1
|
Sánchez-Hernández R, Benítez-Angeles M, Hernández-Vega AM, Rosenbaum T. Recent advances on the structure and the function relationships of the TRPV4 ion channel. Channels (Austin) 2024; 18:2313323. [PMID: 38354101 PMCID: PMC10868539 DOI: 10.1080/19336950.2024.2313323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.
Collapse
Affiliation(s)
- Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Ana M. Hernández-Vega
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
2
|
Qi WH, Tang N, Zhao ZJ, Li XQ. Transient receptor potential channels in viral infectious diseases: Biological characteristics and regulatory mechanisms. J Adv Res 2024:S2090-1232(24)00541-1. [PMID: 39551130 DOI: 10.1016/j.jare.2024.11.022] [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: 06/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Viral infectious diseases have long posed a challenge to humanity. In recent decades, transient receptor potential (TRP) channels have emerged as newly investigated cation channels. Increasing evidence suggests that TRP channel-mediated Ca2+ homeostasis disruptions, along with associated pathological changes, are critical factors in the onset and progression of viral infectious diseases. However, the precise roles and mechanisms of TRP channels in these diseases remain to be systematically elucidated. AIM OF REVIEW The aim of this review is to systematically summarize recent advances in understanding TRP channels in viral infections, and based on current progress and challenges, propose future directions for research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review summarizes the classification and biological functions of the TRP family, explores the mechanisms by which TRP channels contribute to viral infections, and highlights specific mechanisms at three levels: virus, host, and outcome. These include the direct role in viral biology and replication, the indirect role in host immunity and inflammation, and the resulting pathological changes. Additionally, we discuss the potential applications of the TRP family in the treatment of viral infectious diseases and propose future research directions.
Collapse
Affiliation(s)
- Wen-Hui Qi
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Na Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Zhi-Jing Zhao
- Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China; Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
3
|
Koskimäki S, Tojkander S. TRPV4-A Multifunctional Cellular Sensor Protein with Therapeutic Potential. SENSORS (BASEL, SWITZERLAND) 2024; 24:6923. [PMID: 39517820 PMCID: PMC11548305 DOI: 10.3390/s24216923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channel proteins belong to the superfamily of TRP proteins that form cationic channels in the animal cell membranes. These proteins have various subtype-specific functions, serving, for example, as sensors for pain, pressure, pH, and mechanical extracellular stimuli. The sensing of extracellular cues by TRPV4 triggers Ca2+-influx through the channel, subsequently coordinating numerous intracellular signaling cascades in a spatio-temporal manner. As TRPV channels play such a wide role in various cellular and physiological functions, loss or impaired TRPV protein activity naturally contributes to many pathophysiological processes. This review concentrates on the known functions of TRPV4 sensor proteins and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Sanna Koskimäki
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | | |
Collapse
|
4
|
Hernández-Vega AM, Llorente I, Sánchez-Hernández R, Segura Y, Tusié-Luna T, Morales-Buenrostro LE, García-Villegas R, Islas LD, Rosenbaum T. Identification and Properties of TRPV4 Mutant Channels Present in Polycystic Kidney Disease Patients. FUNCTION 2024; 5:zqae031. [PMID: 38984987 PMCID: PMC11384909 DOI: 10.1093/function/zqae031] [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: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Polycystic kidney disease (PKD), a disease characterized by the enlargement of the kidney through cystic growth is the fourth leading cause of end-stage kidney disease world-wide. Transient receptor potential Vanilloid 4 (TRPV4), a calcium-permeable TRP, channel participates in kidney cell physiology and since TRPV4 forms complexes with another channel whose malfunction is associated to PKD, TRPP2 (or PKD2), we sought to determine whether patients with PKD, exhibit previously unknown mutations in TRPV4. Here, we report the presence of mutations in the TRPV4 gene in patients diagnosed with PKD and determine that they produce gain-of-function (GOF). Mutations in the sequence of the TRPV4 gene have been associated to a broad spectrum of neuropathies and skeletal dysplasias but not PKD, and their biophysical effects on channel function have not been elucidated. We identified and examined the functional behavior of a novel E6K mutant and of the previously known S94L and A217S mutant TRVP4 channels. The A217S mutation has been associated to mixed neuropathy and/or skeletal dysplasia phenotypes, however, the PKD carriers of these variants had not been diagnosed with these reported clinical manifestations. The presence of certain mutations in TRPV4 may influence the progression and severity of PKD through GOF mechanisms. PKD patients carrying TRVP4 mutations are putatively more likely to require dialysis or renal transplant as compared to those without these mutations.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yayoi Segura
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis E Morales-Buenrostro
- Departmento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México 07360, Mexico
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina. Universidad Nacional Autónoma de México,Ciudad de México 04510, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
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
|
Yan Q, Gao C, Li M, Lan R, Wei S, Fan R, Cheng W. TRP Ion Channels in Immune Cells and Their Implications for Inflammation. Int J Mol Sci 2024; 25:2719. [PMID: 38473965 DOI: 10.3390/ijms25052719] [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: 01/24/2024] [Revised: 02/16/2024] [Accepted: 02/24/2024] [Indexed: 03/14/2024] Open
Abstract
The transient receptor potential (TRP) ion channels act as cellular sensors and mediate a plethora of physiological processes, including somatosensation, proliferation, apoptosis, and metabolism. Under specific conditions, certain TRP channels are involved in inflammation and immune responses. Thus, focusing on the role of TRPs in immune system cells may contribute to resolving inflammation. In this review, we discuss the distribution of five subfamilies of mammalian TRP ion channels in immune system cells and how these ion channels function in inflammatory mechanisms. This review provides an overview of the current understanding of TRP ion channels in mediating inflammation and may offer potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Qiyue Yan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Chuanzhou Gao
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Mei Li
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Rui Lan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Shaohan Wei
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Runsong Fan
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| | - Wei Cheng
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
7
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
8
|
HU WEI, WARTMANN THOMAS, STRECKER MARCO, PERRAKIS ARISTOTELIS, CRONER ROLAND, SZALLASI ARPAD, SHI WENJIE, KAHLERT ULFD. Transient receptor potential channels as predictive marker and potential indicator of chemoresistance in colon cancer. Oncol Res 2023; 32:227-239. [PMID: 38188686 PMCID: PMC10767253 DOI: 10.32604/or.2023.043053] [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: 06/20/2023] [Accepted: 10/13/2023] [Indexed: 01/09/2024] Open
Abstract
Transient receptor potential (TRP) channels are strongly associated with colon cancer development and progression. This study leveraged a multivariate Cox regression model on publicly available datasets to construct a TRP channels-associated gene signature, with further validation of signature in real world samples from our hospital treated patient samples. Kaplan-Meier (K-M) survival analysis and receiver operating characteristic (ROC) curves were employed to evaluate this gene signature's predictive accuracy and robustness in both training and testing cohorts, respectively. Additionally, the study utilized the CIBERSORT algorithm and single-sample gene set enrichment analysis to explore the signature's immune infiltration landscape and underlying functional implications. The support vector machine algorithm was applied to evaluate the signature's potential in predicting chemotherapy outcomes. The findings unveiled a novel three TRP channels-related gene signature (MCOLN1, TRPM5, and TRPV4) in colon adenocarcinoma (COAD). The ROC and K-M survival curves in the training dataset (AUC = 0.761; p = 1.58e-05) and testing dataset (AUC = 0.699; p = 0.004) showed the signature's robust predictive capability for the overall survival of COAD patients. Analysis of the immune infiltration landscape associated with the signature revealed higher immune infiltration, especially an increased presence of M2 macrophages, in high-risk group patients compared to their low-risk counterparts. High-risk score patients also exhibited potential responsiveness to immune checkpoint inhibitor therapy, evident through increased CD86 and PD-1 expression profiles. Moreover, the TRPM5 gene within the signature was highly expressed in the chemoresistance group (p = 0.00095) and associated with poor prognosis (p = 0.036) in COAD patients, highlighting its role as a hub gene of chemoresistance. Ultimately, this signature emerged as an independent prognosis factor for COAD patients (p = 6.48e-06) and expression of model gene are validated by public data and real-world patients. Overall, this bioinformatics study provides valuable insights into the prognostic implications and potential chemotherapy resistance mechanisms associated with TRPs-related genes in colon cancer.
