1
|
Liu G, Dong BB, Devanarayana S, Chen RC, Liu Q. Emerging roles of mechanosensitive ion channels in ventilator induced lung injury: a systematic review. Front Immunol 2024; 15:1479230. [PMID: 39664395 PMCID: PMC11631737 DOI: 10.3389/fimmu.2024.1479230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
Background The pathogenetic mechanisms of ventilator-induced lung injury (VILI) still need to be elucidated. The mechanical forces during mechanical ventilation are continually sensed and transmitted by mechanosensitive ion channels (MSICs) in pulmonary endothelial, epithelial, and immune cells. In recent years, MSICs have been shown to be involved in VILI. Methods A systematic search across PubMed, the Cochrane Library, Web of Science, and ScienceDirect was performed from inception to March 2024, and the review was conducted in accordance with PRISMA guidelines. The potential eligible studies were evaluated by two authors independently. Study characteristics, quality assessment, and potential mechanisms were analyzed. Results We included 23 eligible studies, most of which were performed with murine animals in vivo. At the in vitro level, 52% and 48% of the experiments were conducted with human or animal cells, respectively. No clinical studies were found. The most reported MSICs include Piezo channels, transient receptor potential channels, potassium channels, and stretch-activated sodium channels. Piezo1 has been the most concerned channel in the recent five years. This study found that signal pathways, such as RhoA/ROCK1, could be enhanced by cyclic stretch-activated MSICs, which contribute to VILI through dysregulated inflammation and immune responses mediated by ion transport. The review indicates the emerging role of MSICs in the pathogenesis of VILI, especially as a signal-transmitting link between mechanical stretch and pathogenesis such as inflammation, disruption of cell junctions, and edema formation. Conclusions Mechanical stretch stimulates MSICs to increase transcellular ion exchange and subsequently generates VILI through inflammation and other pathogeneses mediated by MSICs signal-transmitting pathways. These findings make it possible to identify potential therapeutic targets for the prevention of lung injury through further exploration and more studies. Systematic review registration https://inplasy.com/inplasy-2024-10-0115/, identifier INPLASY2024100115.
Collapse
Affiliation(s)
- Gang Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin-bin Dong
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shalika Devanarayana
- School of International Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Rong-Chang Chen
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Shenzhen Institute of Respiratory Diseases, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Qi Liu
- Department of Emergency Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Xia T, Pan Z, Wan H, Li Y, Mao G, Zhao J, Zhang F, Pan S. Mechanisms of mechanical stimulation in the development of respiratory system diseases. Am J Physiol Lung Cell Mol Physiol 2024; 327:L724-L739. [PMID: 39316681 DOI: 10.1152/ajplung.00122.2024] [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: 04/09/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
During respiration, mechanical stress can initiate biological responses that impact the respiratory system. Mechanical stress plays a crucial role in the development of the respiratory system. However, pathological mechanical stress can impact the onset and progression of respiratory diseases by influencing the extracellular matrix and cell transduction processes. In this article, we explore the mechanisms by which mechanical forces communicate with and influence cells. We outline the basic knowledge of respiratory mechanics, elucidating the important role of mechanical stimulation in influencing respiratory system development and differentiation from a microscopic perspective. We also explore the potential mechanisms of mechanical transduction in the pathogenesis and development of respiratory diseases such as asthma, lung injury, pulmonary fibrosis, and lung cancer. Finally, we look forward to new research directions in cellular mechanotransduction, aiming to provide fresh insights for future therapeutic research on respiratory diseases.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoxin Wan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongsen Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Guocai Mao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Fangbiao Zhang
- Department of Cardiothoracic Surgery, Lishui Municipal Central Hospital, Lishui, People's Republic of China
| | - Shu Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
3
|
Strobel RJ, Ta HQ, Young AM, Wisniewski AM, Norman AV, Rotar EP, Stoler MH, Kron IL, Sonkusare SK, Roeser ME, Laubach VE. Transient receptor potential vanilloid 4 channel inhibition attenuates lung ischemia-reperfusion injury in a porcine lung transplant model. J Thorac Cardiovasc Surg 2024; 168:e121-e132. [PMID: 38678474 PMCID: PMC11416340 DOI: 10.1016/j.jtcvs.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/19/2024] [Accepted: 03/03/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel important in many physiological and pathophysiological processes, including pulmonary disease. Using a murine model, we previously demonstrated that TRPV4 mediates lung ischemia-reperfusion injury, the major cause of primary graft dysfunction after transplant. The current study tests the hypothesis that treatment with a TRPV4 inhibitor will attenuate lung ischemia-reperfusion injury in a clinically relevant porcine lung transplant model. METHODS A porcine left-lung transplant model was used. Animals were randomized to 2 treatment groups (n = 5/group): vehicle or GSK2193874 (selective TRPV4 inhibitor). Donor lungs underwent 30 minutes of warm ischemia and 24 hours of cold preservation before left lung allotransplantation and 4 hours of reperfusion. Vehicle or GSK2193874 (1 mg/kg) was administered to the recipient as a systemic infusion after recipient lung explant. Lung function, injury, and inflammatory biomarkers were compared. RESULTS After transplant, left lung oxygenation was significantly improved in the TRPV4 inhibitor group after 3 and 4 hours of reperfusion. Lung histology scores and edema were significantly improved, and neutrophil infiltration was significantly reduced in the TRPV4 inhibitor group. TRPV4 inhibitor-treated recipients had significantly reduced expression of interleukin-8, high mobility group box 1, P-selectin, and tight junction proteins (occludin, claudin-5, and zonula occludens-1) in bronchoalveolar lavage fluid as well as reduced angiopoietin-2 in plasma, all indicative of preservation of endothelial barrier function. CONCLUSIONS Treatment of lung transplant recipients with TRPV4 inhibitor significantly improves lung function and attenuates ischemia-reperfusion injury. Thus, selective TRPV4 inhibition may be a promising therapeutic strategy to prevent primary graft dysfunction after transplant.
Collapse
Affiliation(s)
- Raymond J Strobel
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Huy Q Ta
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Andrew M Young
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Alex M Wisniewski
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Anthony V Norman
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Evan P Rotar
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Mark H Stoler
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Va
| | - Irving L Kron
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center and the Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Va
| | - Mark E Roeser
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va.
| |
Collapse
|
4
|
Chen J, Gao Y, Liu N, Hai D, Wei W, Liu Y, Lan X, Jin X, Yu J, Ma L. Mechanism of NLRP3 Inflammasome in Epilepsy and Related Therapeutic Agents. Neuroscience 2024; 546:157-177. [PMID: 38574797 DOI: 10.1016/j.neuroscience.2024.03.029] [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: 12/31/2023] [Revised: 03/05/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
Epilepsy is one of the most widespread and complex diseases in the central nervous system (CNS), affecting approximately 65 million people globally, an important factor resulting in neurological disability-adjusted life year (DALY) and progressive cognitive dysfunction. Medication is the most essential treatment. The currently used drugs have shown drug resistance in some patients and only control symptoms; the development of novel and more efficacious pharmacotherapy is imminent. Increasing evidence suggests neuroinflammation is involved in the occurrence and development of epilepsy, and high expression of NLRP3 inflammasome has been observed in the temporal lobe epilepsy (TLE) brain tissue of patients and animal models. The inflammasome is a crucial cause of neuroinflammation by activating IL-1β and IL-18. Many preclinical studies have confirmed that regulating NLRP3 inflammasome pathway can prevent the development of epilepsy, reduce the severity of epilepsy, and play a neuroprotective role. Therefore, regulating NLRP3 inflammasome could be a potential target for epilepsy treatment. In summary, this review describes the priming and activation of inflammasome and its biological function in the progression of epilepsy. In addition, we reviewes the current pharmacological researches for epilepsy based on the regulation of NLRP3 inflammasome, aiming to provide a basis and reference for developing novel antiepileptic drugs.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Ning Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Dongmei Hai
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Wei Wei
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Yue Liu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xiaobing Lan
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China
| | - Xueqin Jin
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Jianqiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| | - Lin Ma
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
5
|
Vermillion MS, Saari N, Bray M, Nelson AM, Bullard RL, Rudolph K, Gigliotti AP, Brendler J, Jantzi J, Kuehl PJ, McDonald JD, Burgert ME, Weber W, Sucoloski S, Behm DJ. Effect of TRPV4 Antagonist GSK2798745 on Chlorine Gas-Induced Acute Lung Injury in a Swine Model. Int J Mol Sci 2024; 25:3949. [PMID: 38612759 PMCID: PMC11011849 DOI: 10.3390/ijms25073949] [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: 01/09/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
As a regulator of alveolo-capillary barrier integrity, Transient Receptor Potential Vanilloid 4 (TRPV4) antagonism represents a promising strategy for reducing pulmonary edema secondary to chemical inhalation. In an experimental model of acute lung injury induced by exposure of anesthetized swine to chlorine gas by mechanical ventilation, the dose-dependent effects of TRPV4 inhibitor GSK2798745 were evaluated. Pulmonary function and oxygenation were measured hourly; airway responsiveness, wet-to-dry lung weight ratios, airway inflammation, and histopathology were assessed 24 h post-exposure. Exposure to 240 parts per million (ppm) chlorine gas for ≥50 min resulted in acute lung injury characterized by sustained changes in the ratio of partial pressure of oxygen in arterial blood to the fraction of inspiratory oxygen concentration (PaO2/FiO2), oxygenation index, peak inspiratory pressure, dynamic lung compliance, and respiratory system resistance over 24 h. Chlorine exposure also heightened airway response to methacholine and increased wet-to-dry lung weight ratios at 24 h. Following 55-min chlorine gas exposure, GSK2798745 marginally improved PaO2/FiO2, but did not impact lung function, airway responsiveness, wet-to-dry lung weight ratios, airway inflammation, or histopathology. In summary, in this swine model of chlorine gas-induced acute lung injury, GSK2798745 did not demonstrate a clinically relevant improvement of key disease endpoints.
