victor Han
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Tiffany Tran
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Jenna Meanor
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, North Carolina 27710
| | - Evan W Miller
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
- Department of Chemistry, University of California, Berkeley, California 94720
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Richard H Kramer
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720
- Helen Wills Neuroscience Institute, University of California, Berkeley, California 94720
| |
Collapse
|
27
|
Fan J, Guo C, Liao D, Ke H, Lei J, Xie W, Tang Y, Tominaga M, Huang Z, Lei X. Structural Pharmacology of TRPV4 Antagonists. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401583. [PMID: 38659239 PMCID: PMC11220649 DOI: 10.1002/advs.202401583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/21/2024] [Indexed: 04/26/2024]
Abstract
The nonselective calcium-permeable Transient Receptor Potential Cation Channel Subfamily V Member4 (TRPV4) channel regulates various physiological activities. Dysfunction of TRPV4 is linked to many severe diseases, including edema, pain, gastrointestinal disorders, lung diseases, and inherited neurodegeneration. Emerging TRPV4 antagonists show potential clinical benefits. However, the molecular mechanisms of TRPV4 antagonism remain poorly understood. Here, cryo-electron microscopy (cryo-EM) structures of human TRPV4 are presented in-complex with two potent antagonists, revealing the detailed binding pockets and regulatory mechanisms of TRPV4 gating. Both antagonists bind to the voltage-sensing-like domain (VSLD) and stabilize the channel in closed states. These two antagonists induce TRPV4 to undergo an apparent fourfold to twofold symmetry transition. Moreover, it is demonstrated that one of the antagonists binds to the VSLD extended pocket, which differs from the canonical VSLD pocket. Complemented with functional and molecular dynamics simulation results, this study provides crucial mechanistic insights into TRPV4 regulation by small-molecule antagonists, which may facilitate future drug discovery targeting TRPV4.
Collapse
Affiliation(s)
- Junping Fan
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Chang Guo
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science CenterBeijing100191China
| | | | - Han Ke
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Jing Lei
- Division of Cell SignalingNational Institute for Physiological SciencesThermal Biology GroupExploratory Research Center on Life and Living SystemsNational Institutes of Natural SciencesOkazaki444‐8787Japan
| | - Wenjun Xie
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Yuliang Tang
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| | - Makoto Tominaga
- Division of Cell SignalingNational Institute for Physiological SciencesThermal Biology GroupExploratory Research Center on Life and Living SystemsNational Institutes of Natural SciencesOkazaki444‐8787Japan
- Nagoya Advanced Research and Developmet CenterNagoya City UniversityNagoya467‐8601Japan
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic DrugsDepartment of Molecular and Cellular PharmacologySchool of Pharmaceutical SciencesPeking University Health Science CenterBeijing100191China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular SciencesKey Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of EducationCollege of Chemistry and Molecular EngineeringPeking‐Tsinghua Center for Life SciencesPeking UniversityBeijing100871China
| |
Collapse
|
28
|
Takayama Y, Tominaga M. Interaction between TRP channels and anoctamins. Cell Calcium 2024; 121:102912. [PMID: 38823351 DOI: 10.1016/j.ceca.2024.102912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Anoctamin 1 (ANO1) binds to transient receptor potential (TRP) channels (protein-protein interaction) and then is activated by TRP channels (functional interaction). TRP channels are non-selective cation channels that are expressed throughout the body and play roles in multiple physiological functions. Studies on TRP channels increased after the identification of TRP vanilloid 1 (TRPV1) in 1997. Calcium-activated chloride channel anoctamin 1 (ANO1, also called TMEM16A and DOG1) was identified in 2008. ANO1 plays a major role in TRP channel-mediated functions, as first shown in 2014 with the demonstration of a protein-protein interaction between TRPV4 and ANO1. In cells that co-express TRP channels and ANO1, calcium entering cells through activated TRP channels causes ANO1 activation. Therefore, in many tissues, the physiological functions related to TRP channels are modulated through chloride flux associated with ANO1 activation. In this review, we summarize the latest understanding of TRP-ANO1 interactions, particularly interaction of ANO1 with TRPV4, TRP canonical 6 (TRPC6), TRPV3, TRPV1, and TRPC2 in the salivary glands, blood vessels, skin keratinocytes, primary sensory neurons, and vomeronasal organs, respectively.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, Japan.
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaiji, Okazaki, Aichi, Japan; Thermal Biology Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Aza-Higashiyama, Myodaiji, Okazaki, Aichi, Japan; Thermal Biology Research Group, Nagoya Advanced Research and Development Center, Nagoya City University, Kawasumi 1, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, Japan.