Collapse
Affiliation(s)
- WEI HU
- The Fourth Clinical Medical College of Yangzhou University, Nantong Rich Hospital, Nantong, China
| | - THOMAS WARTMANN
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - MARCO STRECKER
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ARISTOTELIS PERRAKIS
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ROLAND CRONER
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ARPAD SZALLASI
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - WENJIE SHI
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - ULF D. KAHLERT
- Molecular and Experimental Surgery, Clinic for General-, Visceral-, Vascular and Transplant Surgery, Faculty of Medicine and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
9
|
Yang K, Liu S, Yan H, Lu W, Shan X, Chen H, Bao C, Feng H, Liao J, Liang S, Xu L, Tang H, Yuan JXJ, Zhong N, Wang J. SARS-CoV-2 spike protein receptor-binding domain perturbates intracellular calcium homeostasis and impairs pulmonary vascular endothelial cells. Signal Transduct Target Ther 2023; 8:276. [PMID: 37452066 PMCID: PMC10349149 DOI: 10.1038/s41392-023-01556-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/09/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023] Open
Abstract
Exposure to the spike protein or receptor-binding domain (S-RBD) of SARS-CoV-2 significantly influences endothelial cells and induces pulmonary vascular endotheliopathy. In this study, angiotensin-converting enzyme 2 humanized inbred (hACE2 Tg) mice and cultured pulmonary vascular endothelial cells were used to investigate how spike protein/S-RBD impacts pulmonary vascular endothelium. Results show that S-RBD leads to acute-to-prolonged induction of the intracellular free calcium concentration ([Ca2+]i) via acute activation of TRPV4, and prolonged upregulation of mechanosensitive channel Piezo1 and store-operated calcium channel (SOCC) key component Orai1 in cultured human pulmonary arterial endothelial cells (PAECs). In mechanism, S-RBD interacts with ACE2 to induce formation of clusters involving Orai1, Piezo1 and TRPC1, facilitate the channel activation of Piezo1 and SOCC, and lead to elevated apoptosis. These effects are blocked by Kobophenol A, which inhibits the binding between S-RBD and ACE2, or intracellular calcium chelator, BAPTA-AM. Blockade of Piezo1 and SOCC by GsMTx4 effectively protects the S-RBD-induced pulmonary microvascular endothelial damage in hACE2 Tg mice via normalizing the elevated [Ca2+]i. Comparing to prototypic strain, Omicron variants (BA.5.2 and XBB) of S-RBD induces significantly less severe cell apoptosis. Transcriptomic analysis indicates that prototypic S-RBD confers more severe acute impacts than Delta or Lambda S-RBD. In summary, this study provides compelling evidence that S-RBD could induce persistent pulmonary vascular endothelial damage by binding to ACE2 and triggering [Ca2+]i through upregulation of Piezo1 and Orai1. Targeted inhibition of ACE2-Piezo1/SOCC-[Ca2+]i axis proves a powerful strategy to treat S-RBD-induced pulmonary vascular diseases.
Collapse
Affiliation(s)
- Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Han Yan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqian Shan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haixia Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Pathology, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Changlei Bao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huazhuo Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Liao
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
10
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|
11
|
Luo Z, Zhan Z, Qin X, Pan W, Liang M, Li C, Weng S, He J, Guo C. Interaction of Teleost Fish TRPV4 with DEAD Box RNA Helicase 1 Regulates Iridovirus Replication. J Virol 2023; 97:e0049523. [PMID: 37289063 PMCID: PMC10308943 DOI: 10.1128/jvi.00495-23] [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: 04/02/2023] [Accepted: 05/18/2023] [Indexed: 06/09/2023] Open
Abstract
Viral diseases are a significant risk to the aquaculture industry. Transient receptor potential vanilloid 4 (TRPV4) has been reported to be involved in regulating viral activity in mammals, but its regulatory effect on viruses in teleost fish remains unknown. Here, the role of the TRPV4-DEAD box RNA helicase 1 (DDX1) axis in viral infection was investigated in mandarin fish (Siniperca chuatsi). Our results showed that TRPV4 activation mediates Ca2+ influx and facilitates infectious spleen and kidney necrosis virus (ISKNV) replication, whereas this promotion was nearly eliminated by an M709D mutation in TRPV4, a channel Ca2+ permeability mutant. The concentration of cellular Ca2+ increased during ISKNV infection, and Ca2+ was critical for viral replication. TRPV4 interacted with DDX1, and the interaction was mediated primarily by the N-terminal domain (NTD) of TRPV4 and the C-terminal domain (CTD) of DDX1. This interaction was attenuated by TRPV4 activation, thereby enhancing ISKNV replication. DDX1 could bind to viral mRNAs and facilitate ISKNV replication, which required the ATPase/helicase activity of DDX1. Furthermore, the TRPV4-DDX1 axis was verified to regulate herpes simplex virus 1 replication in mammalian cells. These results suggested that the TRPV4-DDX1 axis plays an important role in viral replication. Our work provides a novel molecular mechanism for host involvement in viral regulation, which would be of benefit for new insights into the prevention and control of aquaculture diseases. IMPORTANCE In 2020, global aquaculture production reached a record of 122.6 million tons, with a total value of $281.5 billion. Meanwhile, frequent outbreaks of viral diseases have occurred in aquaculture, and about 10% of farmed aquatic animal production has been lost to infectious diseases, resulting in more than $10 billion in economic losses every year. Therefore, an understanding of the potential molecular mechanism of how aquatic organisms respond to and regulate viral replication is of great significance. Our study suggested that TRPV4 enables Ca2+ influx and interactions with DDX1 to collectively promote ISKNV replication, providing novel insights into the roles of the TRPV4-DDX1 axis in regulating the proviral effect of DDX1. This advances our understanding of viral disease outbreaks and would be of benefit for studies on preventing aquatic viral diseases.
Collapse
Affiliation(s)
- Zhiyong Luo
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Zhipeng Zhan
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xiaowei Qin
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Weiqiang Pan
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Mincong Liang
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chuanrui Li
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Shaoping Weng
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Jianguo He
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Changjun Guo
- State Key Laboratory for Biocontrol, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- Guangdong Province Key Laboratory for Aquatic Economic Animals, School of Marine Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
12
|
Zhu L, Xie C, Chen L, Dai X, Zhou Y, Pan H, Tian K. Transport of microplastics in the body and interaction with biological barriers, and controlling of microplastics pollution. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 255:114818. [PMID: 36958263 DOI: 10.1016/j.ecoenv.2023.114818] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 03/05/2023] [Accepted: 03/19/2023] [Indexed: 06/18/2023]
Abstract
Microplastics (MPs) are one novel environmental pollutant sized < 5 mm that is ubiquitously present in numerous environmental media and particularly susceptible to interact with various toxic chemicals. Importantly, MPs can enter the food chain, and are bio-enriched and bio-accumulated with trophic levels, eventually endangering ecosystems and human health. However, there need to be more understanding regarding the bio-interaction of MPs with the host, particularly for biological barriers. This review aimed to summarize the latest findings regarding the main exposure routes of MPs that generated health burdens on humans. Furthermore, their interactions with biological barriers that generate adverse health effects and the underlying mechanisms were also reviewed. Additionally, we provided a comprehensive overview of recent advances regarding the removing and controlling of MPs. Finally, we discussed the future directions for MPs hazard prevention to provide helpful information for regulating decision-making and guiding safer plastics applications.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Occupational and Environmental Health, Zunyi Medical University, Zunyi 563000, China
| | - Caiyan Xie
- Department of Occupational and Environmental Health, Zunyi Medical University, Zunyi 563000, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xingyu Dai
- Department of Clinical Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yuanzhong Zhou
- Department of Occupational and Environmental Health, Zunyi Medical University, Zunyi 563000, China
| | - Hong Pan
- Department of Occupational and Environmental Health, Zunyi Medical University, Zunyi 563000, China.