Collapse
Affiliation(s)
- Meghan S. Vermillion
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Nathan Saari
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Mathieu Bray
- GSK, Collegeville, PA 19426, USA; (M.B.); (S.S.); (D.J.B.)
| | - Andrew M. Nelson
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Robert L. Bullard
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Karin Rudolph
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Andrew P. Gigliotti
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Jeffrey Brendler
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Jacob Jantzi
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Philip J. Kuehl
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | - Jacob D. McDonald
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | | | - Waylon Weber
- Lovelace Biomedical Research Institute, Albuquerque, NM 87108, USA; (N.S.); (A.M.N.); (R.L.B.); (K.R.); (A.P.G.); (J.B.); (J.J.); (P.J.K.); (J.D.M.); (W.W.)
| | | | - David J. Behm
- GSK, Collegeville, PA 19426, USA; (M.B.); (S.S.); (D.J.B.)
| |
Collapse
|
6
|
Babaniamansour P, Jacho D, Niedzielski S, Rabino A, Garcia-Mata R, Yildirim-Ayan E. Modulating TRPV4 Channel Activity in Pro-Inflammatory Macrophages within the 3D Tissue Analog. Biomedicines 2024; 12:230. [PMID: 38275401 PMCID: PMC10813551 DOI: 10.3390/biomedicines12010230] [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: 12/21/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Investigating macrophage plasticity emerges as a promising strategy for promoting tissue regeneration and can be exploited by regulating the transient receptor potential vanilloid 4 (TRPV4) channel. The TRPV4 channel responds to various stimuli including mechanical, chemical, and selective pharmacological compounds. It is well documented that treating cells such as epithelial cells and fibroblasts with a TRPV4 agonist enhances the Ca2+ influx to the cells, which leads to secretion of pro-inflammatory cytokines, while a TRPV4 antagonist reduces both Ca2+ influx and pro-inflammatory cytokine secretion. In this work, we investigated the effect of selective TRPV4 modulator compounds on U937-differentiated macrophages encapsulated within three-dimensional (3D) matrices. Despite offering a more physiologically relevant model than 2D cultures, pharmacological treatment of macrophages within 3D collagen matrices is largely overlooked in the literature. In this study, pro-inflammatory macrophages were treated with an agonist, 500 nM of GSK1016790A (TRPV4(+)), and an antagonist, 10 mM of RN-1734 (TRPV4(-)), to elucidate the modulation of the TRPV4 channel at both cellular and extracellular levels. To evaluate macrophage phenotypic alterations within 3D collagen matrices following TRPV4 modulator treatment, we employed structural techniques (SEM, Masson's trichrome, and collagen hybridizing peptide (CHP) staining), quantitative morphological measures for phenotypic assessment, and genotypic methods such as quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC). Our data reveal that pharmacological modulation of the macrophage TRPV4 channel alters the cytoskeletal structure of macrophages and influences the 3D structure encapsulating them. Moreover, we proved that treating macrophages with a TRPV4 agonist and antagonist enhances the expression of pro- and anti-inflammatory genes, respectively, leading to the upregulation of surface markers CD80 and CD206. In the TRPV4(-) group, the CD206 gene and CD206 surface marker were significantly upregulated by 9- and 2.5-fold, respectively, compared to the control group. These findings demonstrate that TRPV4 modulation can be utilized to shift macrophage phenotype within the 3D matrix toward a desired state. This is an innovative approach to addressing inflammation in musculoskeletal tissues.
Collapse
Affiliation(s)
- Parto Babaniamansour
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Diego Jacho
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Skyler Niedzielski
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| | - Agustin Rabino
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Rafael Garcia-Mata
- Department of Biological Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, College of Engineering, University of Toledo, Toledo, OH 43606, USA; (P.B.); (S.N.)
| |
Collapse
|
7
|
Luo M, Wang C, Guo J, Wen K, Yang C, Ni K, Liu L, Pan Y, Li J, Deng L. High Stretch Modulates cAMP/ATP Level in Association with Purine Metabolism via miRNA-mRNA Interactions in Cultured Human Airway Smooth Muscle Cells. Cells 2024; 13:110. [PMID: 38247802 PMCID: PMC10813996 DOI: 10.3390/cells13020110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/06/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
High stretch (>10% strain) of airway smooth muscle cells (ASMCs) due to mechanical ventilation (MV) is postulated to contribute to ventilator-induced lung injury (VILI), but the underlying mechanisms remain largely unknown. We hypothesized that ASMCs may respond to high stretch via regulatory miRNA-mRNA interactions, and thus we aimed to identify high stretch-responsive cellular events and related regulating miRNA-mRNA interactions in cultured human ASMCs with/without high stretch. RNA-Seq analysis of whole genome-wide miRNAs revealed 12 miRNAs differentially expressed (DE) in response to high stretch (7 up and 5 down, fold change >2), which target 283 DE-mRNAs as identified by a parallel mRNA sequencing and bioinformatics analysis. The KEGG and GO analysis further indicated that purine metabolism was the first enriched event in the cells during high stretch, which was linked to miR-370-5p-PDE4D/AK7. Since PDE4D/AK7 have been previously linked to cAMP/ATP metabolism in lung diseases and now to miR-370-5p in ASMCs, we thus evaluated the effect of high stretch on the cAMP/ATP level inside ASMCs. The results demonstrated that high stretch modulated the cAMP/ATP levels inside ASMCs, which could be largely abolished by miR-370-5p mimics. Together, these findings indicate that miR-370-5p-PDE4D/AK7 mediated high stretch-induced modulation of cAMP and ATP synthesis inside ASMCs. Furthermore, such interactive miRNA-mRNA pairs may provide new insights for the discovery of effective biomarkers/therapeutic targets for the diagnosis and treatment of VILI and other MV-associated respiratory diseases.
Collapse
Affiliation(s)
- Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Chunhong Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Jia Guo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Kang Wen
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Chongxin Yang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Kai Ni
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Lei Liu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Yan Pan
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Jingjing Li
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| |
Collapse
|
8
|
王 文, 杨 泳, 王 莉, 郭 欣, 田 玲, 王 鹤, 胡 玉, 刘 睿. [Sevoflurane alleviates ventilator-induced lung injury in rats by down-regulating the TRPV4/C-PLA2 signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1886-1891. [PMID: 38081606 PMCID: PMC10713460 DOI: 10.12122/j.issn.1673-4254.2023.11.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Indexed: 12/18/2023]
Abstract
OBJECTIVE To explore the mechanism underlying the protective effect of sevoflurane against ventilator-induced lung injury (VILI). METHODS Thirty-two SD rats were randomized into mechanical ventilation (MV) group, MV+sevoflurane group (MS group), MV+sevoflurane+transient receptor potential vanillate subtype 4 (TRPV4) agonist group (MST group) and MV+ sevoflurane + vehicle group (MSV group). Arachidonic acid (AA) in the lung tissues was quantified with ELISA. TRPV4, cytoplasmic phospholipase A2 (C-PLA2) and myosin light chain kinase (MLCK) protein expressions were detected by Western blotting. Lung injury in the rats was evaluated by assessing MLCK protein expression level, pulmonary permeability index, lung wet/dry ratio, leukocyte count in the bronchoalveolar lavage fluid (BALF), myeloperoxidase content in lung tissue, and histological score of the lungs. RESULTS The rats in MV group showed significantly increased TRPV4 and C-PLA2 expression levels in the lung tissues with increased lung permeability and obvious lung inflammation compared with those in the other 3 groups (P < 0.05). No significant differences were found in the parameters associated with lung injuries between MS group and MSV group. Compared with those in MST group, the rats in MS group and MSV group showed significantly reduced AA production and TRPV4 and C-PLA2 expressions in the lungs (P < 0.05) with alleviated lung hyper-permeability and inflammation (P < 0.05). CONCLUSION Sevoflurane protects against VILI in rats by down-regulating the TRPV4/C-PLA2 signaling pathway.
Collapse
Affiliation(s)
- 文法 王
- 楚雄彝族自治州人民医院麻醉科,云南 楚雄 675000Department of Anesthesiology, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong 675000, China
| | - 泳 杨
- 昆明医科大学医学机能实验中心,云南 昆明 650500Experimental Center of Medical Function, Kunming Medical University, Kunming 650500, China
| | - 莉 王
- 云南省第一人民医院//昆明理工大学附属医院麻醉科,云南 昆明 650032Department of Anesthesiology, First People's Hospital of Yunnan Province/Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - 欣 郭
- 云南省第一人民医院//昆明理工大学附属医院麻醉科,云南 昆明 650032Department of Anesthesiology, First People's Hospital of Yunnan Province/Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - 玲芳 田
- 楚雄彝族自治州人民医院麻醉科,云南 楚雄 675000Department of Anesthesiology, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong 675000, China
| | - 鹤 王
- 楚雄彝族自治州人民医院麻醉科,云南 楚雄 675000Department of Anesthesiology, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong 675000, China
| | - 玉珍 胡
- 楚雄彝族自治州人民医院麻醉科,云南 楚雄 675000Department of Anesthesiology, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong 675000, China
| | - 睿 刘
- 云南省第一人民医院//昆明理工大学附属医院麻醉科,云南 昆明 650032Department of Anesthesiology, First People's Hospital of Yunnan Province/Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| |
Collapse
|
9
|
Liu H, Fan P, Jin F, Ren H, Xu F, Li J. Targeting biophysical microenvironment for improved treatment of chronic obstructive pulmonary disease. Trends Mol Med 2023; 29:926-938. [PMID: 37704492 DOI: 10.1016/j.molmed.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is responsible for high disability rates, high death rates, and significant cost to health systems. Growing evidence in recent decades shows significant biophysical microenvironment changes in COPD, impacting lung tissues, cells, and treatment response. Furthermore, such biophysical changes have shown great potential as novel targets for improved therapeutic strategy of COPD, where both pharmacological and non-pharmacological therapies focusing on repairing the biophysical microenvironment of the lung have emerged. We present the first comprehensive review of four distinct biophysical hallmarks [i.e., extracellular matrix (ECM) microarchitecture, stiffness, fluid shear stress, and mechanical stretch] in COPD, the possible involvement of pathological changes, possible effects, and correlated in vitro models and sum up the emerging COPD treatments targeting these biophysical hallmarks.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Hui Ren
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China.
| |
Collapse
|
10
|
To KKW, Cho WC. Drug Repurposing to Circumvent Immune Checkpoint Inhibitor Resistance in Cancer Immunotherapy. Pharmaceutics 2023; 15:2166. [PMID: 37631380 PMCID: PMC10459070 DOI: 10.3390/pharmaceutics15082166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have achieved unprecedented clinical success in cancer treatment. However, drug resistance to ICI therapy is a major hurdle that prevents cancer patients from responding to the treatment or having durable disease control. Drug repurposing refers to the application of clinically approved drugs, with characterized pharmacological properties and known adverse effect profiles, to new indications. It has also emerged as a promising strategy to overcome drug resistance. In this review, we summarized the latest research about drug repurposing to overcome ICI resistance. Repurposed drugs work by either exerting immunostimulatory activities or abolishing the immunosuppressive tumor microenvironment (TME). Compared to the de novo drug design strategy, they provide novel and affordable treatment options to enhance cancer immunotherapy that can be readily evaluated in the clinic. Biomarkers are exploited to identify the right patient population to benefit from the repurposed drugs and drug combinations. Phenotypic screening of chemical libraries has been conducted to search for T-cell-modifying drugs. Genomics and integrated bioinformatics analysis, artificial intelligence, machine and deep learning approaches are employed to identify novel modulators of the immunosuppressive TME.