| |
Collapse
|
29
|
Zheng H, Kim M, Kim C, Kim Y, Cho PS, Lim JY, Lee H, Yun HI, Choi J, Hwang SW. GnRH peripherally modulates nociceptor functions, exacerbating mechanical pain. Front Mol Neurosci 2024; 17:1160435. [PMID: 38783903 PMCID: PMC11111891 DOI: 10.3389/fnmol.2024.1160435] [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: 02/07/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The function of peripheral nociceptors, the neurons that relay pain signals to the brain, are frequently tuned by local and systemic modulator substances. In this context, neurohormonal effects are emerging as an important modulatory mechanism, but many aspects remain to be elucidated. Here we report that gonadotropin-releasing hormone (GnRH), a brain-specific neurohormone, can aggravate pain by acting on nociceptors in mice. GnRH and GnRHR, the receptor for GnRH, are expressed in a nociceptor subpopulation. Administration of GnRH and its analogue, localized for selectively affecting the peripheral neurons, deteriorated mechanical pain, which was reproducible in neuropathic conditions. Nociceptor function was promoted by GnRH treatment in vitro, which appears to involve specific sensory transient receptor potential ion channels. These data suggest that peripheral GnRH can positively modulate nociceptor activities in its receptor-specific manner, contributing to pain exacerbation. Our study indicates that GnRH plays an important role in neurohormonal pain modulation via a peripheral mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sun Wook Hwang
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
30
|
Liu W, Deng W, Hu L, Zou H. Advances in TRPV6 inhibitors for tumors by targeted therapies: Macromolecular proteins, synthetic small molecule compounds, and natural compounds. Eur J Med Chem 2024; 270:116379. [PMID: 38588625 DOI: 10.1016/j.ejmech.2024.116379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 03/31/2024] [Indexed: 04/10/2024]
Abstract
TRPV6, a Ca2+-selective member of the transient receptor potential vanilloid (TRPV) family, plays a key role in extracellular calcium transport, calcium ion reuptake, and maintenance of a local low calcium environment. An increasing number of studies have shown that TRPV6 is involved in the regulation of various diseases. Notably, overexpression of TRPV6 is closely related to the occurrence of various cancers. Research confirmed that knocking down TRPV6 could effectively reduce the proliferation and invasiveness of tumors by mainly mediating the calcium signaling pathway. Hence, TRPV6 has become a promising new drug target for numerous tumor treatments. However, the development of TRPV6 inhibitors is still in the early stage, and the existing TRPV6 inhibitors have poor selectivity and off-target effects. In this review, we focus on summarizing and describing the structure characters, and mechanisms of existing TRPV6 inhibitors to provide new ideas and directions for the development of novel TRPV6 inhibitors.
Collapse
Affiliation(s)
- Weikang Liu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Wenwen Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China.
| | - Hui Zou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410013, China.
| |
Collapse
|
31
|
Hansen CE, Kamermans A, Mol K, Berve K, Rodriguez-Mogeda C, Fung WK, van Het Hof B, Fontijn RD, van der Pol SMA, Michalick L, Kuebler WM, Kenkhuis B, van Roon-Mom W, Liedtke W, Engelhardt B, Kooij G, Witte ME, de Vries HE. Inflammation-induced TRPV4 channels exacerbate blood-brain barrier dysfunction in multiple sclerosis. J Neuroinflammation 2024; 21:72. [PMID: 38521959 PMCID: PMC10960997 DOI: 10.1186/s12974-024-03069-9] [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: 12/21/2023] [Accepted: 03/18/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) dysfunction and immune cell migration into the central nervous system (CNS) are pathogenic drivers of multiple sclerosis (MS). Ways to reinstate BBB function and subsequently limit neuroinflammation present promising strategies to restrict disease progression. However, to date, the molecular players directing BBB impairment in MS remain poorly understood. One suggested candidate to impact BBB function is the transient receptor potential vanilloid-type 4 ion channel (TRPV4), but its specific role in MS pathogenesis remains unclear. Here, we investigated the role of TRPV4 in BBB dysfunction in MS. MAIN TEXT In human post-mortem MS brain tissue, we observed a region-specific increase in endothelial TRPV4 expression around mixed active/inactive lesions, which coincided with perivascular microglia enrichment in the same area. Using in vitro models, we identified that microglia-derived tumor necrosis factor-α (TNFα) induced brain endothelial TRPV4 expression. Also, we found that TRPV4 levels influenced brain endothelial barrier formation via expression of the brain endothelial tight junction molecule claudin-5. In contrast, during an inflammatory insult, TRPV4 promoted a pathological endothelial molecular signature, as evidenced by enhanced expression of inflammatory mediators and cell adhesion molecules. Moreover, TRPV4 activity mediated T cell extravasation across the brain endothelium. CONCLUSION Collectively, our findings suggest a novel role for endothelial TRPV4 in MS, in which enhanced expression contributes to MS pathogenesis by driving BBB dysfunction and immune cell migration.
Collapse
Grants
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 813294 European Union´s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant (ENTRAIN)
- 91719305 Dutch Research Council, NWO, Vidi grant
- 91719305 Dutch Research Council, NWO, Vidi grant
- 91719305 Dutch Research Council, NWO, Vidi grant
- 18-1023MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 18-1023MS Stichting MS Research
- 20-1106MS Stichting MS Research
- 81X3100216 Deutsches Zentrum für Herz-Kreislaufforschung
- SFB-TR84 : subprojects A02 & C09, SFB-1449 subproject B01, SFB 1470 subproject A04, KU1218/9-1, KU1218/11-1, and KU1218/12-1 Deutsche Forschungsgemeinschaft
- PROVID (01KI20160A) and SYMPATH (01ZX1906A) Bundesministerium für Bildung und Forschung
- HA2016-02-02 Hersenstichting
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Kevin Mol
- Department of Biomedical Engineering and Physics, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Kristina Berve
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Carla Rodriguez-Mogeda
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Wing Ka Fung
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Ruud D Fontijn
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Laura Michalick
- Institute of Physiology, Corporate member of the Freie Universität Berlin and Humboldt Universität to Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Corporate member of the Freie Universität Berlin and Humboldt Universität to Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| | - Boyd Kenkhuis
- Department of Human Genetics, Leiden University Medical Center Leiden, Leiden, The Netherlands
- UK Dementia Research Institute at University of Edinburgh, Edinburgh, UK
| | - Willeke van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center Leiden, Leiden, The Netherlands
| | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, NY, USA
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, USA
| | | | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maarten E Witte
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Mao A, Zhang K, Kan H, Gao M, Wang Z, Zhou T, Shao J, He D. Single-Cell RNA-Seq Reveals Coronary Heterogeneity and Identifies CD133 +TRPV4 high Endothelial Subpopulation in Regulating Flow-Induced Vascular Tone in Mice. Arterioscler Thromb Vasc Biol 2024; 44:653-665. [PMID: 38269590 PMCID: PMC10880935 DOI: 10.1161/atvbaha.123.319516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Single-cell RNA-Seq analysis can determine the heterogeneity of cells between different tissues at a single-cell level. Coronary artery endothelial cells (ECs) are important to coronary blood flow. However, little is known about the heterogeneity of coronary artery ECs, and cellular identity responses to flow. Identifying endothelial subpopulations will contribute to the precise localization of vascular endothelial subpopulations, thus enabling the precision of vascular injury treatment. METHODS Here, we performed a single-cell RNA sequencing of 31 962 cells and functional assays of 3 branches of the coronary arteries (right coronary artery/circumflex left coronary artery/anterior descending left coronary artery) in wild-type mice. RESULTS We found a compendium of 7 distinct cell types in mouse coronary arteries, mainly ECs, granulocytes, cardiac myocytes, smooth muscle cells, lymphocytes, myeloid cells, and fibroblast cells, and showed spatial heterogeneity between arterial branches. Furthermore, we revealed a subpopulation of coronary artery ECs, CD133+TRPV4high ECs. TRPV4 (transient receptor potential vanilloid 4) in CD133+TRPV4high ECs is important for regulating vasodilation and coronary blood flow. CONCLUSIONS Our study elucidates the nature and range of coronary arterial cell diversity and highlights the importance of coronary CD133+TRPV4high ECs in regulating coronary vascular tone.