| | - Kunming Tian
- Department of Occupational and Environmental Health, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
13
|
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
|
14
|
Wu H, Hou J, Wang X. A review of microplastic pollution in aquaculture: Sources, effects, removal strategies and prospects. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 252:114567. [PMID: 36706522 DOI: 10.1016/j.ecoenv.2023.114567] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/07/2023] [Accepted: 01/18/2023] [Indexed: 06/18/2023]
Abstract
As microplastic pollution has become an emerging environmental issue of global concern, microplastics in aquaculture have become a research hotspot. For environmental safety, economic efficiency and food safety considerations, a comprehensive understanding of microplastic pollution in aquaculture is necessary. This review outlines an overview of sources and effects of microplastics in aquaculture. External environmental inputs and aquaculture processes are sources of microplastics in aquaculture. Microplastics may release harmful additives and adsorb pollutants in aquaculture environment, cause deterioration of aquaculture environment, as well as cause toxicological effects, affect the behavior, growth and reproduction of aquaculture products, ultimately reducing the economic benefits of aquaculture. Microplastics entering the human body through aquaculture products also pose potential health risks at multiple levels. Microplastic pollution removal strategies used in aquaculture in various countries are also reviewed. Ecological interception and purification are considered to be effective methods. In addition, strengthening aquaculture management and improving fishing gear and packaging are also currently feasible solutions. As proactive measures, new portable microplastic monitoring system and remote sensing technology are considered to have broad application prospects. And it was encouraged to comprehensively strengthen the supervision of microplastic pollution in aquaculture through talent exchange and strengthening the construction of laws and regulations.
Collapse
Affiliation(s)
- Haodi Wu
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| | - Jing Hou
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China.
| | - Xiangke Wang
- MOE Key Laboratory of Resources and Environmental Systems Optimization, College of Environmental Science and Engineering, North China Electric Power University, Beijing 102206, China
| |
Collapse
|
15
|
Ziani K, Ioniță-Mîndrican CB, Mititelu M, Neacșu SM, Negrei C, Moroșan E, Drăgănescu D, Preda OT. Microplastics: A Real Global Threat for Environment and Food Safety: A State of the Art Review. Nutrients 2023; 15:617. [PMID: 36771324 PMCID: PMC9920460 DOI: 10.3390/nu15030617] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Microplastics are small plastic particles that come from the degradation of plastics, ubiquitous in nature and therefore affect both wildlife and humans. They have been detected in many marine species, but also in drinking water and in numerous foods, such as salt, honey and marine organisms. Exposure to microplastics can also occur through inhaled air. Data from animal studies have shown that once absorbed, plastic micro- and nanoparticles can distribute to the liver, spleen, heart, lungs, thymus, reproductive organs, kidneys and even the brain (crosses the blood-brain barrier). In addition, microplastics are transport operators of persistent organic pollutants or heavy metals from invertebrate organisms to other higher trophic levels. After ingestion, the additives and monomers in their composition can interfere with important biological processes in the human body and can cause disruption of the endocrine, immune system; can have a negative impact on mobility, reproduction and development; and can cause carcinogenesis. The pandemic caused by COVID-19 has affected not only human health and national economies but also the environment, due to the large volume of waste in the form of discarded personal protective equipment. The remarkable increase in global use of face masks, which mainly contain polypropylene, and poor waste management have led to worsening microplastic pollution, and the long-term consequences can be extremely devastating if urgent action is not taken.
Collapse
Affiliation(s)
- Khaled Ziani
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Corina-Bianca Ioniță-Mîndrican
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020945 Bucharest, Romania
| | - Magdalena Mititelu
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | | | - Carolina Negrei
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020945 Bucharest, Romania
| | - Elena Moroșan
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Doina Drăgănescu
- Department of Pharmaceutical Physics and Informatics, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Olivia-Teodora Preda
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, 020945 Bucharest, Romania
| |
Collapse
|
16
|
Erfinanda L, Zou L, Gutbier B, Kneller L, Weidenfeld S, Michalick L, Lei D, Reppe K, Teixeira Alves LG, Schneider B, Zhang Q, Li C, Fatykhova D, Schneider P, Liedtke W, Sohara E, Mitchell TJ, Gruber AD, Hocke A, Hippenstiel S, Suttorp N, Olschewski A, Mall MA, Witzenrath M, Kuebler WM. Loss of endothelial CFTR drives barrier failure and edema formation in lung infection and can be targeted by CFTR potentiation. Sci Transl Med 2022; 14:eabg8577. [PMID: 36475904 DOI: 10.1126/scitranslmed.abg8577] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pneumonia is the most common cause of the acute respiratory distress syndrome (ARDS). Here, we identified loss of endothelial cystic fibrosis transmembrane conductance regulator (CFTR) as an important pathomechanism leading to lung barrier failure in pneumonia-induced ARDS. CFTR was down-regulated after Streptococcus pneumoniae infection ex vivo or in vivo in human or murine lung tissue, respectively. Analysis of isolated perfused rat lungs revealed that CFTR inhibition increased endothelial permeability in parallel with intracellular chloride ion and calcium ion concentrations ([Cl-]i and [Ca2+]i). Inhibition of the chloride ion-sensitive with-no-lysine kinase 1 (WNK1) protein with tyrphostin 47 or WNK463 replicated the effect of CFTR inhibition on endothelial permeability and endothelial [Ca2+]i, whereas WNK1 activation by temozolomide attenuated it. Endothelial [Ca2+]i transients and permeability in response to inhibition of either CFTR or WNK1 were prevented by inhibition of the cation channel transient receptor potential vanilloid 4 (TRPV4). Mice deficient in Trpv4 (Trpv4-/-) developed less lung edema and protein leak than their wild-type littermates after infection with S. pneumoniae. The CFTR potentiator ivacaftor prevented lung CFTR loss, edema, and protein leak after S. pneumoniae infection in wild-type mice. In conclusion, lung infection caused loss of CFTR that promoted lung edema formation through intracellular chloride ion accumulation, inhibition of WNK1, and subsequent disinhibition of TRPV4, resulting in endothelial calcium ion influx and vascular barrier failure. Ivacaftor prevented CFTR loss in the lungs of mice with pneumonia and may, therefore, represent a possible therapeutic strategy in people suffering from ARDS due to severe pneumonia.