Collapse
Affiliation(s)
- Kenneth K. W. To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| |
Collapse
|
11
|
Xu P, Lin H, Jiao H, Zhao J, Wang X. Chicken embryo thermal manipulation alleviates postnatal heat stress-induced jejunal inflammation by inhibiting Transient Receptor Potential V4. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 256:114851. [PMID: 37004430 DOI: 10.1016/j.ecoenv.2023.114851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/16/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Intestinal inflammation induced by heat stress is an important factor restricting the healthy growth of broilers. The aim of this study was to evaluate the effect of chicken embryo thermal manipulation (39.5 ℃ and 65 % RH for 3 h daily during 16-18 th embryonic age) on intestinal inflammation in broilers under postnatal heat stress and to investigate whether transient receptor potential V4 (TRPV4) plays a role in this process. Our results suggest that broilers with embryo thermal manipulation experience could delay the rising of rectal temperature during postnatal heat stress (P < 0.05), and had better production performance (P < 0.05), intestinal morphological parameters (P < 0.05) and higher expression of tight junction related genes (P < 0.05). The increased serum lipopolysaccharide (LPS) content, activation of nuclear factor-kappa B (NF-κB) signaling pathway and the increased expression of pro-inflammatory cytokines interleukin (IL)-1β, IL-6 and tumor necrosis factor alpha (TNF-α) in jejunum during postnatal heat stress were alleviated by embryo thermal manipulation (P < 0.05). Postnatal heat stress induced an increase in mRNA and protein expression of TRPV4 in jejunum (P < 0.05), but had no effect on broilers which experienced embryo thermal manipulation (P > 0.05). Inhibition of TRPV4 reduced LPS-induced Ca2+ influx and restrained the activation of NF-κB signaling pathway and the expression of downstream pro-inflammatory cytokines (P < 0.05). The expression of DNA methyltransferase (DNMT) in the jejunum of broilers exposed to postnatal heat stress was increased by embryo thermal manipulation (P < 0.05). The DNA methylation level of TRPV4 promoter region was detected, and the results showed that embryo thermal manipulation increased the DNA methylation level of TRPV4 promoter region (P < 0.05). In conclusion, Chicken embryo thermal manipulation can alleviate jejunal inflammation in broilers under postnatal heat stress. This may be due to the decreased circulating LPS or the increased DNA methylation level in the promoter region of TRPV4, which inhibits TRPV4 expression, thereby reducing Ca2+ influx, and finally alleviating inflammation by affecting NF-κB signaling pathway. The work is an attempt to understand the mechanism involved in alleviation of adverse effects of heat stress during postnatal life through prenatal thermal manipulation and to reveal the important role of epigenetics.
Collapse
Affiliation(s)
- Peng Xu
- College of Animal Science & Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, Shandong, China
| | - Hai Lin
- College of Animal Science & Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, Shandong, China
| | - Hongchao Jiao
- College of Animal Science & Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, Shandong, China
| | - Jingpeng Zhao
- College of Animal Science & Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaojuan Wang
- College of Animal Science & Technology, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Agricultural University, Taian, Shandong, China.
| |
Collapse
|
12
|
Benítez-Angeles M, Juárez-González E, Vergara-Jaque A, Llorente I, Rangel-Yescas G, Thébault SC, Hiriart M, Islas LD, Rosenbaum T. Unconventional interactions of the TRPV4 ion channel with beta-adrenergic receptor ligands. Life Sci Alliance 2023; 6:6/3/e202201704. [PMID: 36549871 PMCID: PMC9780703 DOI: 10.26508/lsa.202201704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is present in different tissues including those of the airways. This channel is activated in response to stimuli such as changes in temperature, hypoosmotic conditions, mechanical stress, and chemicals from plants, lipids, and others. TRPV4's overactivity and/or dysfunction has been associated with several diseases, such as skeletal dysplasias, neuromuscular disorders, and lung pathologies such as asthma and cardiogenic lung edema and COVID-19-related respiratory malfunction. TRPV4 antagonists and blockers have been described; nonetheless, the mechanisms involved in achieving inhibition of the channel remain scarce, and the search for safe use of these molecules in humans continues. Here, we show that the widely used bronchodilator salbutamol and other ligands of β-adrenergic receptors inhibit TRPV4's activation. We also demonstrate that inhibition of TRPV4 by salbutamol is achieved through interaction with two residues located in the outer region of the pore and that salbutamol leads to channel closing, consistent with an allosteric mechanism. Our study provides molecular insights into the mechanisms that regulate the activity of this physiopathologically important ion channel.
Collapse
Affiliation(s)
- Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - Emmanuel Juárez-González
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - Ariela Vergara-Jaque
- Center for Bioinformatics, Simulation and Modeling, Faculty of Engineering, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | | | | | - Marcia Hiriart
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina, UNAM, México, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, México
| |
Collapse
|
13
|
Yang C, Guo J, Ni K, Wen K, Qin Y, Gu R, Wang C, Liu L, Pan Y, Li J, Luo M, Deng L. Mechanical Ventilation-Related High Stretch Mainly Induces Endoplasmic Reticulum Stress and Thus Mediates Inflammation Response in Cultured Human Primary Airway Smooth Muscle Cells. Int J Mol Sci 2023; 24:3811. [PMID: 36835223 PMCID: PMC9958795 DOI: 10.3390/ijms24043811] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Ventilator-induced lung injury (VILI) occurs in mechanically ventilated patients of respiratory disease and is typically characterized by airway inflammation. However, recent studies increasingly indicate that a major cause of VILI may be the excessive mechanical loading such as high stretch (>10% strain) on airway smooth muscle cells (ASMCs) due to mechanical ventilation (MV). Although ASMCs are the primary mechanosensitive cells in airways and contribute to various airway inflammation diseases, it is still unclear how they respond to high stretch and what mediates such a response. Therefore, we used whole genome-wide mRNA-sequencing (mRNA-Seq), bioinformatics, and functional identification to systematically analyze the mRNA expression profiles and signaling pathway enrichment of cultured human ASMCs exposed to high stretch (13% strain), aiming to screen the susceptible signaling pathway through which cells respond to high stretch. The data revealed that in response to high stretch, 111 mRNAs with count ≥100 in ASMCs were significantly differentially expressed (defined as DE-mRNAs). These DE-mRNAs are mainly enriched in endoplasmic reticulum (ER) stress-related signaling pathways. ER stress inhibitor (TUDCA) abolished high-stretch-enhanced mRNA expression of genes associated with ER stress, downstream inflammation signaling, and major inflammatory cytokines. These results demonstrate in a data-driven approach that in ASMCs, high stretch mainly induced ER stress and activated ER stress-related signaling and downstream inflammation response. Therefore, it suggests that ER stress and related signaling pathways in ASMCs may be potential targets for timely diagnosis and intervention of MV-related pulmonary airway diseases such as VILI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Mingzhi Luo
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| |
Collapse
|
14
|
Abstract
The ability to detect stimuli from the environment plays a pivotal role in our survival. The molecules that allow the detection of such signals include ion channels, which are proteins expressed in different cells and organs. Among these ion channels, the transient receptor potential (TRP) family responds to the presence of diverse chemicals, temperature, and osmotic changes, among others. This family of ion channels includes the TRPV or vanilloid subfamily whose members serve several physiological functions. Although these proteins have been studied intensively for the last two decades, owing to their structural and functional complexities, a number of controversies regarding their function still remain. Here, we discuss some salient features of their regulation in light of these controversies and outline some of the efforts pushing the field forward.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Department of Cognitive Neuroscience, Neuroscience Division, Institute for Cellular Physiology, National Autonomous University of Mexico, Coyoacán, México;
| | - León D Islas
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Coyoacán, México
| |
Collapse
|
15
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
16
|
Blocking TRPV4 Ameliorates Osteoarthritis by Inhibiting M1 Macrophage Polarization via the ROS/NLRP3 Signaling Pathway. Antioxidants (Basel) 2022; 11:antiox11122315. [PMID: 36552524 PMCID: PMC9774183 DOI: 10.3390/antiox11122315] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/12/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA) is a low-level inflammatory disease in which synovial macrophage M1 polarization exacerbates the progression of synovitis and OA. Notedly, the ROS (reactive oxygen species) level in macrophages is intimately implicated in macrophage M1 polarization. TRPV4 (transient receptor potential channel subfamily V member 4), as an ion channel, plays a pivotal role in oxidative stress and inflammation. In this study, we investigated the role of TRPV4 in OA progression and M1 macrophage polarization. Male adult Sprague-Dawley (SD) rats underwent a medial meniscus radial transection operation to create an OA model in vivo and RAW 264.7 cells were intervened with 100 ng/mL LPS (lipopolysaccharide) to induce M1-polarized macrophages in vitro. We demonstrated that the infiltration of M1 synovial macrophages and the expression of TRPV4 were increased significantly in OA synovium. In addition, intra-articular injection of HC067074 (a specific inhibitor of TRPV4) alleviated the progression of rat OA and significantly decreased synovial macrophage M1 polarization. Further mechanisms suggested that ROS production by M1 macrophages was decreased after TRPV4 inhibition. In addition, NLRP3 (pyrin domain containing protein 3) as a downstream effector of ROS in M1-polarized macrophage, was significantly suppressed following TRPV4 inhibition. In conclusion, this study discovered that inhibition of TRPV4 delays OA progression by inhibiting M1 synovial macrophage polarization through the ROS/NLRP3 pathway.
Collapse
|
17
|
Kuronuma K, Otsuka M, Wakabayashi M, Yoshioka T, Kobayashi T, Kameda M, Morioka Y, Chiba H, Takahashi H. Role of transient receptor potential vanilloid 4 in therapeutic anti-fibrotic effects of pirfenidone. Am J Physiol Lung Cell Mol Physiol 2022; 323:L193-L205. [PMID: 35787697 DOI: 10.1152/ajplung.00565.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fatal lung disorder characterized by aberrant extracellular matrix deposition in the interstitium. Pirfenidone is an anti-fibrotic agent used to treat patients with IPF. Pirfenidone shows a pleiotropic mode of action, but its underlying anti-fibrotic mechanism is unclear. Transient receptor potential vanilloid 4 (TRPV4), which is a mechanosensitive calcium channel, was recently shown to be related to pulmonary fibrosis. To clarify the anti-fibrotic mechanisms of pirfenidone, we investigated whether TRPV4 blockade has a pharmacological effect in a murine model of pulmonary fibrosis and whether pirfenidone contributes to suppression of TRPV4. Our synthetic TRPV4 antagonist and pirfenidone treatment attenuated lung injury in the bleomycin mouse model. TRPV4-mediated increases in intracellular calcium were inhibited by pirfenidone. Additionally, TRPV4-stimulated interleukin-8 release from cells was reduced and a delay in cell migration was abolished by pirfenidone. Furthermore, pirfenidone decreased TRPV4 endogenous ligands in bleomycin-administered mouse lungs and their production by microsomes of human lungs. We found TRPV4 expression in the bronchiolar and alveolar epithelium and activated fibroblasts of the lungs in patients with IPF. Finally, we showed that changes in forced vital capacity of patients with IPF treated with pirfenidone were significantly correlated with metabolite levels of TRPV4 endogenous ligands in bronchoalveolar lavage fluid. These results suggest that the anti-fibrotic action of pirfenidone is partly mediated by TRPV4 and that TRPV4 endogenous ligands in bronchoalveolar lavage fluid may be biomarkers for distinguishing responders to pirfenidone.