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
- School of Food Science and Technology (A.M., D.H.), Jiangnan University, China
| | - Ka Zhang
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Hao Kan
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Mengru Gao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Zhiwei Wang
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Tingting Zhou
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Jing Shao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Dongxu He
- School of Food Science and Technology (A.M., D.H.), Jiangnan University, China
| |
Collapse
|
33
|
Jing L, Liu K, Wang F, Su Y. Role of mechanically-sensitive cation channels Piezo1 and TRPV4 in trabecular meshwork cell mechanotransduction. Hum Cell 2024; 37:394-407. [PMID: 38316716 DOI: 10.1007/s13577-024-01035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Glaucoma is one of the leading causes of irreversible blindness in developed countries, and intraocular pressure (IOP) is primary and only treatable risk factor, suggesting that to a significant extent, glaucoma is a disease of IOP disorder and pathological mechanotransduction. IOP-lowering ways are limited to decreaseing aqueous humour (AH) production or increasing the uveoscleral outflow pathway. Still, therapeutic approaches have been lacking to control IOP by enhancing the trabecular meshwork (TM) pathway. Trabecular meshwork cells (TMCs) have endothelial and myofibroblast properties and are responsible for the renewal of the extracellular matrix (ECM). Mechanosensitive cation channels, including Piezo1 and TRPV4, are abundantly expressed in primary TMCs and trigger mechanostress-dependent ECM and cytoskeletal remodelling. However, prolonged mechanical stimulation severely affects cellular biosynthesis through TMC mechanotransduction, including signaling, gene expression, ECM remodelling, and cytoskeletal structural changes, involving outflow facilities and elevating IOP. As for the functional coupling relationship between Piezo1 and TRPV4 channels, inspired by VECs and osteoblasts, we hypothesized that Piezo1 may also act upstream of TRPV4 in glaucomatous TM tissue, mediating the activation of TRPV4 via Ca2+ inflow or Ca2+ binding to phospholipase A2(PLA2), and thus be involved in increasing TM outflow resistance and elevated IOP. Therefore, this review aims to help identify new potential targets for IOP stabilization in ocular hypertension and primary open-angle glaucoma by understanding the mechanical transduction mechanisms associated with the development of glaucoma and may provide ideas into novel treatments for preventing the progression of glaucoma by targeting mechanotransduction.
Collapse
Affiliation(s)
- Lingling Jing
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Kexin Liu
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Feng Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Ying Su
- Eye Hospital, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
34
|
Abstract
Mechanical forces influence different cell types in our bodies. Among the earliest forces experienced in mammals is blood movement in the vascular system. Blood flow starts at the embryonic stage and ceases when the heart stops. Blood flow exposes endothelial cells (ECs) that line all blood vessels to hemodynamic forces. ECs detect these mechanical forces (mechanosensing) through mechanosensors, thus triggering physiological responses such as changes in vascular diameter. In this review, we focus on endothelial mechanosensing and on how different ion channels, receptors, and membrane structures detect forces and mediate intricate mechanotransduction responses. We further highlight that these responses often reflect collaborative efforts involving several mechanosensors and mechanotransducers. We close with a consideration of current knowledge regarding the dysregulation of endothelial mechanosensing during disease. Because hemodynamic disruptions are hallmarks of cardiovascular disease, studying endothelial mechanosensing holds great promise for advancing our understanding of vascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine and Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont, USA;
| |
Collapse
|
35
|
Cherry AL, Wheeler MJ, Mathisova K, Di Miceli M. In silico analyses of the involvement of GPR55, CB1R and TRPV1: response to THC, contribution to temporal lobe epilepsy, structural modeling and updated evolution. Front Neuroinform 2024; 18:1294939. [PMID: 38404644 PMCID: PMC10894036 DOI: 10.3389/fninf.2024.1294939] [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: 09/28/2023] [Accepted: 01/19/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction The endocannabinoid (eCB) system is named after the discovery that endogenous cannabinoids bind to the same receptors as the phytochemical compounds found in Cannabis. While endogenous cannabinoids include anandamide (AEA) and 2-arachidonoylglycerol (2-AG), exogenous phytocannabinoids include Δ-9 tetrahydrocannabinol (THC) and cannabidiol (CBD). These compounds finely tune neurotransmission following synapse activation, via retrograde signaling that activates cannabinoid receptor 1 (CB1R) and/or transient receptor potential cation channel subfamily V member 1 (TRPV1). Recently, the eCB system has been linked to several neurological diseases, such as neuro-ocular abnormalities, pain insensitivity, migraine, epilepsy, addiction and neurodevelopmental disorders. In the current study, we aim to: (i) highlight a potential link between the eCB system and neurological disorders, (ii) assess if THC exposure alters the expression of eCB-related genes, and (iii) identify evolutionary-conserved residues in CB1R or TRPV1 in light of their function. Methods To address this, we used several bioinformatic approaches, such as transcriptomic (Gene Expression Omnibus), protein-protein (STRING), phylogenic (BLASTP, MEGA) and structural (Phyre2, AutoDock, Vina, PyMol) analyzes. Results Using RNA sequencing datasets, we did not observe any dysregulation of eCB-related transcripts in major