Collapse
Affiliation(s)
- Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Lin Zou
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Heart Center, 13353 Berlin, Germany.,Department of Endocrinology, Shanghai Pudong New Area Gongli Hospital, 200135 Shanghai, China
| | - Birgitt Gutbier
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Laura Kneller
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Sarah Weidenfeld
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Laura Michalick
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Disi Lei
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Heart Center, 13353 Berlin, Germany
| | - Katrin Reppe
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Luiz Gustavo Teixeira Alves
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Bill Schneider
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Qi Zhang
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Caihong Li
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Diana Fatykhova
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Paul Schneider
- Department for General and Thoracic Surgery, DRK Clinics, 13359 Berlin, Germany
| | - Wolfgang Liedtke
- Departments of Neurology, Neurobiology, and Clinics for Pain and Palliative Care, Duke University Medical Center, Durham, NC 27710, USA
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Timothy J Mitchell
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B15-2TT, UK
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, 14163 Berlin, Germany
| | - Andreas Hocke
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany
| | - Andrea Olschewski
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Marcus A Mall
- German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany.,Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Center for Lung Research (DZL), associated partner site, 10117 Berlin, Germany
| |
Collapse
|
17
|
Goretzki B, Tebbe F, Mitrovic SA, Hellmich UA. Backbone NMR assignments of the extensive human and chicken TRPV4 N-terminal intrinsically disordered regions as important players in ion channel regulation. BIOMOLECULAR NMR ASSIGNMENTS 2022; 16:205-212. [PMID: 35451798 PMCID: PMC9027025 DOI: 10.1007/s12104-022-10080-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
Transient receptor potential (TRP) channels are important pharmacological targets due to their ability to act as sensory transducers on the organismic and cellular level, as polymodal signal integrators and because of their role in numerous diseases. However, a detailed molecular understanding of the structural dynamics of TRP channels and their integration into larger cellular signalling networks remains challenging, in part due to the systematic absence of highly dynamic regions pivotal for channel regulation from available structures. In human TRP vanilloid 4 (TRPV4), a ubiquitously expressed homotetrameric cation channel involved in temperature, osmo- and mechano-sensation and in a multitude of (patho)physiological processes, the intrinsically disordered N-terminus encompasses 150 amino acids and thus represents > 17% of the entire channel sequence. Its deletion renders the channel significantly less excitable to agonists supporting a crucial role in TRPV4 activation and regulation. For a structural understanding and a comparison of its properties across species, we determined the NMR backbone assignments of the human and chicken TRPV4 N-terminal IDRs.
Collapse
Affiliation(s)
- Benedikt Goretzki
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry and Cluster of Excellence "Balance of the Microverse", Friedrich Schiller University Jena, Humboldtstrasse 10, 07443, Jena, Germany
- Center for Biomolecular Magnetic Resonance, Goethe-University, Max-von-Laue-Strasse 9, 60438, Frankfurt, Germany
| | - Frederike Tebbe
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry and Cluster of Excellence "Balance of the Microverse", Friedrich Schiller University Jena, Humboldtstrasse 10, 07443, Jena, Germany
| | - Sarah-Ana Mitrovic
- Department of Chemistry, Division Biochemistry, Johannes-Gutenberg-University Mainz, Johann-Joachim Becher-Weg 30, 55128, Mainz, Germany
| | - Ute A Hellmich
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry and Cluster of Excellence "Balance of the Microverse", Friedrich Schiller University Jena, Humboldtstrasse 10, 07443, Jena, Germany.
- Center for Biomolecular Magnetic Resonance, Goethe-University, Max-von-Laue-Strasse 9, 60438, Frankfurt, Germany.
| |
Collapse
|
18
|
Müller I, Alt P, Rajan S, Schaller L, Geiger F, Dietrich A. Transient Receptor Potential (TRP) Channels in Airway Toxicity and Disease: An Update. Cells 2022; 11:2907. [PMID: 36139480 PMCID: PMC9497104 DOI: 10.3390/cells11182907] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Our respiratory system is exposed to toxicants and pathogens from both sides: the airways and the vasculature. While tracheal, bronchial and alveolar epithelial cells form a natural barrier in the airways, endothelial cells protect the lung from perfused toxic compounds, particulate matter and invading microorganism in the vascular system. Damages induce inflammation by our immune response and wound healing by (myo)fibroblast proliferation. Members of the transient receptor potential (TRP) superfamily of ion channel are expressed in many cells of the respiratory tract and serve multiple functions in physiology and pathophysiology. TRP expression patterns in non-neuronal cells with a focus on TRPA1, TRPC6, TRPM2, TRPM5, TRPM7, TRPV2, TRPV4 and TRPV6 channels are presented, and their roles in barrier function, immune regulation and phagocytosis are summarized. Moreover, TRP channels as future pharmacological targets in chronic obstructive pulmonary disease (COPD), asthma, cystic and pulmonary fibrosis as well as lung edema are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Alexander Dietrich
- Walther-Straub-Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), LMU-Munich, Nussbaumstr. 26, 80336 Munich, Germany
| |
Collapse
|
19
|
Voelkel NF, Bogaard HJ, Kuebler WM. ARDS in the Time of Corona: Context and Perspective. Am J Physiol Lung Cell Mol Physiol 2022; 323:L431-L437. [PMID: 35997290 PMCID: PMC9529269 DOI: 10.1152/ajplung.00432.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
For more than 2 years, COVID-19 has been holding the world at awe with new waves of infections, novel mutants, and still limited (albeit improved) means to combat SARS-CoV-2-induced respiratory failure, the most common and fatal presentation of severe COVID-19. In the present perspective, we draw from the successes and—mostly—failures in previous acute respiratory distress syndrome (ARDS) work and the experiences from COVID-19 to define conceptual barriers that have so far hindered therapeutic breakthroughs in this deadly disease, and to open up new avenues of thinking and thus, ultimately of therapy.
Collapse
Affiliation(s)
- Norbert F Voelkel
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Harm Jan Bogaard
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| |
Collapse
|
20
|
Zhang Y, Liang P, Yang L, Shan KZ, Feng L, Chen Y, Liedtke W, Coyne CB, Yang H. Functional coupling between TRPV4 channel and TMEM16F modulates human trophoblast fusion. eLife 2022; 11:e78840. [PMID: 35670667 PMCID: PMC9236608 DOI: 10.7554/elife.78840] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/05/2022] [Indexed: 11/15/2022] Open
Abstract
TMEM16F, a Ca2+-activated phospholipid scramblase (CaPLSase), is critical for placental trophoblast syncytialization, HIV infection, and SARS-CoV2-mediated syncytialization, however, how TMEM16F is activated during cell fusion is unclear. Here, using trophoblasts as a model for cell fusion, we demonstrate that Ca2+ influx through the Ca2+ permeable transient receptor potential vanilloid channel TRPV4 is critical for TMEM16F activation and plays a role in subsequent human trophoblast fusion. GSK1016790A, a TRPV4 specific agonist, robustly activates TMEM16F in trophoblasts. We also show that TRPV4 and TMEM16F are functionally coupled within Ca2+ microdomains in a human trophoblast cell line using patch-clamp electrophysiology. Pharmacological inhibition or gene silencing of TRPV4 hinders TMEM16F activation and subsequent trophoblast syncytialization. Our study uncovers the functional expression of TRPV4 and one of the physiological activation mechanisms of TMEM16F in human trophoblasts, thus providing us with novel strategies to regulate CaPLSase activity as a critical checkpoint of physiologically and disease-relevant cell fusion events.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liheng Yang
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
| | - Ke Zoe Shan
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical CentreDurhamUnited States
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua HospitalShanghaiChina
| | - Yong Chen
- Department of Neurology, Duke University Medical CenterDurhamUnited States
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical CenterDurhamUnited States
- Department of Anesthesiology, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
- College of Dentistry, Department of Molecular Pathobiology, NYUNew YorkUnited States
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
21
|
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
|
22
|
Nadeali Z, Mohammad-Rezaei F, Aria H, Nikpour P. Possible role of pannexin 1 channels and purinergic receptors in the pathogenesis and mechanism of action of SARS-CoV-2 and therapeutic potential of targeting them in COVID-19. Life Sci 2022; 297:120482. [PMID: 35288174 PMCID: PMC8915746 DOI: 10.1016/j.lfs.2022.120482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 01/08/2023]
Abstract
Identifying signaling pathways and molecules involved in SARS-CoV-2 pathogenesis is pivotal for developing new effective therapeutic or preventive strategies for COVID-19. Pannexins (PANX) are ATP-release channels in the plasma membrane essential in many physiological and immune responses. Activation of pannexin channels and downstream purinergic receptors play dual roles in viral infection, either by facilitating viral replication and infection or inducing host antiviral defense. The current review provides a hypothesis demonstrating the possible contribution of the PANX1 channel and purinergic receptors in SARS-CoV-2 pathogenesis and mechanism of action. Moreover, we discuss whether targeting these signaling pathways may provide promising preventative therapies and treatments for patients with progressive COVID-19 resulting from excessive pro-inflammatory cytokines and chemokines production. Several inhibitors of this pathway have been developed for the treatment of other viral infections and pathological consequences. Specific PANX1 inhibitors could be potentially included as part of the COVID-19 treatment regimen if, in future, studies demonstrate the role of PANX1 in COVID-19 pathogenesis. Of note, any ATP therapeutic modulation for COVID-19 should be carefully designed and monitored because of the complex role of extracellular ATP in cellular physiology.