Collapse
Affiliation(s)
- Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine, Sapporo-Kosei General Hospital, Sapporo, Japan
| | - Masato Wakabayashi
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Takeshi Yoshioka
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Tomofumi Kobayashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masami Kameda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuhide Morioka
- Drug Discovery and Disease Research Laboratory, Shionogi Co., Ltd., Osaka, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
18
|
Jo AO, Lakk M, Rudzitis CN, Križaj D. TRPV4 and TRPC1 channels mediate the response to tensile strain in mouse Müller cells. Cell Calcium 2022; 104:102588. [PMID: 35398674 PMCID: PMC9119919 DOI: 10.1016/j.ceca.2022.102588] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/10/2022] [Accepted: 04/01/2022] [Indexed: 11/24/2022]
Abstract
Müller glia, a pillar of metabolic, volume regulatory and immune/inflammatory signaling in the mammalian retina, are among the earliest responders to mechanical stressors in the eye. Ocular trauma, edema, detachment and glaucoma evoke early inflammatory activation of Müller cells yet the identity of their mechanotransducers and signaling mechanisms downstream remains unknown. Here, we investigate expression of genes that encode putative stretch-activated calcium channels (SACs) in mouse Müller cells and study their responses to dynamical tensile loading in cells loaded with a calcium indicator dye. Transcript levels in purified glia were Trpc1>Piezo1>Trpv2>Trpv4>>Trpv1>Trpa1. Cyclic radial deformation of matrix-coated substrates produced dose-dependent increases in [Ca2+]i that were suppressed by the TRPV4 channel antagonist HC-067047 and by ablation of the Trpv4 gene. Stretch-evoked calcium responses were also reduced by knockdown and pharmacological inhibition of TRPC1 channels whereas the TRPV2 inhibitor tranilast had no effect. These data demonstrate that Müller cells are intrinsically mechanosensitive, with the response to tensile loading mediated through synergistic activation of TRPV4 and TRPC1 channels. Coupling between mechanical stress and Müller Ca2+ homeostasis has treatment implications, since many neuronal injury paradigms in the retina involve calcium dysregulation associated with inflammatory and immune signaling.
Collapse
Affiliation(s)
- Andrew O Jo
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Monika Lakk
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Christopher N Rudzitis
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132; Interdepartmental Program in Neuroscience
| | - David Križaj
- Department of Ophthalmology & Visual Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132; Interdepartmental Program in Neuroscience; Department of Neurobiology, University of Utah, Salt Lake City, UT 84112; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112.
| |
Collapse
|
19
|
Calcium–Permeable Channels and Endothelial Dysfunction in Acute Lung Injury. Curr Issues Mol Biol 2022; 44:2217-2229. [PMID: 35678679 PMCID: PMC9164020 DOI: 10.3390/cimb44050150] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
The increased permeability of the lung microvascular endothelium is one critical initiation of acute lung injury (ALI). The disruption of vascular-endothelium integrity results in leakiness of the endothelial barrier and accumulation of protein-rich fluid in the alveoli. During ALI, increased endothelial-cell (EC) permeability is always companied by high frequency and amplitude of cytosolic Ca2+ oscillations. Mechanistically, cytosolic calcium oscillations include calcium release from internal stores and calcium entry via channels located in the cell membrane. Recently, numerous publications have shown substantial evidence that calcium-permeable channels play an important role in maintaining the integrity of the endothelium barrier function of the vessel wall in ALI. These novel endothelial signaling pathways are future targets for the treatment of lung injury. This short review focuses on the up-to-date research and provide insight into the contribution of calcium influx via ion channels to the disruption of lung microvascular endothelial-barrier function during ALI.
Collapse
|
20
|
Nguyen TN, Siddiqui G, Veldhuis NA, Poole DP. Diverse Roles of TRPV4 in Macrophages: A Need for Unbiased Profiling. Front Immunol 2022; 12:828115. [PMID: 35126384 PMCID: PMC8811046 DOI: 10.3389/fimmu.2021.828115] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Nicholas A. Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| | - Daniel P. Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| |
Collapse
|
21
|
Zhou L, Xue C, Chen Z, Jiang W, He S, Zhang X. c-Fos is a mechanosensor that regulates inflammatory responses and lung barrier dysfunction during ventilator-induced acute lung injury. BMC Pulm Med 2022; 22:9. [PMID: 34986829 PMCID: PMC8734268 DOI: 10.1186/s12890-021-01801-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/13/2021] [Indexed: 12/05/2022] Open
Abstract
Background As one of the basic treatments performed in the intensive care unit, mechanical ventilation can cause ventilator-induced acute lung injury (VILI). The typical features of VILI are an uncontrolled inflammatory response and impaired lung barrier function; however, its pathogenesis is not fully understood, and c-Fos protein is activated under mechanical stress. c-Fos/activating protein-1 (AP-1) plays a role by binding to AP-1 within the promoter region, which promotes inflammation and apoptosis. T-5224 is a specific inhibitor of c-Fos/AP-1, that controls the gene expression of many proinflammatory cytokines. This study investigated whether T-5224 attenuates VILI in rats by inhibiting inflammation and apoptosis. Methods The SD rats were divided into six groups: a control group, low tidal volume group, high tidal volume group, DMSO group, T-5224 group (low concentration), and T-5224 group (high concentration). After 3 h, the pathological damage, c-Fos protein expression, inflammatory reaction and apoptosis degree of lung tissue in each group were detected. Results c-Fos protein expression was increased within the lung tissue of VILI rats, and the pathological damage degree, inflammatory reaction and apoptosis in the lung tissue of VILI rats were significantly increased; T-5224 inhibited c-Fos protein expression in lung tissues, and T-5224 inhibit the inflammatory reaction and apoptosis of lung tissue by regulating the Fas/Fasl pathway. Conclusions c-Fos is a regulatory factor during ventilator-induced acute lung injury, and the inhibition of its expression has a protective effect. Which is associated with the antiinflammatory and antiapoptotic effects of T-5224.
Collapse
Affiliation(s)
- Leilei Zhou
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China
| | - Chunju Xue
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China
| | - Zongyu Chen
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China
| | - Wenqing Jiang
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China
| | - Shuang He
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China
| | - Xianming Zhang
- School of Clinical Medicine, Guizhou Medical University, 550004, Guiyang, China. .,Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, 550004, Guiyang, China.
| |
Collapse
|
22
|
Jia Q, Yang Y, Chen X, Yao S, Hu Z. Emerging roles of mechanosensitive ion channels in acute lung injury/acute respiratory distress syndrome. Respir Res 2022; 23:366. [PMID: 36539808 PMCID: PMC9764320 DOI: 10.1186/s12931-022-02303-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a devastating respiratory disorder with high rates of mortality and morbidity, but the detailed underlying mechanisms of ALI/ARDS remain largely unknown. Mechanosensitive ion channels (MSCs), including epithelial sodium channel (ENaC), Piezo channels, transient receptor potential channels (TRPs), and two-pore domain potassium ion (K2P) channels, are highly expressed in lung tissues, and the activity of these MSCs can be modulated by mechanical forces (e.g., mechanical ventilation) and other stimuli (e.g., LPS, hyperoxia). Dysfunction of MSCs has been found in various types of ALI/ARDS, and MSCs play a key role in regulating alveolar fluid clearance, alveolar epithelial/endothelial barrier function, the inflammatory response and surfactant secretion in ALI/ARDS lungs. Targeting MSCs exerts therapeutic effects in the treatment of ALI/ARDS. In this review, we summarize the structure and functions of several well-recognized MSCs, the role of MSCs in the pathogenesis of ALI/ARDS and recent advances in the pharmacological and molecular modulation of MSCs in the treatment of ALI/ARDS. According to the current literature, targeting MSCs might be a very promising therapeutic approach against ALI/ARDS.
Collapse
Affiliation(s)
- Qi Jia
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyi Yang
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglong Yao
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Hu
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Abstract
The alveolo-capillary barrier is relatively impermeable, and facilitates gas exchange via the large alveolar surface in the lung. Disruption of alveolo-capillary barrier leads to accumulation of edema fluid in lung injury. Studies in animal models of various forms of lung injury provide evidence that TRPV4 channels play a critical role in disruption of the alveolo-capillary barrier and pathogenesis of lung injury. TRPV4 channels from capillary endothelial cells, alveolar epithelial cells, and immune cells have been implicated in the pathogenesis of lung injury. Recent studies in endothelium-specific TRPV4 knockout mice point to a central role for endothelial TRPV4 channels in lung injury. In this chapter, we review the findings on the pathological roles of endothelial TRPV4 channels in different forms of lung injury and future directions for further investigation.
Collapse
|
24
|
Kono M, Nanushaj D, Sakatani H, Murakami D, Hijiya M, Kinoshita T, Shiga T, Kaneko F, Enomoto K, Sugita G, Miyajima M, Okada Y, Saika S, Hotomi M. The Roles of Transient Receptor Potential Vanilloid 1 and 4 in Pneumococcal Nasal Colonization and Subsequent Development of Invasive Disease. Front Immunol 2021; 12:732029. [PMID: 34804016 PMCID: PMC8595402 DOI: 10.3389/fimmu.2021.732029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Transient receptor potential (TRP) channels, neuronal stimulations widely known to be associated with thermal responses, pain induction, and osmoregulation, have been shown in recent studies to have underlying mechanisms associated with inflammatory responses. The role of TRP channels on inflammatory milieu during bacterial infections has been widely demonstrated. It may vary among types of channels/pathogens, however, and it is not known how TRP channels function during pneumococcal infections. Streptococcus pneumoniae can cause severe infections such as pneumonia, bacteremia, and meningitis, with systemic inflammatory responses. This study examines the role of TRP channels (TRPV1 and TRPV4) for pneumococcal nasal colonization and subsequent development of invasive pneumococcal disease in a mouse model. Both TRPV1 and TRPV4 channels were shown to be related to regulation of the development of pneumococcal diseases. In particular, the influx of neutrophils (polymorphonuclear cells) in the nasal cavity and the bactericidal activity were significantly suppressed among TRPV4 knockout mice. This may lead to severe pneumococcal pneumonia, resulting in dissemination of the bacteria to various organs and causing high mortality during influenza virus coinfection. Regulating host immune responses by TRP channels could be a novel strategy against pathogenic microorganisms causing strong local/systemic inflammation.