depressive disorders, bipolar disorder or schizophrenia in the anterior cingulate cortex, nucleus accumbens or dorsolateral striatum. Following in vivo THC exposure in adolescent mice, GPR55 was significantly upregulated in neurons from the ventral tegmental area, while other transcripts involved in the eCB system were not affected by THC exposure. Our results also suggest that THC likely induces neuroinflammation following in vitro application on mice microglia. Significant downregulation of TPRV1 occurred in the hippocampi of mice in which a model of temporal lobe epilepsy was induced, confirming previous observations. In addition, several transcriptomic dysregulations were observed in neurons of both epileptic mice and humans, which included transcripts involved in neuronal death. When scanning known interactions for transcripts involved in the eCB system (n = 12), we observed branching between the eCB system and neurophysiology, including proteins involved in the dopaminergic system. Our protein phylogenic analyzes revealed that CB1R forms a clade with CB2R, which is distinct from related paralogues such as sphingosine-1-phosphate, receptors, lysophosphatidic acid receptors and melanocortin receptors. As expected, several conserved residues were identified, which are crucial for CB1R receptor function. The anandamide-binding pocket seems to have appeared later in evolution. Similar results were observed for TRPV1, with conserved residues involved in receptor activation. Conclusion The current study found that GPR55 is upregulated in neurons following THC exposure, while TRPV1 is downregulated in temporal lobe epilepsy. Caution is advised when interpreting the present results, as we have employed secondary analyzes. Common ancestors for CB1R and TRPV1 diverged from jawless vertebrates during the late Ordovician, 450 million years ago. Conserved residues are identified, which mediate crucial receptor functions.
Collapse
Affiliation(s)
- Amy L. Cherry
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| | - Michael J. Wheeler
- Sustainable Environments Research Group, School of Science and the Environment University of Worcester, Worcester, United Kingdom
| | - Karolina Mathisova
- School of Science and the Environment University of Worcester, Worcester, United Kingdom
| | - Mathieu Di Miceli
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Worcester, United Kingdom
| |
Collapse
|
36
|
Li Y, Li L, Wei S, Yao J, Liang B, Chu X, Wang L, Liu H, Liao D, Liu D, Jiang P. Integrating transcriptomics and metabolomics to elucidate the mechanism by which taurine protects against DOX-induced depression. Sci Rep 2024; 14:2686. [PMID: 38302509 PMCID: PMC10834502 DOI: 10.1038/s41598-023-51138-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/31/2023] [Indexed: 02/03/2024] Open
Abstract
Doxorubicin (DOX) is an effective anticancer drug with potent antitumour activity. However, the application of DOX is limited by its adverse reactions, such as depression. Taurine can alleviate depression induced by multiple factors. However, it is still unclear whether and how taurine improves DOX-induced depression. To address this question, the aim of this study was to explore the potential mechanism by which taurine protects against DOX-induced depression. Mice were randomly divided into three groups (n = 8): (1) the control group, (2) the DOX group, and (3) the DOX + taurine group. The open field test (OFT), elevated plus maze test, and forced swim test (FST) were first performed to assess the effects of DOX and taurine on the behaviour of mice. Next, a combined transcriptomic and metabolomic analysis was performed to analyse the possible antidepressive effect of taurine. Taurine pretreatment increased the total distance travelled and speed of mice in the OFT, increased the number of entries into the open arm and the time spent in the open arm, and reduced the immobility time in the FST. In addition, 179 differential genes and 51 differentially abundant metabolites were detected in the DOX + taurine group compared to the DOX group. Furthermore, differential genes and differentially abundant metabolites were found to be jointly involved in 21 pathways, which may be closely related to the antidepressant effect of taurine. Taurine alleviated DOX-induced depressive behaviour. The various pathways identified in this study, such as the serotonergic synapse and the inflammatory mediator regulation of TRP channels, may be key regulatory pathways related to depression and antidepressant effects.
Collapse
Affiliation(s)
- Yanan Li
- College of Marine Life Sciences, Ocean University of China, No.5 Yushan Road, Qingdao, 266003, China
| | - Luxi Li
- College of Marine Life Sciences, Ocean University of China, No.5 Yushan Road, Qingdao, 266003, China
| | - Shanshan Wei
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Graduate, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, China
| | - Jia Yao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, China
| | - Benhui Liang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, China
| | - Lei Wang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, China
| | - Hui Liu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, China
| | - Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Daotong Liu
- Department of Breast and Thyroid Surgery, Jining First People's Hospital, Shandong First Medical University, Jining, China.
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, China.
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China.
| |
Collapse
|
37
|
Appendino G, Gaeta S. Tigliane Diterpenoids. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2024; 125:1-189. [PMID: 39546131 DOI: 10.1007/978-3-031-67180-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The distribution, chemistry, and molecular bioactivity of tiglianes are reviewed from the very beginning of the studies on these diterpenoids, summarizing their clinical and toxicological literature mostly in its more recent and controversial aspects, and critically analyzing various proposals for their biosynthesis.