Collapse
Affiliation(s)
- Zakiye Nadeali
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Mohammad-Rezaei
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Aria
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
23
|
Boudaka A, Tominaga M. Physiological and Pathological Significance of Esophageal TRP Channels: Special Focus on TRPV4 in Esophageal Epithelial Cells. Int J Mol Sci 2022; 23:ijms23094550. [PMID: 35562940 PMCID: PMC9099744 DOI: 10.3390/ijms23094550] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 12/10/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel that is broadly expressed in different human tissues, including the digestive system, where it acts as a molecular sensor and a transducer that regulates a variety of functional activities. Despite the extensive research to determine the role of this channel in the physiology and pathophysiology of different organs, the unique morphological and functional features of TRPV4 in the esophagus remain largely unknown. Ten years ago, TRPV4 was shown to be highly expressed in esophageal epithelial cells where its activation induces Ca2+-dependent ATP release, which, in turn, mediates several functions, ranging from mechanosensation to wound healing. This review summarizes the research progress on TRPV4, and focuses on the functional expression of TRPV4 in esophageal epithelium and its possible role in different esophageal diseases that would support TRPV4 as a candidate target for future therapeutic approaches to treat patients with these conditions.
Collapse
Affiliation(s)
- Ammar Boudaka
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Al-Khoud, P.O. Box 35, Muscat 123, Oman
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki 444-8787, Aichi, Japan;
- Correspondence:
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki 444-8787, Aichi, Japan;
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Aichi, Japan
- Exploratory Research Center on Life and Living Systems, Thermal Biology Group, Okazaki 444-8787, Aichi, Japan
| |
Collapse
|
24
|
Armenta-Medina D, Brambila-Tapia AJL, Miranda-Jiménez S, Rodea-Montero ER. A Web Application for Biomedical Text Mining of Scientific Literature Associated with Coronavirus-Related Syndromes: Coronavirus Finder. Diagnostics (Basel) 2022; 12:887. [PMID: 35453935 PMCID: PMC9028729 DOI: 10.3390/diagnostics12040887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/10/2022] Open
Abstract
In this study, a web application was developed that comprises scientific literature associated with the Coronaviridae family, specifically for those viruses that are members of the Genus Betacoronavirus, responsible for emerging diseases with a great impact on human health: Middle East Respiratory Syndrome-Related Coronavirus (MERS-CoV) and Severe Acute Respiratory Syndrome-Related Coronavirus (SARS-CoV, SARS-CoV-2). The information compiled on this webserver aims to understand the basics of these viruses' infection, and the nature of their pathogenesis, enabling the identification of molecular and cellular components that may function as potential targets on the design and development of successful treatments for the diseases associated with the Coronaviridae family. Some of the web application's primary functions are searching for keywords within the scientific literature, natural language processing for the extraction of genes and words, the generation and visualization of gene networks associated with viral diseases derived from the analysis of latent semantic space, and cosine similarity measures. Interestingly, our gene association analysis reveals drug targets in understudies, and new targets suggested in the scientific literature to treat coronavirus.
Collapse
Affiliation(s)
- Dagoberto Armenta-Medina
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México 03940, Mexico;
- Centro de Investigación e Innovación en Tecnologías de la Información y Comunicación (INFOTEC), Aguascalientes 20326, Mexico
| | | | - Sabino Miranda-Jiménez
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México 03940, Mexico;
- Centro de Investigación e Innovación en Tecnologías de la Información y Comunicación (INFOTEC), Aguascalientes 20326, Mexico
| | | |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Shahgolzari M, Yavari A, Arjeini Y, Miri SM, Darabi A, Mozaffari Nejad AS, Keshavarz M. Immunopathology and Immunopathogenesis of COVID-19, what we know and what we should learn. GENE REPORTS 2021; 25:101417. [PMID: 34778602 PMCID: PMC8570409 DOI: 10.1016/j.genrep.2021.101417] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 02/08/2023]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) directly interacts with host's epithelial and immune cells, leading to inflammatory response induction, which is considered the hallmark of infection. The host immune system is programmed to facilitate the clearance of viral infection by establishing a modulated response. However, SARS-CoV-2 takes the initiative and its various structural and non-structural proteins directly or indirectly stimulate the uncontrolled activation of injurious inflammatory pathways through interaction with innate immune system mediators. Upregulation of cell-signaling pathways such as mitogen-activate protein kinase (MAPK) in response to recognition of SARS-CoV-2 antigens by innate immune system receptors mediates unbridled production of proinflammatory cytokines and cells causing cytokine storm, tissue damage, increased pulmonary edema, acute respiratory distress syndrome (ARDS), and mortality. Moreover, this acute inflammatory state hinders the immunomodulatory effect of T helper cells and timely response of CD4+ and CD8+ T cells against infection. Furthermore, inflammation-induced overproduction of Th17 cells can downregulate the antiviral response of Th1 and Th2 cells. In fact, the improperly severe response of the innate immune system is the key to conversion from a non-severe to severe disease state and needs to be investigated more deeply. The virus can also modulate the protective immune responses by developing immune evasion mechanisms, and thereby provide a more stable niche. Overall, combination of detrimental immunostimulatory and immunomodulatory properties of both the SARS-CoV-2 and immune cells does complicate the immune interplay. Thorough understanding of immunopathogenic basis of immune responses against SARS-CoV-2 has led to developing several advanced vaccines and immune-based therapeutics and should be expanded more rapidly. In this review, we tried to delineate the immunopathogenesis of SARS-CoV-2 in humans and to provide insight into more effective therapeutic and prophylactic strategies.