Collapse
Affiliation(s)
- Masamitsu Kono
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Denisa Nanushaj
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hideki Sakatani
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Daichi Murakami
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Masayoshi Hijiya
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Tetsuya Kinoshita
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Tatsuya Shiga
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Fumie Kaneko
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Keisuke Enomoto
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Gen Sugita
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Yuka Okada
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Muneki Hotomi
- Department of Otorhinolaryngology-Head and Neck Surgery, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
25
|
Zhang Y, Jiang L, Huang T, Lu D, Song Y, Wang L, Gao J. Mechanosensitive cation channel Piezo1 contributes to ventilator-induced lung injury by activating RhoA/ROCK1 in rats. Respir Res 2021; 22:250. [PMID: 34548087 PMCID: PMC8456630 DOI: 10.1186/s12931-021-01844-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 09/13/2021] [Indexed: 01/13/2023] Open
Abstract
Background Mechanical ventilation can induce or aggravate lung injury, which is termed ventilator-induced lung injury (VILI). Piezo1 is a key element of the mechanotransduction process and can transduce mechanical signals into biological signals by mediating Ca2+ influx, which in turn regulates cytoskeletal remodeling and stress alterations. We hypothesized that it plays an important role in the occurrence of VILI, and investigated the underlying mechanisms. Methods High tidal volume mechanical ventilation and high magnitude cyclic stretch were performed on Sprague–Dawley rats, and A549 and human pulmonary microvascular endothelial cells, respectively, to establish VILI models. Immunohistochemical staining, flow cytometry, histological examination, enzyme-linked immunosorbent assay, western blotting, quantitative real-time polymerase chain reaction and survival curves were used to assess the effect of Piezo1 on induction of lung injury, as well as the signaling pathways involved. Results We observed that Piezo1 expression increased in the lungs after high tidal volume mechanical ventilation and in cyclic stretch-treated cells. Mechanistically, we observed the enhanced expression of RhoA/ROCK1 in both cyclic stretch and Yoda1-treated cells, while the deficiency or inhibition of Piezo1 dramatically antagonized RhoA/ROCK1 expression. Furthermore, blockade of RhoA/ROCK1 signaling using an inhibitor did not affect Piezo1 expression. GSMTx4 was used to inhibit Piezo1, which alleviated VILI-induced pathologic changes, water content and protein leakage in the lungs, and the induction of systemic inflammatory mediators, and improved the 7-day mortality rate in the model rats. Conclusions These findings indicate that Piezo1 affects the development and progression of VILI through promotion of RhoA/ROCK1 signaling.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China
| | - Lulu Jiang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tianfeng Huang
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China
| | - Dahao Lu
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China
| | - Yue Song
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China
| | - Lihui Wang
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China
| | - Ju Gao
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People's Hospital, 98 Nan Tong Western Road, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
26
|
Wong NR, Mohan J, Kopecky BJ, Guo S, Du L, Leid J, Feng G, Lokshina I, Dmytrenko O, Luehmann H, Bajpai G, Ewald L, Bell L, Patel N, Bredemeyer A, Weinheimer CJ, Nigro JM, Kovacs A, Morimoto S, Bayguinov PO, Fisher MR, Stump WT, Greenberg M, Fitzpatrick JAJ, Epelman S, Kreisel D, Sah R, Liu Y, Hu H, Lavine KJ. Resident cardiac macrophages mediate adaptive myocardial remodeling. Immunity 2021; 54:2072-2088.e7. [PMID: 34320366 PMCID: PMC8446343 DOI: 10.1016/j.immuni.2021.07.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Cardiac macrophages represent a heterogeneous cell population with distinct origins, dynamics, and functions. Recent studies have revealed that C-C Chemokine Receptor 2 positive (CCR2+) macrophages derived from infiltrating monocytes regulate myocardial inflammation and heart failure pathogenesis. Comparatively little is known about the functions of tissue resident (CCR2-) macrophages. Herein, we identified an essential role for CCR2- macrophages in the chronically failing heart. Depletion of CCR2- macrophages in mice with dilated cardiomyopathy accelerated mortality and impaired ventricular remodeling and coronary angiogenesis, adaptive changes necessary to maintain cardiac output in the setting of reduced cardiac contractility. Mechanistically, CCR2- macrophages interacted with neighboring cardiomyocytes via focal adhesion complexes and were activated in response to mechanical stretch through a transient receptor potential vanilloid 4 (TRPV4)-dependent pathway that controlled growth factor expression. These findings establish a role for tissue-resident macrophages in adaptive cardiac remodeling and implicate mechanical sensing in cardiac macrophage activation.
Collapse
Affiliation(s)
- Nicole R Wong
- Departmental of Medicine, Washington University School of Medicine
| | - Jay Mohan
- Departmental of Medicine, Washington University School of Medicine
| | | | - Shuchi Guo
- Departmental of Medicine, Washington University School of Medicine
| | - Lixia Du
- Department of Anesthesiology, Washington University School of Medicine
| | - Jamison Leid
- Departmental of Medicine, Washington University School of Medicine
| | - Guoshuai Feng
- Departmental of Medicine, Washington University School of Medicine
| | - Inessa Lokshina
- Departmental of Medicine, Washington University School of Medicine
| | | | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine
| | - Geetika Bajpai
- Departmental of Medicine, Washington University School of Medicine
| | - Laura Ewald
- Departmental of Medicine, Washington University School of Medicine
| | - Lauren Bell
- Departmental of Medicine, Washington University School of Medicine
| | - Nikhil Patel
- Departmental of Genetics, Washington University School of Medicine
| | | | | | - Jessica M Nigro
- Departmental of Medicine, Washington University School of Medicine
| | - Attila Kovacs
- Departmental of Medicine, Washington University School of Medicine
| | - Sachio Morimoto
- Department of Physical Therapy, International University of Health and Welfare, Japan
| | - Peter O Bayguinov
- Department of Biochemistry, Washington University School of Medicine
| | - Max R Fisher
- Department of Biochemistry, Washington University School of Medicine
| | - W Tom Stump
- Department of Biochemistry, Washington University School of Medicine
| | - Michael Greenberg
- Department of Biochemistry, Washington University School of Medicine
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine; Departments of Neuroscience, Cell Biology & Physiology, and Biomedical Engineering, Washington University School of Medicine
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network
| | - Daniel Kreisel
- Department of Pathology and Immunology, Washington University School of Medicine; Department of Surgery, Washington University School of Medicine
| | - Rajan Sah
- Departmental of Medicine, Washington University School of Medicine
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine
| | - Hongzhen Hu
- Department of Anesthesiology, Washington University School of Medicine
| | - Kory J Lavine
- Departmental of Medicine, Washington University School of Medicine; Department of Pathology and Immunology, Washington University School of Medicine; Department of Developmental Biology, Washington University School of Medicine.
| |
Collapse
|
27
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
28
|
Endothelial Transient Receptor Potential V4 Channels Mediate Lung Ischemia-Reperfusion Injury. Ann Thorac Surg 2021; 113:1256-1264. [PMID: 33961815 DOI: 10.1016/j.athoracsur.2021.04.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Lung ischemia-reperfusion injury (IRI), involving severe inflammation and edema, is a major cause of primary graft dysfunction following transplant. Activation of transient receptor potential vanilloid 4 (TRPV4) channels modulates vascular permeability. Thus, this study tests the hypothesis that endothelial TRPV4 channels mediate lung IRI. METHODS C57BL/6 wild-type (WT), TRPV4-/-, tamoxifen-inducible endothelial TRPV4 knockout (TRPV4EC-/-), and tamoxifen-treated control (TRPV4fl/fl) mice underwent lung IR using a left lung hilar-ligation model (n≥6 mice/group). WT mice were also treated with a TRPV4-specific inhibitor (GSK2193874; 1mg/kg) (WT+GSK219). Partial pressure of oxygen (PaO2), edema (wet-to-dry weight ratio), compliance, neutrophil infiltration, and cytokine concentrations in bronchioalveolar lavage fluid were assessed. Pulmonary microvascular endothelial cells (PMVECs) were characterized in vitro following exposure to hypoxia-reoxygenation. RESULTS Compared to WT, PaO2 following IR was significantly improved in TRPV4-/- mice (133.1±43.9 vs 427.8±83.1 mmHg, p<0.001) and WT+GSK219 mice (133.1±43.9 vs 447.0±67.6 mmHg, p<0.001). Pulmonary edema and neutrophil infiltration were also significantly reduced after IR in TRPV4-/- and WT+GSK219 mice versus WT. TRPV4EC-/- mice following IR demonstrated significantly improved oxygenation versus control (109.2±21.6 vs 405.3±41.4 mmHg, p<0.001) as well as significantly improved compliance, and significantly less edema, neutrophil infiltration and proinflammatory cytokine production (TNF-α, CXCL1, IL-17, IFN-γ). Hypoxia-reoxygenation-induced permeability and CXCL1 expression by PMVECs was significantly attenuated by TRPV4 inhibitors. CONCLUSIONS Endothelial TRPV4 plays a key role in vascular permeability and lung inflammation following IR. TRPV4 channels may be a promising therapeutic target to mitigate lung IRI and decrease the incidence of primary graft dysfunction following transplant. (Word Count: 249/250).
Collapse
|
29
|
Gu Q, Lee LY. TRP channels in airway sensory nerves. Neurosci Lett 2021; 748:135719. [PMID: 33587987 PMCID: PMC7988689 DOI: 10.1016/j.neulet.2021.135719] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Transient Receptor Potential (TRP) channels expressed in specific subsets of airway sensory nerves function as transducers and integrators of a diverse range of sensory inputs including chemical, mechanical and thermal signals. These TRP sensors can detect inhaled irritants as well as endogenously released chemical substances. They play an important role in generating the afferent activity carried by these sensory nerves and regulating the centrally mediated pulmonary defense reflexes. Increasing evidence reported in recent investigations has revealed important involvements of several TRP channels (TRPA1, TRPV1, TRPV4 and TRPM8) in the manifestation of various symptoms and pathogenesis of certain acute and chronic airway diseases. This mini-review focuses primarily on these recent findings of the responses of these TRP sensors to the biological stresses emerging under the pathophysiological conditions of the lung and airways.
Collapse
Affiliation(s)
- Qihai Gu
- Department of Biomedical Sciences, Mercer University School of Medicine, 1501 Mercer University Drive, Macon, GA, 31207, USA.
| | - Lu-Yuan Lee
- Department of Physiology, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA.
| |
Collapse
|
30
|
Issa NT, Stathias V, Schürer S, Dakshanamurthy S. Machine and deep learning approaches for cancer drug repurposing. Semin Cancer Biol 2021; 68:132-142. [PMID: 31904426 PMCID: PMC7723306 DOI: 10.1016/j.semcancer.2019.12.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/31/2019] [Accepted: 12/15/2019] [Indexed: 02/07/2023]
Abstract
Knowledge of the underpinnings of cancer initiation, progression and metastasis has increased exponentially in recent years. Advanced "omics" coupled with machine learning and artificial intelligence (deep learning) methods have helped elucidate targets and pathways critical to those processes that may be amenable to pharmacologic modulation. However, the current anti-cancer therapeutic armamentarium continues to lag behind. As the cost of developing a new drug remains prohibitively expensive, repurposing of existing approved and investigational drugs is sought after given known safety profiles and reduction in the cost barrier. Notably, successes in oncologic drug repurposing have been infrequent. Computational in-silico strategies have been developed to aid in modeling biological processes to find new disease-relevant targets and discovering novel drug-target and drug-phenotype associations. Machine and deep learning methods have especially enabled leaps in those successes. This review will discuss these methods as they pertain to cancer biology as well as immunomodulation for drug repurposing opportunities in oncologic diseases.
Collapse
Affiliation(s)
- Naiem T Issa
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami School of Medicine, Miami, FL, USA
| | - Vasileios Stathias
- Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, FL, USA
| | - Stephan Schürer
- Department of Molecular and Cellular Pharmacology, University of Miami School of Medicine, Miami, FL, USA
| | - Sivanesan Dakshanamurthy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
31
|
Lu Q, Zemskov EA, Sun X, Wang H, Yegambaram M, Wu X, Garcia-Flores A, Song S, Tang H, Kangath A, Cabanillas GZ, Yuan JXJ, Wang T, Fineman JR, Black SM. Activation of the mechanosensitive Ca 2+ channel TRPV4 induces endothelial barrier permeability via the disruption of mitochondrial bioenergetics. Redox Biol 2021; 38:101785. [PMID: 33221570 PMCID: PMC7691184 DOI: 10.1016/j.redox.2020.101785] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/20/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS), a refractory lung disease with an unacceptable high mortality rate. Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The specific mechanisms involved in VILI-induced pulmonary capillary leakage, a key pathologic feature of VILI are still far from resolved. The mechanoreceptor, transient receptor potential cation channel subfamily V member 4, TRPV4 plays a key role in the development of VILI through unresolved mechanism. Endothelial nitric oxide synthase (eNOS) uncoupling plays an important role in sepsis-mediated ARDS so in this study we investigated whether there is a role for eNOS uncoupling in the barrier disruption associated with TRPV4 activation during VILI. Our data indicate that the TRPV4 agonist, 4α-Phorbol 12,13-didecanoate (4αPDD) induces pulmonary arterial endothelial cell (EC) barrier disruption through the disruption of mitochondrial bioenergetics. Mechanistically, this occurs via the mitochondrial redistribution of uncoupled eNOS secondary to a PKC-dependent phosphorylation of eNOS at Threonine 495 (T495). A specific decoy peptide to prevent T495 phosphorylation reduced eNOS uncoupling and mitochondrial redistribution and preserved PAEC barrier function under 4αPDD challenge. Further, our eNOS decoy peptide was able to preserve lung vascular integrity in a mouse model of VILI. Thus, we have revealed a functional link between TRPV4 activation, PKC-dependent eNOS phosphorylation at T495, and EC barrier permeability. Reducing pT495-eNOS could be a new therapeutic approach for the prevention of VILI.