Collapse
Affiliation(s)
- Giovanni Appendino
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Largo Donegani, 2, 28100, Novara, Italy.
| | - Simone Gaeta
- Research & Development-Chemistry Research, QBiotics Group Limited, 165, Moggill Road, Taringa, QLD, 4068, Australia
| |
Collapse
|
38
|
Shibasaki K. Regulation of Neural Functions by Brain Temperature and Thermo-TRP Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1461:199-211. [PMID: 39289283 DOI: 10.1007/978-981-97-4584-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Body temperature is an important determinant in regulating the activities of animals. In humans, a mild 0.5 °C hyperthermia can cause headaches, demonstrating that the maintenance of normal body temperature is a key for our health. In a more extreme example, accidental acute hypothermia can lead to severe shivering, loss of consciousness, or death, although the details of these mechanisms are poorly understood. We previously found that the TRPV4 ion channel is constitutively activated by normal body temperature. The activation threshold of TRPV4 is >34 °C in the brain, which enables TRPV4 to convert thermal information into cellular signaling. Here we review the data that describe how the deletion of TRPV4 evokes abnormal behavior in mice. These studies demonstrate that the maintenance of body temperature and the sensory system for detecting body temperature, such as via TRPV4, are critical components for normal cellular function. Moreover, abnormal TRPV4 activation exacerbates cell death, epilepsy, stroke, or brain edema. Notably, TRPV4 can detect mechanical stimuli and contributes to various neural functions similar to the mechanosensitive characteristics of TRPV2. In this review, I summarize the findings related to TRPV2/TRPV4 and neural functions.
Collapse
Affiliation(s)
- Koji Shibasaki
- Laboratory of Neurochemistry, Department of Nutrition Science, University of Nagasaki, Nagasaki, Japan.
| |
Collapse
|
39
|
Matsumoto T, Taguchi K, Kobayashi T. Role of TRPV4 on vascular tone regulation in pathophysiological states. Eur J Pharmacol 2023; 959:176104. [PMID: 37802278 DOI: 10.1016/j.ejphar.2023.176104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023]
Abstract
Vascular tone regulation is a key event in controlling blood flow in the body. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) help regulate the vascular tone. Abnormal vascular responsiveness to various stimuli, including constrictors and dilators, has been observed in pathophysiological states although EC and VSMC coordinate to maintain the exquisite balance between contraction and relaxation in vasculatures. Thus, investigating the mechanisms underlying vascular tone abnormality is very important in maintaining vascular health and treating vasculopathy. Increased intracellular free Ca2+ concentration ([Ca2+]i) is one of the major triggers initiating each EC and VSMC response. Transient receptor potential vanilloid family member 4 (TRPV4) is a Ca2+-permeable non-selective ion channel, which is activated by several stimuli, and is presented in both ECs and VSMCs. Therefore, TRPV4 plays an important role in vascular responses. Emerging evidence indicates the role of TRPV4 on the functions of ECs and VSMCs in various pathophysiological states, including hypertension, diabetes, and obesity. This review focused on the link between TRPV4 and the functions of ECs/VSMCs, particularly its role in vascular tone and responsiveness to vasoactive substances.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Pharmaceutical Education and Research, Pharmaceutical Education and Research Center, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan.
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| |
Collapse
|
40
|
Huang J, Chen J. Hydrophobic gating in bundle-crossing ion channels: a case study of TRPV4. Commun Biol 2023; 6:1094. [PMID: 37891195 PMCID: PMC10611814 DOI: 10.1038/s42003-023-05471-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Transmembrane ion channels frequently regulate ion permeation by forming bundle crossing of the pore-lining helices when deactivated. The resulting physical constriction is believed to serve as the de facto gate that imposes the major free energy barrier to ion permeation. Intriguingly, many ion channels also contain highly hydrophobic inner pores enclosed by bundle crossing, which can undergo spontaneous dewetting and give rise to a "vapor barrier" to block ion flow even in the absence of physical constriction. Using atomistic simulations, we show that hydrophobic gating and bundle-crossing mechanisms co-exist and complement one and another in the human TRPV4 channel. In particular, a single hydrophilic mutation in the lower pore can increase pore hydration and reduce the ion permeation free energy barrier by about half without affecting the bundle crossing. We believe that hydrophobic gating may play a key role in other bundle-crossing ion channels with hydrophobic inner pores.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Jianhan Chen
- Department of Chemistry University of Massachusetts Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
41
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
42
|
Wen X, Peng Y, Peng Y, Zhu Y, Yu F, Geng L, Zhou T, Wang X, Feng L, Meng Q. Aortic smooth muscle TRPV4 channels regulate vasoconstriction in high salt-induced hypertension. Hypertens Res 2023; 46:2356-2367. [PMID: 37532951 DOI: 10.1038/s41440-023-01363-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 08/04/2023]
Abstract
Recent studies have focused on the contribution of vascular endothelial transient receptor potential vanilloid 4 (TRPV4) channels to hypertension. However, in hypertension, TRPV4 channels in vascular smooth muscle remain unexplored. In the present study, we performed wire myograph experiments in isolated aortas from endothelial cell specific TRPV4 channel knockout (TRPV4EC-/-) mice to demonstrate that GSK1016790A (a specific TRPV4 channel agonist) triggered aortic smooth muscle-dependent contractions from mice on a normal-salt diet, and the contractions were enhanced in high-salt diet (HSD) mice. Intracellular Ca2+ concentration ([Ca2+]i) and Ca2+ imaging assays showed that TRPV4-induced [Ca2+]i was significantly higher in aortic smooth muscle cells (ASMCs) from HSD-induced hypertensive mice, and application of an inositol trisphosphate receptor (IP3R) inhibitor markedly attenuated TRPV4-induced [Ca2+]i. IP3R2 expression was enhanced in ASMCs from HSD-induced hypertensive mice and the contractile response induced by TRPV4 was inhibited by the IP3R inhibitor. Whole-transcriptome analysis by RNA-seq and western blot assays revealed the involvement of interferon regulatory factor 7 (IRF7) in TRPV4-IRF7-IP3R2 signaling in HSD-induced hypertension. These results suggested that TRPV4 channels regulate smooth muscle-dependent contractions in high salt-induced hypertension, and this contraction involves increased [Ca2+]i, IP3R2, and IRF7 activity. Our study revealed a considerable effect of TRPV4 channels in smooth muscle-dependent contraction in mice during high-salt induced hypertension.