Collapse
Affiliation(s)
- Mehdi Shahgolzari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afagh Yavari
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Yaser Arjeini
- Department of Research and Development, Production and Research Complex, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Mohammad Miri
- Freelance Researcher of Biomedical Sciences, No 32, Vaezi Street, Tehran, Iran
| | - Amirhossein Darabi
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Amir Sasan Mozaffari Nejad
- Department of Microbiology, Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Keshavarz
- The Persian Gulf Tropical Medicine Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
28
|
Peng S, Poole DP, Veldhuis NA. Mini-review: Dissecting receptor-mediated stimulation of TRPV4 in nociceptive and inflammatory pathways. Neurosci Lett 2021; 770:136377. [PMID: 34856355 DOI: 10.1016/j.neulet.2021.136377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a polymodal, non-selective cation channel that detects thermal, mechanical, and environmental cues and contributes to a range of diverse physiological processes. The effects of chronic TRPV4 stimulation and gain-of-function genetic mutations suggest that TRPV4 may also be a valuable therapeutic target for pathophysiological events including neurogenic inflammation, peripheral neuropathies, and impaired wound healing. There has been significant interest in defining how and where TRPV4 may promote inflammation and pain. Endogenous stimuli such as osmotic stress and lipid binding are established TRPV4 activators. The TRP channel family is also well-known to be controlled by 'receptor-operated' pathways. For example, G protein-coupled receptors (GPCRs) expressed by primary afferent neurons or other cells in inflammatory pathways utilize TRPV4 as an effector protein to amplify nociceptive and inflammatory signaling. Contributing to disorders including arthritis, neuropathies, and pulmonary edema, GPCRs such as the protease-activated receptor PAR2 mediate activation of kinase signaling cascades to increase TRPV4 phosphorylation, resulting in sensitization and enhanced neuronal excitability. Phospholipase activity also leads to production of polyunsaturated fatty acid lipid mediators that directly activate TRPV4. Consistent with the contribution of TRPV4 to disease, pharmacological inhibition or genetic ablation of TRPV4 can diminish receptor-mediated inflammatory events. This review outlines how receptor-mediated signaling is a major endogenous driver of TRPV4 gating and discusses key signaling pathways and emerging TRPV4 modulators such as the mechanosensitive Piezo1 ion channel. A collective understanding of how endogenous stimuli can influence TRPV4 function is critical for future therapeutic endeavors to modulate this channel.
Collapse
Affiliation(s)
- Scott Peng
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Daniel P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| | - Nicholas A Veldhuis
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
29
|
Pulmonary Edema in COVID-19 Treated with Furosemide and Negative Fluid Balance (NEGBAL): A Different and Promising Approach. J Clin Med 2021; 10:jcm10235599. [PMID: 34884300 PMCID: PMC8658626 DOI: 10.3390/jcm10235599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 12/27/2022] Open
Abstract
In COVID-19, pulmonary edema has been attributed to “cytokine storm”. However, it is known that SARS-CoV2 promotes angiotensin-converting enzyme 2 deficit, increases angiotensin II, and this triggers volume overload. Our report is based on COVID-19 patients with tomographic evidence of pulmonary edema and volume overload to whom established a standard treatment with diuretic (furosemide) guided by objectives: Negative Fluid Balance (NEGBAL approach). Retrospective observational study. We reviewed data from medical records: demographic, clinical, laboratory, blood gas, and chest tomography (CT) before and while undergoing NEGBAL, from 20 critically ill patients. Once the NEGBAL strategy was started, no patient required mechanical ventilation. All cases reverted to respiratory failure with NEGBAL, but subsequently two patients died from sepsis and acute myocardial infarction (AMI). The regressive analysis between PaO2/FiO2BAL and NEGBAL demonstrated correlation (p < 0.032). The results comparing the Pao2Fio2 between admission to NEGBAL to NEGBAL day 4, were statistically significant (p < 0.001). We noted between admission to NEGBAL and day 4 improvement in CT score (p < 0.001), decrease in the superior vena cava diameter (p < 0.001) and the decrease of cardiac axis (p < 0.001). Though our study has several limitations, we believe the promising results encourage further investigation of this different pathophysiological approach.
Collapse
|
30
|
Orsini EM, Perelas A, Southern BD, Grove LM, Olman MA, Scheraga RG. Stretching the Function of Innate Immune Cells. Front Immunol 2021; 12:767319. [PMID: 34795674 PMCID: PMC8593101 DOI: 10.3389/fimmu.2021.767319] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
The importance of innate immune cells to sense and respond to their physical environment is becoming increasingly recognized. Innate immune cells (e.g. macrophages and neutrophils) are able to receive mechanical signals through several mechanisms. In this review, we discuss the role of mechanosensitive ion channels, such as Piezo1 and transient receptor potential vanilloid 4 (TRPV4), and cell adhesion molecules, such as integrins, selectins, and cadherins in biology and human disease. Furthermore, we explain that these mechanical stimuli activate intracellular signaling pathways, such as MAPK (p38, JNK), YAP/TAZ, EDN1, NF-kB, and HIF-1α, to induce protein conformation changes and modulate gene expression to drive cellular function. Understanding the mechanisms by which immune cells interpret mechanosensitive information presents potential targets to treat human disease. Important areas of future study in this area include autoimmune, allergic, infectious, and malignant conditions.
Collapse
Affiliation(s)
- Erica M Orsini
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Apostolos Perelas
- Department of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, United States
| | - Brian D Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M Grove
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Rachel G Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
31
|
Advances in TRP channel drug discovery: from target validation to clinical studies. Nat Rev Drug Discov 2021; 21:41-59. [PMID: 34526696 PMCID: PMC8442523 DOI: 10.1038/s41573-021-00268-4] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/20/2022]
Abstract
Transient receptor potential (TRP) channels are multifunctional signalling molecules with many roles in sensory perception and cellular physiology. Therefore, it is not surprising that TRP channels have been implicated in numerous diseases, including hereditary disorders caused by defects in genes encoding TRP channels (TRP channelopathies). Most TRP channels are located at the cell surface, which makes them generally accessible drug targets. Early drug discovery efforts to target TRP channels focused on pain, but as our knowledge of TRP channels and their role in health and disease has grown, these efforts have expanded into new clinical indications, ranging from respiratory disorders through neurological and psychiatric diseases to diabetes and cancer. In this Review, we discuss recent findings in TRP channel structural biology that can affect both drug development and clinical indications. We also discuss the clinical promise of novel TRP channel modulators, aimed at both established and emerging targets. Last, we address the challenges that these compounds may face in clinical practice, including the need for carefully targeted approaches to minimize potential side-effects due to the multifunctional roles of TRP channels.
Collapse
|
32
|
Retamal JS, Grace MS, Dill LK, Ramirez-Garcia P, Peng S, Gondin AB, Bennetts F, Alvi S, Rajasekhar P, Almazi JG, Carbone SE, Bunnett NW, Davis TP, Veldhuis NA, Poole DP, McIntyre P. Serotonin-induced vascular permeability is mediated by transient receptor potential vanilloid 4 in the airways and upper gastrointestinal tract of mice. J Transl Med 2021; 101:851-864. [PMID: 33859334 PMCID: PMC8047529 DOI: 10.1038/s41374-021-00593-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 01/07/2023] Open
Abstract
Endothelial and epithelial cells form physical barriers that modulate the exchange of fluid and molecules. The integrity of these barriers can be influenced by signaling through G protein-coupled receptors (GPCRs) and ion channels. Serotonin (5-HT) is an important vasoactive mediator of tissue edema and inflammation. However, the mechanisms that drive 5-HT-induced plasma extravasation are poorly defined. The Transient Receptor Potential Vanilloid 4 (TRPV4) ion channel is an established enhancer of signaling by GPCRs that promote inflammation and endothelial barrier disruption. Here, we investigated the role of TRPV4 in 5-HT-induced plasma extravasation using pharmacological and genetic approaches. Activation of either TRPV4 or 5-HT receptors promoted significant plasma extravasation in the airway and upper gastrointestinal tract of mice. 5-HT-mediated extravasation was significantly reduced by pharmacological inhibition of the 5-HT2A receptor subtype, or with antagonism or deletion of TRPV4, consistent with functional interaction between 5-HT receptors and TRPV4. Inhibition of receptors for the neuropeptides substance P (SP) or calcitonin gene-related peptide (CGRP) diminished 5-HT-induced plasma extravasation. Supporting studies assessing treatment of HUVEC with 5-HT, CGRP, or SP was associated with ERK phosphorylation. Exposure to the TRPV4 activator GSK1016790A, but not 5-HT, increased intracellular Ca2+ in these cells. However, 5-HT pre-treatment enhanced GSK1016790A-mediated Ca2+ signaling, consistent with sensitization of TRPV4. The functional interaction was further characterized in HEK293 cells expressing 5-HT2A to reveal that TRPV4 enhances the duration of 5-HT-evoked Ca2+ signaling through a PLA2 and PKC-dependent mechanism. In summary, this study demonstrates that TRPV4 contributes to 5-HT2A-induced plasma extravasation in the airways and upper GI tract, with evidence supporting a mechanism of action involving SP and CGRP release.