Collapse
Affiliation(s)
- Qing Lu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xutong Sun
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Hui Wang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Manivannan Yegambaram
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Alejandro Garcia-Flores
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Shanshan Song
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Haiyang Tang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Archana Kangath
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Gabriela Zubiate Cabanillas
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemist-Biological Sciences, Universidad de Sonora, Hermosillo, SON, Mexico
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, CA, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Stephen M Black
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
32
|
Fu S, Meng H, Inamdar S, Das B, Gupta H, Wang W, Thompson CL, Knight MM. Activation of TRPV4 by mechanical, osmotic or pharmaceutical stimulation is anti-inflammatory blocking IL-1β mediated articular cartilage matrix destruction. Osteoarthritis Cartilage 2021; 29:89-99. [PMID: 33395574 PMCID: PMC7799379 DOI: 10.1016/j.joca.2020.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Cartilage health is maintained in response to a range of mechanical stimuli including compressive, shear and tensile strains and associated alterations in osmolality. The osmotic-sensitive ion channel Transient Receptor Potential Vanilloid 4 (TRPV4) is required for mechanotransduction. Mechanical stimuli inhibit interleukin-1β (IL-1β) mediated inflammatory signalling, however the mechanism is unclear. This study aims to clarify the role of TRPV4 in this response. DESIGN TRPV4 activity was modulated glycogen synthase kinase (GSK205 antagonist or GSK1016790 A (GSK101) agonist) in articular chondrocytes and cartilage explants in the presence or absence of IL-1β, mechanical (10% cyclic tensile strain (CTS), 0.33 Hz, 24hrs) or osmotic loading (200mOsm, 24hrs). Nitric oxide (NO), prostaglandin E2 (PGE2) and sulphated glycosaminoglycan (sGAG) release and cartilage biomechanics were analysed. Alterations in post-translational tubulin modifications and primary cilia length regulation were examined. RESULTS In isolated chondrocytes, mechanical loading inhibited IL-1β mediated NO and PGE2 release. This response was inhibited by GSK205. Similarly, osmotic loading was anti-inflammatory in cells and explants, this response was abrogated by TRPV4 inhibition. In explants, GSK101 inhibited IL-1β mediated NO release and prevented cartilage degradation and loss of mechanical properties. Upon activation, TRPV4 cilia localisation was increased resulting in histone deacetylase 6 (HDAC6)-dependent modulation of soluble tubulin and altered cilia length regulation. CONCLUSION Mechanical, osmotic or pharmaceutical activation of TRPV4 regulates HDAC6-dependent modulation of ciliary tubulin and is anti-inflammatory. This study reveals for the first time, the potential of TRPV4 manipulation as a novel therapeutic mechanism to supress pro-inflammatory signalling and cartilage degradation.
Collapse
Affiliation(s)
- S Fu
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - H Meng
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - S Inamdar
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - B Das
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK
| | - H Gupta
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - W Wang
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - C L Thompson
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| | - M M Knight
- Centre for Predictive In Vitro Models, School of Engineering and Materials Science, Queen Mary University of London, UK.
| |
Collapse
|
33
|
Lee K, Byun J, Kim B, Yeon J, Tai J, Lee SH, Kim TH. TRPV4-Mediated Epithelial Junction Disruption in Allergic Rhinitis Triggered by House Dust Mites. Am J Rhinol Allergy 2020; 35:432-440. [PMID: 33012175 DOI: 10.1177/1945892420964169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Epithelial barrier disruption is a crucial feature of allergic rhinitis (AR). Previous reports have indicated the role of transient receptor potential vanilloid (TRPV) 4 in regulating the intercellular junctions in various cells. However, the role of TRPV4 and its regulation by T helper 2 cell cytokines in the epithelial cells of patients with AR remains unclear. OBJECTIVE We aimed to elucidate the expression of TRPV4 in nasal epithelial cells and its cytokine-induced regulation, and to reveal its role in house dust mite-induced junction disruption in AR. METHODS The expression of TRPV4 in nasal epithelial cells was measured using real-time polymerase chain reaction, western blot, and immunohistochemical assays, and the expression levels were compared between the patients with AR and healthy controls. Altered expression of TRPV4 was induced in cultured nasal epithelial cells by stimulation of interleukin (IL) 4, IL-13, and tumor necrosis factor alpha. In addition, expression of E-cadherin and zonula occludens 1 was induced in Der p 1-stimulated epithelial cells by treatment with either a TRPV4 agonist (GSK1016790A) or a TRPV4 antagonist (RN1734). RESULTS TRPV4 expression was increased in epithelial cells harvested from the affected turbinates compared to those from the normal turbinates. The stimulation of cultured epithelial cells with IL-4 and IL-13 resulted in TRPV4 upregulation. Additionally, E-cadherin and zonula occludens 1 expression levels decreased in the cultured epithelial cells treated with GSK1016790A after stimulation with Der p 1, whereas Der p 1 stimulation alone showed no effect on junctional protein expression. CONCLUSIONS Increased TRPV4 expression occurred in epithelial cells harvested from patients with AR and epithelial cells stimulated by Th2 cytokines. Decreased junctional protein expression in epithelial cells after the stimulation by house dust mite allergen with TRPV4 agonist indicates a possible role of TRPV4 in the pathogenesis of allergen-induced epithelial barrier disruption in AR.
Collapse
Affiliation(s)
- Kijeong Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| | - Junhyoung Byun
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| | - Byoungjae Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea.,Neuroscience Research Institute, College of Medicine, Korea University, Seoul, Korea
| | - Jiwoo Yeon
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| | - Junhu Tai
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| | - Sang Hag Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
34
|
Lapajne L, Lakk M, Yarishkin O, Gubeljak L, Hawlina M, Križaj D. Polymodal Sensory Transduction in Mouse Corneal Epithelial Cells. Invest Ophthalmol Vis Sci 2020; 61:2. [PMID: 32271891 PMCID: PMC7401707 DOI: 10.1167/iovs.61.4.2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Contact lenses, osmotic stressors, and chemical burns may trigger severe discomfort and vision loss by damaging the cornea, but the signaling mechanisms used by corneal epithelial cells (CECs) to sense extrinsic stressors are not well understood. We therefore investigated the mechanisms of swelling, temperature, strain, and chemical transduction in mouse CECs. Methods Intracellular calcium imaging in conjunction with electrophysiology, pharmacology, transcript analysis, immunohistochemistry, and bioluminescence assays of adenosine triphosphate (ATP) release were used to track mechanotransduction in dissociated CECs and epithelial sheets isolated from the mouse cornea. Results The transient receptor potential vanilloid (TRPV) transcriptome in the mouse corneal epithelium is dominated by Trpv4, followed by Trpv2, Trpv3, and low levels of Trpv1 mRNAs. TRPV4 protein was localized to basal and intermediate epithelial strata, keratocytes, and the endothelium in contrast to the cognate TRPV1, which was confined to intraepithelial afferents and a sparse subset of CECs. The TRPV4 agonist GSK1016790A induced cation influx and calcium elevations, which were abolished by the selective blocker HC067047. Hypotonic solutions, membrane strain, and moderate heat elevated [Ca2+]CEC with swelling- and temperature-, but not strain-evoked signals, sensitive to HC067047. GSK1016790A and swelling evoked calcium-dependent ATP release, which was suppressed by HC067027 and the hemichannel blocker probenecid. Conclusions These results demonstrate that cation influx via TRPV4 transduces osmotic and thermal but not strain inputs to CECs and promotes hemichannel-dependent ATP release. The TRPV4-hemichannel-ATP signaling axis might modulate corneal pain induced by excessive mechanical, osmotic, and chemical stimulation.
Collapse
|
35
|
Kuebler WM, Jordt SE, Liedtke WB. Urgent reconsideration of lung edema as a preventable outcome in COVID-19: inhibition of TRPV4 represents a promising and feasible approach. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1239-L1243. [PMID: 32401673 PMCID: PMC7276984 DOI: 10.1152/ajplung.00161.2020] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Lethality of coronavirus disease (COVID-19) during the 2020 pandemic, currently still in the exponentially accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We argue for inhibition of the transient receptor potential vanilloid 4 (TRPV4) calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in COVID-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in COVID-19 patients with respiratory malfunction and at risk for lung edema. Perplexingly, among the currently pursued therapeutic strategies against COVID-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce COVID-19 lethality but will also preempt a distressing healthcare scenario with insufficient capacity to provide ventilator-assisted respiration.
Collapse
Affiliation(s)
- Wolfgang M. Kuebler
- 1Institute of Physiology, Charité Medical University of Berlin, Berlin, Germany
| | - Sven-Eric Jordt
- 2Department of Anesthesiology, Duke University, Durham, North Carolina
| | - Wolfgang B. Liedtke
- 2Department of Anesthesiology, Duke University, Durham, North Carolina,3Department of Neurology, Duke University, Durham, North Carolina,4Department of Neurobiology, Duke University, Durham, North Carolina
| |
Collapse
|
36
|
Rosenbaum T, Benítez-Angeles M, Sánchez-Hernández R, Morales-Lázaro SL, Hiriart M, Morales-Buenrostro LE, Torres-Quiroz F. TRPV4: A Physio and Pathophysiologically Significant Ion Channel. Int J Mol Sci 2020; 21:ijms21113837. [PMID: 32481620 PMCID: PMC7312103 DOI: 10.3390/ijms21113837] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/23/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023] Open
Abstract
Transient Receptor Potential (TRP) channels are a family of ion channels whose members are distributed among all kinds of animals, from invertebrates to vertebrates. The importance of these molecules is exemplified by the variety of physiological roles they play. Perhaps, the most extensively studied member of this family is the TRPV1 ion channel; nonetheless, the activity of TRPV4 has been associated to several physio and pathophysiological processes, and its dysfunction can lead to severe consequences. Several lines of evidence derived from animal models and even clinical trials in humans highlight TRPV4 as a therapeutic target and as a protein that will receive even more attention in the near future, as will be reviewed here.
Collapse
Affiliation(s)
- Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.B.-A.); (R.S.-H.); (S.L.M.-L.); (M.H.)