Collapse
Affiliation(s)
- Xin Wen
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yidi Peng
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214000, China
| | - Yuefeng Peng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yuzhong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Xianfeng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Qingyou Meng
- Department of Vascular Surgery, General Surgery Clinical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
43
|
Niu L, Lu YJ, Zu XW, Yang W, Shen FK, Xu YY, Jiang M, Xie Y, Li SY, Gao J, Bai G. Magnolol alleviates pulmonary fibrosis inchronic obstructive pulmonary disease by targeting transient receptor potential vanilloid 4-ankyrin repeat domain. Phytother Res 2023; 37:4282-4297. [PMID: 37282760 DOI: 10.1002/ptr.7907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/08/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) plays a role in regulating pulmonary fibrosis (PF). While several TRPV4 antagonists including magnolol (MAG), have been discovered, the mechanism of action is not fully understood. This study aimed to investigate the effect of MAG on alleviating fibrosis in chronic obstructive pulmonary disease (COPD) based on TRPV4, and to further analyze its mechanism of action on TRPV4. COPD was induced using cigarette smoke and LPS. The therapeutic effect of MAG on COPD-induced fibrosis was evaluated. TRPV4 was identified as the main target protein of MAG using target protein capture with MAG probe and drug affinity response target stability assay. The binding sites of MAG at TRPV4 were analyzed using molecular docking and small molecule interaction with TRPV4-ankyrin repeat domain (ARD). The effects of MAG on TRPV4 membrane distribution and channel activity were analyzed by co-immunoprecipitation, fluorescence co-localization, and living cell assay of calcium levels. By targeting TRPV4-ARD, MAG disrupted the binding between phosphatidylinositol 3 kinase γ and TRPV4, leading to hampered membrane distribution on fibroblasts. Additionally, MAG competitively impaired ATP binding to TRPV4-ARD, inhibiting TRPV4 channel opening activity. MAG effectively blocked the fibrotic process caused by mechanical or inflammatory signals, thus alleviating PF in COPD. Targeting TRPV4-ARD presents a novel treatment strategy for PF in COPD.
Collapse
Affiliation(s)
- Lin Niu
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu-Jie Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Xing-Wang Zu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Fu-Kui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yan-Yan Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yang Xie
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Su-Yun Li
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
44
|
Maccarrone M, Di Marzo V, Gertsch J, Grether U, Howlett AC, Hua T, Makriyannis A, Piomelli D, Ueda N, van der Stelt M. Goods and Bads of the Endocannabinoid System as a Therapeutic Target: Lessons Learned after 30 Years. Pharmacol Rev 2023; 75:885-958. [PMID: 37164640 PMCID: PMC10441647 DOI: 10.1124/pharmrev.122.000600] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/12/2023] Open
Abstract
The cannabis derivative marijuana is the most widely used recreational drug in the Western world and is consumed by an estimated 83 million individuals (∼3% of the world population). In recent years, there has been a marked transformation in society regarding the risk perception of cannabis, driven by its legalization and medical use in many states in the United States and worldwide. Compelling research evidence and the Food and Drug Administration cannabis-derived cannabidiol approval for severe childhood epilepsy have confirmed the large therapeutic potential of cannabidiol itself, Δ9-tetrahydrocannabinol and other plant-derived cannabinoids (phytocannabinoids). Of note, our body has a complex endocannabinoid system (ECS)-made of receptors, metabolic enzymes, and transporters-that is also regulated by phytocannabinoids. The first endocannabinoid to be discovered 30 years ago was anandamide (N-arachidonoyl-ethanolamine); since then, distinct elements of the ECS have been the target of drug design programs aimed at curing (or at least slowing down) a number of human diseases, both in the central nervous system and at the periphery. Here a critical review of our knowledge of the goods and bads of the ECS as a therapeutic target is presented to define the benefits of ECS-active phytocannabinoids and ECS-oriented synthetic drugs for human health. SIGNIFICANCE STATEMENT: The endocannabinoid system plays important roles virtually everywhere in our body and is either involved in mediating key processes of central and peripheral diseases or represents a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of the components of this complex system, and in particular of key receptors (like cannabinoid receptors 1 and 2) and metabolic enzymes (like fatty acid amide hydrolase and monoacylglycerol lipase), will advance our understanding of endocannabinoid signaling and activity at molecular, cellular, and system levels, providing new opportunities to treat patients.