Collapse
Affiliation(s)
- Jeffri S Retamal
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Megan S Grace
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- Department of Physiology, School of Medicine Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
- School of Clinical Medicine, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Larissa K Dill
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Paulina Ramirez-Garcia
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Scott Peng
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Arisbel B Gondin
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Felix Bennetts
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Sadia Alvi
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Pradeep Rajasekhar
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Juhura G Almazi
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
| | - Simona E Carbone
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Nigel W Bunnett
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, USA
| | - Thomas P Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia.
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia.
| | - Peter McIntyre
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| |
Collapse
|
33
|
Sun AM, Hoffman T, Luu BQ, Ashammakhi N, Li S. Application of lung microphysiological systems to COVID-19 modeling and drug discovery: a review. Biodes Manuf 2021; 4:757-775. [PMID: 34178414 PMCID: PMC8213042 DOI: 10.1007/s42242-021-00136-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/13/2021] [Indexed: 01/08/2023]
Abstract
There is a pressing need for effective therapeutics for coronavirus disease 2019 (COVID-19), the respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. The process of drug development is a costly and meticulously paced process, where progress is often hindered by the failure of initially promising leads. To aid this challenge, in vitro human microphysiological systems need to be refined and adapted for mechanistic studies and drug screening, thereby saving valuable time and resources during a pandemic crisis. The SARS-CoV-2 virus attacks the lung, an organ where the unique three-dimensional (3D) structure of its functional units is critical for proper respiratory function. The in vitro lung models essentially recapitulate the distinct tissue structure and the dynamic mechanical and biological interactions between different cell types. Current model systems include Transwell, organoid and organ-on-a-chip or microphysiological systems (MPSs). We review models that have direct relevance toward modeling the pathology of COVID-19, including the processes of inflammation, edema, coagulation, as well as lung immune function. We also consider the practical issues that may influence the design and fabrication of MPS. The role of lung MPS is addressed in the context of multi-organ models, and it is discussed how high-throughput screening and artificial intelligence can be integrated with lung MPS to accelerate drug development for COVID-19 and other infectious diseases.
Collapse
Affiliation(s)
- Argus M. Sun
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- UC San Diego Healthcare, UCSD, La Jolla, CA 92037 USA
| | - Tyler Hoffman
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
| | - Bao Q. Luu
- Pulmonary Diseases and Critical Care, Scripps Green Hospital, Scripps Health, La Jolla, CA 92037 USA
| | - Nureddin Ashammakhi
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI 48824 USA
| | - Song Li
- Department of Bioengineering, Samueli School of Engineering, University of California - Los Angeles, 420 Westwood Plaza 5121 Engineering V University of California, Los Angeles, CA 90095-1600 USA
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095 USA
| |
Collapse
|
34
|
de Bruin S, Bos LD, van Roon MA, Tuip-de Boer AM, Schuurman AR, Koel-Simmelinck MJA, Bogaard HJ, Tuinman PR, van Agtmael MA, Hamann J, Teunissen CE, Wiersinga WJ, Koos Zwinderman AH, Brouwer MC, van de Beek D, Vlaar APJ. Clinical features and prognostic factors in Covid-19: A prospective cohort study. EBioMedicine 2021; 67:103378. [PMID: 34000622 PMCID: PMC8118723 DOI: 10.1016/j.ebiom.2021.103378] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Mortality rates are high among hospitalized patients with COVID-19, especially in those intubated on the ICU. Insight in pathways associated with unfavourable outcome may lead to new treatment strategies. METHODS We performed a prospective cohort study of patients with COVID-19 admitted to general ward or ICU who underwent serial blood sampling. To provide insight in the pathways involved in disease progression, associations were estimated between outcome risk and serial measurements of 64 biomarkers in potential important pathways of COVID-19 infection (inflammation, tissue damage, complement system, coagulation and fibrinolysis) using joint models combining Cox regression and linear mixed-effects models. For patients admitted to the general ward, the primary outcome was admission to the ICU or mortality (unfavourable outcome). For patients admitted to the ICU, the primary outcome was 12-week mortality. FINDINGS A total of 219 patients were included: 136 (62%) on the ward and 119 patients (54%) on the ICU; 36 patients (26%) were included in both cohorts because they were transferred from general ward to ICU. On the general ward, 54 of 136 patients (40%) had an unfavourable outcome and 31 (23%) patients died. On the ICU, 54 out of 119 patients (45%) died. Unfavourable outcome on the general ward was associated with changes in concentrations of IL-6, IL-8, IL-10, soluble Receptor for Advanced Glycation End Products (sRAGE), vascular cell adhesion molecule 1 (VCAM-1) and Pentraxin-3. Death on the ICU was associated with changes in IL-6, IL-8, IL-10, sRAGE, VCAM-1, Pentraxin-3, urokinase-type plasminogen activator receptor, IL-1-receptor antagonist, CD14, procalcitonin, tumor necrosis factor alfa, tissue factor, complement component 5a, Growth arrest-specific 6, angiopoietin 2, and lactoferrin. Pathway analysis showed that unfavourable outcome on the ward was mainly driven by chemotaxis and interleukin production, whereas death on ICU was associated with a variety of pathways including chemotaxis, cell-cell adhesion, innate host response mechanisms, including the complement system, viral life cycle regulation, angiogenesis, wound healing and response to corticosteroids. INTERPRETATION Clinical deterioration in patients with severe COVID-19 involves multiple pathways, including chemotaxis and interleukin production, but also endothelial dysfunction, the complement system, and immunothrombosis. Prognostic markers showed considerable overlap between general ward and ICU patients, but we identified distinct differences between groups that should be considered in the development and timing of interventional therapies in COVID-19. FUNDING Amsterdam UMC, Amsterdam UMC Corona Fund, and Dr. C.J. Vaillant Fonds.
Collapse
Affiliation(s)
- Sanne de Bruin
- From the Department of Intensive Care, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lieuwe D Bos
- From the Department of Intensive Care, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marian A van Roon
- Department of Neurology, University of Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Anita M Tuip-de Boer
- From the Department of Intensive Care, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alex R Schuurman
- Department of Infectious Diseases, University of Amsterdam, Amsterdam UMC, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Marleen J A Koel-Simmelinck
- Neurochemistry Laboratory, Department of Clinical Chemistry, Free University, Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Pieter Roel Tuinman
- Department of Intensive Care, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Michiel A van Agtmael
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jörg Hamann
- Biobank Core Facility, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Free University, Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Department of Infectious Diseases, University of Amsterdam, Amsterdam UMC, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - A H Koos Zwinderman
- Department of Clinical Epidemiology and Biostatistics, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, the Netherlands
| | - Matthijs C Brouwer
- Department of Neurology, University of Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Diederik van de Beek
- Department of Neurology, University of Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Alexander P J Vlaar
- From the Department of Intensive Care, University of Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Daneva Z, Marziano C, Ottolini M, Chen YL, Baker TM, Kuppusamy M, Zhang A, Ta HQ, Reagan CE, Mihalek AD, Kasetti RB, Shen Y, Isakson BE, Minshall RD, Zode GS, Goncharova EA, Laubach VE, Sonkusare SK. Caveolar peroxynitrite formation impairs endothelial TRPV4 channels and elevates pulmonary arterial pressure in pulmonary hypertension. Proc Natl Acad Sci U S A 2021; 118:e2023130118. [PMID: 33879616 PMCID: PMC8092599 DOI: 10.1073/pnas.2023130118] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies have focused on the contribution of capillary endothelial TRPV4 channels to pulmonary pathologies, including lung edema and lung injury. However, in pulmonary hypertension (PH), small pulmonary arteries are the focus of the pathology, and endothelial TRPV4 channels in this crucial anatomy remain unexplored in PH. Here, we provide evidence that TRPV4 channels in endothelial cell caveolae maintain a low pulmonary arterial pressure under normal conditions. Moreover, the activity of caveolar TRPV4 channels is impaired in pulmonary arteries from mouse models of PH and PH patients. In PH, up-regulation of iNOS and NOX1 enzymes at endothelial cell caveolae results in the formation of the oxidant molecule peroxynitrite. Peroxynitrite, in turn, targets the structural protein caveolin-1 to reduce the activity of TRPV4 channels. These results suggest that endothelial caveolin-1-TRPV4 channel signaling lowers pulmonary arterial pressure, and impairment of endothelial caveolin-1-TRPV4 channel signaling contributes to elevated pulmonary arterial pressure in PH. Thus, inhibiting NOX1 or iNOS activity, or lowering endothelial peroxynitrite levels, may represent strategies for restoring vasodilation and pulmonary arterial pressure in PH.