- Correspondence: ; Tel.: +52-555-622-56-24; Fax: +52-555-622-56-07
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.B.-A.); (R.S.-H.); (S.L.M.-L.); (M.H.)
| | - Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.B.-A.); (R.S.-H.); (S.L.M.-L.); (M.H.)
| | - Sara Luz Morales-Lázaro
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.B.-A.); (R.S.-H.); (S.L.M.-L.); (M.H.)
| | - Marcia Hiriart
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.B.-A.); (R.S.-H.); (S.L.M.-L.); (M.H.)
| | - Luis Eduardo Morales-Buenrostro
- Departamento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City 14080, Mexico;
| | - Francisco Torres-Quiroz
- Departamento de Bioquímica y Biología Estructural, División Investigación Básica, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| |
Collapse
|
37
|
Kuebler WM, Jordt SE, Liedtke WB. COVID-19: urgent reconsideration of lung edema as a preventable outcome Inhibition of TRPV4 as a promising and feasible approach. SSRN 2020:3558887. [PMID: 32714108 PMCID: PMC7366813 DOI: 10.2139/ssrn.3558887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/21/2020] [Indexed: 02/06/2023]
Abstract
Lethality of Covid-19 during the 2020 pandemic, currently in the exponentially-accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of SARS-CoV-2 infection. We argue for inhibition of the TRPV4 calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema, and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in Covid-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in Covid-19 patients with respiratory malfunction and at risk for lung edema. We note that among the currently pursued therapeutic strategies against Covid-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce Covid-19 lethality but will pre-empt a catastrophic scenario in healthcare with insufficient capacity to provide ventilator-assisted respiration.
Collapse
Affiliation(s)
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University, Durham NC, USA
| | - Wolfgang B Liedtke
- Department of Anesthesiology, Duke University, Durham NC, USA
- Department of Neurology, Duke University, Durham NC, USA
- Department of Neurobiology, Duke University, Durham NC, USA
| |
Collapse
|
38
|
Michalick L, Kuebler WM. TRPV4-A Missing Link Between Mechanosensation and Immunity. Front Immunol 2020; 11:413. [PMID: 32210976 PMCID: PMC7076180 DOI: 10.3389/fimmu.2020.00413] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential vanilloid-type 4 (TRPV4) cation channel is widely expressed in all tissues as well as in immune cells and its function as mechanosensitive Ca2+ channel seems to be conserved throughout all mammalian species. Of late, emerging evidence has implicated TRPV4 in the activation and differentiation of innate immune cells, especially in neutrophils, monocytes, and macrophages. As such, TRPV4 has been shown to mediate neutrophil adhesion and chemotaxis, as well as production of reactive oxygen species in response to pro-inflammatory stimuli. In macrophages, TRPV4 mediates formation of both reactive oxygen and nitrogen species, and regulates phagocytosis, thus facilitating bacterial clearance and resolution of infection. Importantly, TRPV4 may present a missing link between mechanical forces and immune responses. This connection has been exemplary highlighted by the demonstrated role of TRPV4 in macrophage activation and subsequent induction of lung injury following mechanical overventilation. Mechanosensation via TRPV4 is also expected to activate innate immune cells and establish a pro-inflammatory loop in fibrotic diseases with increased deposition of extracellular matrix (ECM) and substrate stiffness. Likewise, TRPV4 may be activated by cell migration through the endothelium or the extracellular matrix, or even by circulating immune cells squeezing through the narrow passages of the pulmonary or systemic capillary bed, a process that has recently been linked to neutrophil priming and depriming. Here, we provide an overview over the emerging role of TRPV4 in innate immune responses and highlight two distinct modes for the activation of TRPV4 by either mechanical forces ("mechanoTRPV4") or by pathogens ("immunoTRPV4").
Collapse
Affiliation(s)
- Laura Michalick
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Physiology, Berlin Institute of Health, Berlin, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Institute of Physiology, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
39
|
Yu Q, Wang D, Wen X, Tang X, Qi D, He J, Zhao Y, Deng W, Zhu T. Adipose-derived exosomes protect the pulmonary endothelial barrier in ventilator-induced lung injury by inhibiting the TRPV4/Ca 2+ signaling pathway. Am J Physiol Lung Cell Mol Physiol 2020; 318:L723-L741. [PMID: 32073873 PMCID: PMC7191475 DOI: 10.1152/ajplung.00255.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mechanical ventilation (MV) is the main supportive treatment of acute respiratory distress syndrome (ARDS), but it may lead to ventilator-induced lung injury (VILI). Large epidemiological studies have found that obesity was associated with lower mortality in mechanically ventilated patients with acute lung injury, which is known as “obesity paradox.” However, the effects of obesity on VILI are unknown. In the present study, wild-type mice were fed a high-fat diet (HFD) and ventilated with high tidal volume to investigate the effects of obesity on VILI in vivo, and pulmonary microvascular endothelial cells (PMVECs) were subjected to 18% cyclic stretching (CS) to further investigate its underlying mechanism in vitro. We found that HFD protects mice from VILI by alleviating the pulmonary endothelial barrier injury and inflammatory responses in mice. Adipose-derived exosomes can regulate distant tissues as novel adipokines, providing a new mechanism for cell-cell interactions. We extracted three adipose-derived exosomes, including HFD mouse serum exosome (S-Exo), adipose tissue exosome (AT-Exo), and adipose-derived stem cell exosome (ADSC-Exo), and further explored their effects on MV or 18% CS-induced VILI in vivo and in vitro. Administration of three exosomes protected against VILI by suppressing pulmonary endothelial barrier hyperpermeability, repairing the expression of adherens junctions, and alleviating inflammatory response in vivo and in vitro, accompanied by transient receptor potential vanilloid 4 (TRPV4)/Ca2+ pathway inhibition. Collectively, these data indicated that HFD-induced obesity plays a protective role in VILI by alleviating the pulmonary endothelial barrier injury and inflammatory response via adipose-derived exosomes, at least partially, through inhibiting the TRPV4/Ca2+ pathway.
Collapse
Affiliation(s)
- Qian Yu
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daoxin Wang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoting Wen
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xumao Tang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Di Qi
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zhao
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Deng
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Zhu
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Role of macrophage TRPV4 in inflammation. J Transl Med 2020; 100:178-185. [PMID: 31645630 PMCID: PMC7261496 DOI: 10.1038/s41374-019-0334-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/19/2019] [Accepted: 08/30/2019] [Indexed: 01/05/2023] Open
Abstract
Transient receptor ion channels have emerged as immensely important channels/receptors in diverse physiological and pathological responses. Of particular interest is the transient receptor potential channel subfamily V member 4 (TRPV4), which is a polymodal, nonselective, calcium-permeant cation channel, and is activated by both endogenous and exogenous stimuli. Both neuronal and nonneuronal cells express functional TRPV4, which is responsive to a variety of biochemical and biomechanical stimuli. Emerging discoveries have advanced our understanding of the role of macrophage TRPV4 in numerous inflammatory diseases. In lung injury, TRPV4 mediates macrophage phagocytosis, secretion of pro-resolution cytokines, and generation of reactive oxygen species. TRPV4 regulates lipid-laden macrophage foam cell formation, the hallmark of atheroinflammatory conditions, in response to matrix stiffness and lipopolysaccharide stimulation. Accumulating data also point to a role of macrophage TRPV4 in the pathogenesis of the foreign body response, a chronic inflammatory condition, through the formation of foreign body giant cells. Deletion of TRPV4 in macrophages suppresses the allergic and nonallergic itch in a mouse model, suggesting a role of TRPV4 in skin disease. Here, we discuss the current understanding of the role of macrophage TRPV4 in various inflammatory conditions.
Collapse
|
41
|
Li Y, Talotta-Altenburg LM, Silimperi KA, Ciabattoni GO, Lowe-Krentz LJ. Endothelial nitric oxide synthase activation is required for heparin receptor effects on vascular smooth muscle cells. Am J Physiol Cell Physiol 2019; 318:C463-C475. [PMID: 31891520 DOI: 10.1152/ajpcell.00284.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Published studies indicate that TMEM184A is a heparin receptor that interacts with and transduces stimulation from heparin in vascular cells. Previous studies have indicated that heparin increases endothelial nitric oxide synthase (eNOS) activity in bovine endothelial cells. However, the precise mechanism remains unknown. In this study, we investigated the impact of heparin treatment and TMEM184A on eNOS's activation and the role of eNOS in heparin signaling in the cloned A7r5 rat vascular smooth muscle cell line and confirmed results in endothelial cells. We employed a combination of TMEM184A knockdown A7r5 cells along with transient eNOS knockdown and enzyme inhibitor strategies. The results indicate that heparin induces phosphorylation of eNOS. eNOS can be immunoprecipitated with TMEM184A and is internalized to the perinuclear region in a TMEM184A-dependent manner in response to heparin. We also examined how heparin treatment leads to phosphorylation of eNOS and confirmed that TMEM184A and Ca2+ were required to mediate heparin-elicited eNOS phosphorylation. Evidence supporting the involvement of transient receptor potential cation channel subfamily V member 4 with TMEM184A in this eNOS activation process is also presented.
Collapse
Affiliation(s)
- Yaqiu Li
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | | | - Kayli A Silimperi
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | - Grace O Ciabattoni
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| | - Linda J Lowe-Krentz
- Department of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania
| |
Collapse
|
42
|
Li M, Fang XZ, Liu XT, Zheng YF, Xie YB, Ma XD, Xia Y, Shao DH. Inhibition of calcineurin/NFATc4 signaling attenuates ventilator‑induced lung injury. Mol Med Rep 2019; 21:607-614. [PMID: 31789412 PMCID: PMC6947850 DOI: 10.3892/mmr.2019.10851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is a life-threatening condition caused by the inappropriate use of mechanical ventilation (MV). However, the precise molecular mechanism inducing the development of VILI remains to be elucidated. In the present study, it was revealed that the calcineurin/NFATc4 signaling pathway mediates the expression of adhesion molecules and proinflammatory cytokines essential for the development of VILI. The present results revealed that a high tidal volume ventilation (HV) caused lung inflammation and edema in the alveolar walls and the infiltration of inflammatory cells. The calcineurin activity and protein expression in the lungs were increased in animals with VILI, and NFATc4 translocated into the nucleus following calcineurin activation. Furthermore, the translocation of NFATc4 and lung injury were prevented by a calcineurin inhibitor (CsA). Thus, the present results highlighted the critical role of the calcineurin/NFATc4 signaling pathway in VILI and suggest that this pathway coincides with the release of ICAM-1, VCAM-1, TNF-α and IL-1β.