Collapse
Affiliation(s)
- Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Vincenzo Di Marzo
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Jürg Gertsch
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Uwe Grether
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Allyn C Howlett
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Tian Hua
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Alexandros Makriyannis
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Daniele Piomelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Natsuo Ueda
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| | - Mario van der Stelt
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy (M.M.); European Center for Brain Research, Santa Lucia Foundation, Rome, Italy (M.M.); Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, University of Laval, Quebec, Canada (V.D.); Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, Bern, Switzerland (J.G.); Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland (U.G.); Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.C.H.); iHuman Institute, ShanghaiTech University, Shanghai, China (T.H.); Center for Drug Discovery and Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (A.M.); Departments of Pharmaceutical Sciences and Biological Chemistry, University of California, Irvine, California (D.P.); Department of Biochemistry, Kagawa University School of Medicine, Miki, Kagawa, Japan (N.U.); Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands (M.S.)
| |
Collapse
|
45
|
Zhang Y, Tawiah GK, Wu X, Zhang Y, Wang X, Wei X, Qiao X, Zhang Q. Primary cilium-mediated mechanotransduction in cartilage chondrocytes. Exp Biol Med (Maywood) 2023; 248:1279-1287. [PMID: 37897221 PMCID: PMC10625344 DOI: 10.1177/15353702231199079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent joint disorders associated with the degradation of articular cartilage and an abnormal mechanical microenvironment. Mechanical stimuli, including compression, shear stress, stretching strain, osmotic challenge, and the physical properties of the matrix microenvironment, play pivotal roles in the tissue homeostasis of articular cartilage. The primary cilium, as a mechanosensory and chemosensory organelle, is important for detecting and transmitting both mechanical and biochemical signals in chondrocytes within the matrix microenvironment. Growing evidence indicates that primary cilia are critical for chondrocytes signaling transduction and the matrix homeostasis of articular cartilage. Furthermore, the ability of primary cilium to regulate cellular signaling is dynamic and dependent on the cellular matrix microenvironment. In the current review, we aim to elucidate the key mechanisms by which primary cilia mediate chondrocytes sensing and responding to the matrix mechanical microenvironment. This might have potential therapeutic applications in injuries and OA-associated degeneration of articular cartilage.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Godfred K Tawiah
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Yanjun Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaohong Qiao
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- Department of Orthopaedics, Lvliang Hospital Affiliated to Shanxi Medical University, Lvliang 033099, Shanxi, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
46
|
Nadezhdin KD, Talyzina IA, Parthasarathy A, Neuberger A, Zhang DX, Sobolevsky AI. Structure of human TRPV4 in complex with GTPase RhoA. Nat Commun 2023; 14:3733. [PMID: 37353478 PMCID: PMC10290124 DOI: 10.1038/s41467-023-39346-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023] Open
Abstract
Transient receptor potential (TRP) channel TRPV4 is a polymodal cellular sensor that responds to moderate heat, cell swelling, shear stress, and small-molecule ligands. It is involved in thermogenesis, regulation of vascular tone, bone homeostasis, renal and pulmonary functions. TRPV4 is implicated in neuromuscular and skeletal disorders, pulmonary edema, and cancers, and represents an important drug target. The cytoskeletal remodeling GTPase RhoA has been shown to suppress TRPV4 activity. Here, we present a structure of the human TRPV4-RhoA complex that shows RhoA interaction with the membrane-facing surface of the TRPV4 ankyrin repeat domains. The contact interface reveals residues that are mutated in neuropathies, providing an insight into the disease pathogenesis. We also identify the binding sites of the TRPV4 agonist 4α-PDD and the inhibitor HC-067047 at the base of the S1-S4 bundle, and show that agonist binding leads to pore opening, while channel inhibition involves a π-to-α transition in the pore-forming helix S6. Our structures elucidate the interaction interface between hTRPV4 and RhoA, as well as residues at this interface that are involved in TRPV4 disease-causing mutations. They shed light on TRPV4 activation and inhibition and provide a template for the design of future therapeutics for treatment of TRPV4-related diseases.
Collapse
Affiliation(s)
- Kirill D Nadezhdin
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - Irina A Talyzina
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, 10032, USA
| | - Aravind Parthasarathy
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Arthur Neuberger
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
| | - David X Zhang
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
47
|
Kwon DH, Zhang F, McCray BA, Feng S, Kumar M, Sullivan JM, Im W, Sumner CJ, Lee SY. TRPV4-Rho GTPase complex structures reveal mechanisms of gating and disease. Nat Commun 2023; 14:3732. [PMID: 37353484 PMCID: PMC10290081 DOI: 10.1038/s41467-023-39345-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/08/2023] [Indexed: 06/25/2023] Open
Abstract
Crosstalk between ion channels and small GTPases is critical during homeostasis and disease, but little is known about the structural underpinnings of these interactions. TRPV4 is a polymodal, calcium-permeable cation channel that has emerged as a potential therapeutic target in multiple conditions. Gain-of-function mutations also cause hereditary neuromuscular disease. Here, we present cryo-EM structures of human TRPV4 in complex with RhoA in the ligand-free, antagonist-bound closed, and agonist-bound open states. These structures reveal the mechanism of ligand-dependent TRPV4 gating. Channel activation is associated with rigid-body rotation of the intracellular ankyrin repeat domain, but state-dependent interaction with membrane-anchored RhoA constrains this movement. Notably, many residues at the TRPV4-RhoA interface are mutated in disease and perturbing this interface by introducing mutations into either TRPV4 or RhoA increases TRPV4 channel activity. Together, these results suggest that RhoA serves as an auxiliary subunit for TRPV4, regulating TRPV4-mediated calcium homeostasis and disruption of TRPV4-RhoA interactions can lead to TRPV4-related neuromuscular disease. These insights will help facilitate TRPV4 therapeutics development.
Collapse
Affiliation(s)
- Do Hoon Kwon
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Feng Zhang
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Brett A McCray
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Shasha Feng
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Meha Kumar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jeremy M Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Wonpil Im
- Departments of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA, 18015, USA
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
48
|
Chaigne S, Barbeau S, Ducret T, Guinamard R, Benoist D. Pathophysiological Roles of the TRPV4 Channel in the Heart. Cells 2023; 12:1654. [PMID: 37371124 DOI: 10.3390/cells12121654] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) channel is a non-selective cation channel that is mostly permeable to calcium (Ca2+), which participates in intracellular Ca2+ handling in cardiac cells. It is widely expressed through the body and is activated by a large spectrum of physicochemical stimuli, conferring it a role in a variety of sensorial and physiological functions. Within the cardiovascular system, TRPV4 expression is reported in cardiomyocytes, endothelial cells (ECs) and smooth muscle cells (SMCs), where it modulates mitochondrial activity, Ca2+ homeostasis, cardiomyocytes electrical activity and contractility, cardiac embryonic development and fibroblast proliferation, as well as vascular permeability, dilatation and constriction. On the other hand, TRPV4 channels participate in several cardiac pathological processes such as the development of cardiac fibrosis, hypertrophy, ischemia-reperfusion injuries, heart failure, myocardial infarction and arrhythmia. In this manuscript, we provide an overview of TRPV4 channel implications in cardiac physiology and discuss the potential of the TRPV4 channel as a therapeutic target against cardiovascular diseases.
Collapse
Affiliation(s)
- Sébastien Chaigne
- IHU LIRYC Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
- Electrophysiology and Ablation Unit, Bordeaux University Hospital, 33604 Pessac, France
| | - Solène Barbeau
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| | - Romain Guinamard
- UR4650, Physiopathologie et Stratégies d'Imagerie du Remodelage Cardiovasculaire, GIP Cyceron, Université de Caen Normandie, 14032 Caen, France
| | - David Benoist
- IHU LIRYC Electrophysiology and Heart Modeling Institute, Fondation Bordeaux Université, 33600 Bordeaux, France
- Centre de Recherche Cardio-Thoracique de Bordeaux, INSERM U1045, University of Bordeaux, 33600 Pessac, France
| |
Collapse
|
49
|
Wang X, Wang Y, Huo H, Zhou G, Li Y, Liang F, Xue J, Shi X, Yin A, Xiao Q, Yuan R, Pan C, Shen L, He B. Transient Receptor Vanilloid Subtype 4-Mediated Ca 2+ Influx Promotes Glomerular Endothelial Inflammation in Sepsis-Associated Acute Kidney Injury. J Transl Med 2023; 103:100126. [PMID: 36889540 DOI: 10.1016/j.labinv.2023.100126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a frequent complication in patients who are critically ill, which is often initiated by glomerular endothelial cell dysfunction. Although transient receptor vanilloid subtype 4 (TRPV4) ion channels are known to be permeable to Ca2+ and are widely expressed in the kidneys, the role of TRPV4 on glomerular endothelial inflammation in sepsis remains elusive. In the present study, we found that TRPV4 expression in mouse glomerular endothelial cells (MGECs) increased after lipopolysaccharide (LPS) stimulation or cecal ligation and puncture challenge, which increased intracellular Ca2+ in MGECs. Furthermore, the inhibition or knockdown of TRPV4 suppressed LPS-induced phosphorylation and translocation of inflammatory transcription factors NF-κB and IRF-3 in MGECs. Clamping intracellular Ca2+ mimicked LPS-induced responses observed in the absence of TRPV4. In vivo experiments showed that the pharmacologic blockade or knockdown of TRPV4 reduced glomerular endothelial inflammatory responses, increased survival rate, and improved renal function in cecal ligation and puncture-induced sepsis without altering renal cortical blood perfusion. Taken together, our results suggest that TRPV4 promotes glomerular endothelial inflammation in S-AKI and that its inhibition or knockdown alleviates glomerular endothelial inflammation by reducing Ca2+ overload and NF-κB/IRF-3 activation. These findings provide insights that may aid in the development of novel pharmacologic strategies for the treatment of S-AKI.
Collapse
Affiliation(s)
- Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yinhua Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Guo Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jieyuan Xue
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Anwen Yin
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Qingqing Xiao
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ruosen Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Changqing Pan
- Department of General Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
50
|
Santana Nunez D, Malik AB, Lee Q, Ahn SJ, Coctecon-Murillo A, Lazarko D, Levitan I, Mehta D, Komarova YA. Piezo1 induces endothelial responses to shear stress via soluble adenylyl Cyclase-IP 3R2 circuit. iScience 2023; 26:106661. [PMID: 37168565 PMCID: PMC10164902 DOI: 10.1016/j.isci.2023.106661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/30/2023] [Accepted: 04/07/2023] [Indexed: 05/13/2023] Open
Abstract
Endothelial cells (ECs) continuously sense and adapt to changes in shear stress generated by blood flow. Here, we show that the activation of the mechanosensitive channel Piezo1 by defined shear forces induces Ca2+ entry into the endoplasmic reticulum (ER) via the ER Ca2+ ATPase pump. This entry is followed by inositol trisphosphate receptor 2 (IP3R2)-elicited ER Ca2+ release into the cytosol. The mechanism of ER Ca2+ release involves the generation of cAMP by soluble adenylyl cyclase (sAC), leading to IP3R2-evoked Ca2+ gating. Depleting sAC or IP3R2 prevents ER Ca2+ release and blocks EC alignment in the direction of flow. Overexpression of constitutively active Akt1 restores the shear-induced alignment of ECs lacking Piezo1 or IP3R2, as well as the flow-induced vasodilation in endothelial restricted Piezo1 knockout mice. These studies describe an unknown Piezo1-cAMP-IP3R2 circuit as an essential mechanism activating Akt signaling and inducing adaptive changes in ECs to laminar flow.
Collapse
Affiliation(s)
- Dianicha Santana Nunez
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Asrar B. Malik
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Quinn Lee
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Sang Joon Ahn
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Arnold Coctecon-Murillo
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Dana Lazarko
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Irena Levitan
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Dolly Mehta
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yulia A. Komarova
- Department of Pharmacology and Regenerative Medicine, the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL, USA
| |
Collapse
|