Collapse
Affiliation(s)
- Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Xiang-Zhi Fang
- Department of Anesthesiology, Clinical Medical School of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Xiao-Tian Liu
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yong-Feng Zheng
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yun-Bin Xie
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Xiao-Dong Ma
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yan Xia
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Dong-Hua Shao
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| |
Collapse
|
43
|
Yu S, Huang S, Ding Y, Wang W, Wang A, Lu Y. Transient receptor potential ion-channel subfamily V member 4: a potential target for cancer treatment. Cell Death Dis 2019; 10:497. [PMID: 31235786 PMCID: PMC6591233 DOI: 10.1038/s41419-019-1708-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 12/29/2022]
Abstract
The transient receptor potential ion-channel superfamily consists of nonselective cation channels located mostly on the plasma membranes of numerous animal cell types, which are closely related to sensory information transmission (e.g., vision, pain, and temperature perception), as well as regulation of intracellular Ca2+ balance and physiological activities of growth and development. Transient receptor potential ion channel subfamily V (TRPV) is one of the largest and most diverse subfamilies, including TRPV1-TRPV6 involved in the regulation of a variety of cellular functions. TRPV4 can be activated by various physical and chemical stimuli, such as heat, mechanical force, and phorbol ester derivatives participating in the maintenance of normal cellular functions. In recent years, the roles of TRPV4 in cell proliferation, differentiation, apoptosis, and migration have been extensively studied. Its abnormal expression has also been closely related to the onset and progression of multiple tumors, so TRPV4 may be a target for cancer diagnosis and treatment. In this review, we focused on the latest studies concerning the role of TRPV4 in tumorigenesis and the therapeutic potential. As evidenced by the effects on cancerogenesis, TRPV4 is a potential target for anticancer therapy.
Collapse
Affiliation(s)
- Suyun Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Shuai Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yushi Ding
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Wei Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
| |
Collapse
|
44
|
TRPV4-induced inflammatory response is involved in neuronal death in pilocarpine model of temporal lobe epilepsy in mice. Cell Death Dis 2019; 10:386. [PMID: 31097691 PMCID: PMC6522539 DOI: 10.1038/s41419-019-1612-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/21/2019] [Accepted: 04/24/2019] [Indexed: 01/02/2023]
Abstract
Activation of transient receptor potential vanilloid 4 (TRPV4) induces neuronal injury. TRPV4 activation enhances inflammatory response and promotes the proinflammatory cytokine release in various types of tissue and cells. Hyperneuroinflammation contributes to neuronal damage in epilepsy. Herein, we examined the contribution of neuroinflammation to TRPV4-induced neurotoxicity and its involvement in the inflammation and neuronal damage in pilocarpine model of temporal lobe epilepsy in mice. Icv. injection of TRPV4 agonist GSK1016790A (GSK1016790A-injected mice) increased ionized calcium binding adapter molecule-1 (Iba-1) and glial fibrillary acidic protein (GFAP) protein levels and Iba-1-positive (Iba-1+) and GFAP-positive (GFAP+) cells in hippocampi, which indicated TRPV4-induced microglial cell and astrocyte activation. The protein levels of nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3) inflammasome components NLRP3, apoptosis-related spotted protein (ASC) and cysteinyl aspartate-specific protease-1 (caspase-1) were increased in GSK1016790A-injected mice, which indicated NLRP3 inflammasome activation. GSK1016790A also increased proinflammatory cytokine IL-1β, TNF-α and IL-6 protein levels, which were blocked by caspase-1 inhibitor Ac-YVAD-cmk. GSK1016790A-induced neuronal death was attenuated by Ac-YVAD-cmk. Icv. injection of TRPV4-specific antagonist HC-067047 markedly increased the number of surviving cells 3 d post status epilepticus in pilocarpine model of temporal lobe epilepsy in mice (pilocarpine-induced status epilepticus, PISE). HC-067047 also markedly blocked the increase in Iba-1 and GFAP protein levels, as well as Iba-1+ and GFAP+ cells 3 d post-PISE. Finally, the increased protein levels of NLRP3, ASC and caspase-1 as well as IL-1β, TNF-α and IL-6 were markedly blocked by HC-067047. We conclude that TRPV4-induced neuronal death is mediated at least partially by enhancing the neuroinflammatory response, and this action is involved in neuronal injury following status epilepticus.
Collapse
|
45
|
Orduña Ríos M, Noguez Imm R, Hernández Godínez NM, Bautista Cortes AM, López Escalante DD, Liedtke W, Martínez Torres A, Concha L, Thébault S. TRPV4 inhibition prevents increased water diffusion and blood-retina barrier breakdown in the retina of streptozotocin-induced diabetic mice. PLoS One 2019; 14:e0212158. [PMID: 31048895 PMCID: PMC6497373 DOI: 10.1371/journal.pone.0212158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/09/2019] [Indexed: 01/02/2023] Open
Abstract
A better understanding of the molecular and cellular mechanisms involved in retinal hydro-mineral homeostasis imbalance during diabetic macular edema (DME) is needed to gain insights into retinal (patho-)physiology that will help elaborate innovative therapies with lower health care costs. Transient receptor potential cation channel subfamily vanilloid member 4 (TRPV4) plays an intricate role in homeostatic processes that needs to be deciphered in normal and diabetic retina. Based on previous findings showing that TRPV4 antagonists resolve blood-retina barrier (BRB) breakdown in diabetic rats, we evaluated whether TRPV4 channel inhibition prevents and reverts retinal edema in streptozotocin(STZ)-induced diabetic mice. We assessed retinal edema using common metrics, including retinal morphology/thickness (histology) and BRB integrity (albumin-associated tracer), and also by quantifying water mobility through apparent diffusion coefficient (ADC) measures. ADC was measured by diffusion-weighted magnetic resonance imaging (DW-MRI), acquired ex vivo at 4 weeks after STZ injection in diabetes and control groups. DWI images were also used to assess retinal thickness. TRPV4 was genetically ablated or pharmacologically inhibited as follows: left eyes were used as vehicle control and right eyes were intravitreally injected with TRPV4-selective antagonist GSK2193874, 24 h before the end of the 4 weeks of diabetes. Histological data show that retinal thickness was similar in nondiabetic and diabetic wt groups but increased in diabetic Trpv4-/- mice. In contrast, DWI shows retinal thinning in diabetic wt mice that was absent in diabetic Trpv4-/- mice. Disorganized outer nuclear layer was observed in diabetic wt but not in diabetic Trpv4-/- retinas. We further demonstrate increased water diffusion, increased distances between photoreceptor nuclei, reduced nuclear area in all nuclear layers, and BRB hyperpermeability, in diabetic wt mice, effects that were absent in diabetic Trpv4-/- mice. Retinas of diabetic mice treated with PBS showed increased water diffusion that was not normalized by GSK2193874. ADC maps in nondiabetic Trpv4-/- mouse retinas showed restricted diffusion. Our data provide evidence that water diffusion is increased in diabetic mouse retinas and that TRPV4 function contributes to retinal hydro-mineral homeostasis and structure under control conditions, and to the development of BRB breakdown and increased water diffusion in the retina under diabetes conditions. A single intravitreous injection of TRPV4 antagonist is however not sufficient to revert these alterations in diabetic mouse retinas.
Collapse
Affiliation(s)
- Maricruz Orduña Ríos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Ramsés Noguez Imm
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Ana María Bautista Cortes
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | | | - Wolfgang Liedtke
- Department of Medicine and Neurobiology, Center for Translational Neuroscience, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Atáulfo Martínez Torres
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Luis Concha
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
| | - Stéphanie Thébault
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus UNAM-Juriquilla, Querétaro, México
- * E-mail:
| |
Collapse
|
46
|
Kameda T, Zvick J, Vuk M, Sadowska A, Tam WK, Leung VY, Bölcskei K, Helyes Z, Applegate LA, Hausmann ON, Klasen J, Krupkova O, Wuertz-Kozak K. Expression and Activity of TRPA1 and TRPV1 in the Intervertebral Disc: Association with Inflammation and Matrix Remodeling. Int J Mol Sci 2019; 20:E1767. [PMID: 30974795 PMCID: PMC6480240 DOI: 10.3390/ijms20071767] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/28/2019] [Accepted: 03/30/2019] [Indexed: 01/12/2023] Open
Abstract
Transient receptor potential (TRP) channels have emerged as potential sensors and transducers of inflammatory pain. The aims of this study were to investigate (1) the expression of TRP channels in intervertebral disc (IVD) cells in normal and inflammatory conditions and (2) the function of Transient receptor potential ankyrin 1 (TRPA1) and Transient receptor potential vanilloid 1 (TRPV1) in IVD inflammation and matrix homeostasis. RT-qPCR was used to analyze human fetal, healthy, and degenerated IVD tissues for the gene expression of TRPA1 and TRPV1. The primary IVD cell cultures were stimulated with either interleukin-1 beta (IL-1β) or tumor necrosis factor alpha (TNF-α) alone or in combination with TRPA1/V1 agonist allyl isothiocyanate (AITC, 3 and 10 µM), followed by analysis of calcium flux and the expression of inflammation mediators (RT-qPCR/ELISA) and matrix constituents (RT-qPCR). The matrix structure and composition in caudal motion segments from TRPA1 and TRPV1 wild-type (WT) and knock-out (KO) mice was visualized by FAST staining. Gene expression of other TRP channels (A1, C1, C3, C6, V1, V2, V4, V6, M2, M7, M8) was also tested in cytokine-treated cells. TRPA1 was expressed in fetal IVD cells, 20% of degenerated IVDs, but not in healthy mature IVDs. TRPA1 expression was not detectable in untreated cells and it increased upon cytokine treatment, while TRPV1 was expressed and concomitantly reduced. In inflamed IVD cells, 10 µM AITC activated calcium flux, induced gene expression of IL-8, and reduced disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) and collagen 1A1, possibly via upregulated TRPA1. TRPA1 KO in mice was associated with signs of degeneration in the nucleus pulposus and the vertebral growth plate, whereas TRPV1 KO did not show profound changes. Cytokine treatment also affected the gene expression of TRPV2 (increase), TRPV4 (increase), and TRPC6 (decrease). TRPA1 might be expressed in developing IVD, downregulated during its maturation, and upregulated again in degenerative disc disease, participating in matrix homeostasis. However, follow-up studies with larger sample sizes are needed to fully elucidate the role of TRPA1 and other TRP channels in degenerative disc disease.
Collapse
Affiliation(s)
- Takuya Kameda
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
- Department of Orthopaedic Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima City, Fukushima 960-1295, Japan.
| | - Joel Zvick
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
| | - Miriam Vuk
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
| | - Aleksandra Sadowska
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
| | - Wai Kit Tam
- Department of Orthopaedics and Traumatology, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China.
| | - Victor Y Leung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China.
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, University of Pécs, H-7624 Pécs, Szigeti út 12., Hungary.
- János Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, University of Pécs, H-7624 Pécs, Szigeti út 12., Hungary.
- János Szentágothai Research Centre, University of Pécs, H-7624 Pécs, Ifjúság útja 20., Hungary.
| | - Lee Ann Applegate
- Department of Musculoskeletal Medicine, Unit of Regenerative Therapy (UTR), University Hospital Lausanne, EPCR/02 Chemin des Croisettes 22, 1066 Epalinges, Switzerland.
| | - Oliver N Hausmann
- Neuro- and Spine Center, St. Anna Hospital, Sankt-Anna-Strasse 32, 6006 Luzern, Switzerland.
| | - Juergen Klasen
- Clinic Prodorso, Walchestrasse 15, 8006 Zurich, Switzerland.
| | - Olga Krupkova
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
| | - Karin Wuertz-Kozak
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, 8093 Zurich, Switzerland.
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (AU), Harlachinger Str. 51, 81547 Munich, Germany.
- Department of Health Sciences, University of Potsdam, Am Neuen Palais 10, 14469 Potsdam, Germany.
| |
Collapse
|