Collapse
Affiliation(s)
- Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Corina Marziano
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Matteo Ottolini
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Thomas M Baker
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Aimee Zhang
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Huy Q Ta
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Claire E Reagan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908
| | - Andrew D Mihalek
- Department of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA 22908
| | - Ramesh B Kasetti
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Yuanjun Shen
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
| | - Gulab S Zode
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107
| | - Elena A Goncharova
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213
| | - Victor E Laubach
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908;
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
36
|
Role of pirfenidone in TGF-β pathways and other inflammatory pathways in acute respiratory syndrome coronavirus 2 (SARS-Cov-2) infection: a theoretical perspective. Pharmacol Rep 2021; 73:712-727. [PMID: 33880743 PMCID: PMC8057922 DOI: 10.1007/s43440-021-00255-x] [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: 01/18/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes pulmonary injury or multiple-organ injury by various pathological pathways. Transforming growth factor-beta (TGF-β) is a key factor that is released during SARS-CoV-2 infection. TGF-β, by internalization of the epithelial sodium channel (ENaC), suppresses the anti-oxidant system, downregulates the cystic fibrosis transmembrane conductance regulator (CFTR), and activates the plasminogen activator inhibitor 1 (PAI-1) and nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-kB). These changes cause inflammation and lung injury along with coagulopathy. Moreover, reactive oxygen species play a significant role in lung injury, which levels up during SARS-CoV-2 infection. Drug Suggestion Pirfenidone is an anti-fibrotic drug with an anti-oxidant activity that can prevent lung injury during SARS-CoV-2 infection by blocking the maturation process of transforming growth factor-beta (TGF-β) and enhancing the protective role of peroxisome proliferator-activated receptors (PPARs). Pirfenidone is a safe drug for patients with hypertension or diabetes and its side effect tolerated well. Conclusion The drug as a theoretical perspective may be an effective and safe choice for suppressing the inflammatory response during COVID-19. The recommendation would be a combination of pirfenidone and N-acetylcysteine to achieve maximum benefit during SARS-CoV-2 treatment.
Collapse
|
37
|
Rajan S, Schremmer C, Weber J, Alt P, Geiger F, Dietrich A. Ca 2+ Signaling by TRPV4 Channels in Respiratory Function and Disease. Cells 2021; 10:cells10040822. [PMID: 33917551 PMCID: PMC8067475 DOI: 10.3390/cells10040822] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/23/2021] [Accepted: 04/04/2021] [Indexed: 12/14/2022] Open
Abstract
Members of the transient receptor potential (TRP) superfamily are broadly expressed in our body and contribute to multiple cellular functions. Most interestingly, the fourth member of the vanilloid family of TRP channels (TRPV4) serves different partially antagonistic functions in the respiratory system. This review highlights the role of TRPV4 channels in lung fibroblasts, the lung endothelium, as well as the alveolar and bronchial epithelium, during physiological and pathophysiological mechanisms. Data available from animal models and human tissues confirm the importance of this ion channel in cellular signal transduction complexes with Ca2+ ions as a second messenger. Moreover, TRPV4 is an excellent therapeutic target with numerous specific compounds regulating its activity in diseases, like asthma, lung fibrosis, edema, and infections.
Collapse
|
38
|
Haywood N, Ta HQ, Rotar E, Daneva Z, Sonkusare SK, Laubach VE. Role of the purinergic signaling network in lung ischemia-reperfusion injury. Curr Opin Organ Transplant 2021; 26:250-257. [PMID: 33651003 PMCID: PMC9270688 DOI: 10.1097/mot.0000000000000854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD) is the leading cause of early mortality following lung transplantation and is typically caused by lung ischemia-reperfusion injury (IRI). Current management of PGD is largely supportive and there are no approved therapies to prevent lung IRI after transplantation. The purinergic signaling network plays an important role in this sterile inflammatory process, and pharmacologic manipulation of said network is a promising therapeutic strategy. This review will summarize recent findings in this area. RECENT FINDINGS In the past 18 months, our understanding of lung IRI has improved, and it is becoming clear that the purinergic signaling network plays a vital role. Recent works have identified critical components of the purinergic signaling network (Pannexin-1 channels, ectonucleotidases, purinergic P1 and P2 receptors) involved in inflammation in a number of pathologic states including lung IRI. In addition, a functionally-related calcium channel, the transient receptor potential vanilloid type 4 (TRPV4) channel, has recently been linked to purinergic signaling and has also been shown to mediate lung IRI. SUMMARY Agents targeting components of the purinergic signaling network are promising potential therapeutics to limit inflammation associated with lung IRI and thus decrease the risk of developing PGD.
Collapse
Affiliation(s)
- Nathan Haywood
- 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
| | - Evan Rotar
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| | - Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA
| | - Victor E. Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
39
|
Abstract
Introduction: Transient receptor potential vanilloid 4 (TRPV4) is an ion channel that is widely expressed and is activated by numerous chemical, osmotic and mechanical stimuli. By modulating Ca2+ entry, TRPV4 regulates cellular signaling associated with a variety of (patho)physiological processes and is a target of interest for treatment of human diseases including heart failure, respiratory diseases, gastrointestinal disorders, dermatological conditions, pain and cancer, among others.Areas covered: This article reviews small molecule TRPV4 antagonists and new therapeutic use claims disclosed in the patent literature from 2015 to 2020, including applications covering the first potent and selective TRPV4 clinical candidate and other advanced chemotypes.Expert opinion: TRPV4 has proven to be a tractable target and significant progress in discovery of TRPV4 antagonists has been realized in recent years. Several unique chemical templates with drug-like properties inhibit the channel and show efficacy in models that suggest their potential for treatment of a variety of diseases. While compelling clinical efficacy has not yet been seen in the limited early studies conducted with GSK2798745, evaluation of TRPV4 antagonists in larger trials across several indications is warranted given the availability of high-quality candidates and the promise of therapeutic benefit based on pre-clinical evidence.
Collapse
Affiliation(s)
- Brian G Lawhorn
- Medicinal Chemistry, Medicine Design, and Early Development Leaders, GlaxoSmithKline, Collegeville, Pennsylvania, United States
| | - Edward J Brnardic
- Medicinal Chemistry, Medicine Design, and Early Development Leaders, GlaxoSmithKline, Collegeville, Pennsylvania, United States
| | - David J Behm
- Medicinal Chemistry, Medicine Design, and Early Development Leaders, GlaxoSmithKline, Collegeville, Pennsylvania, United States
| |
Collapse
|
40
|
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
|
41
|
Goodman KE, Hare JT, Khamis ZI, Hua T, Sang QXA. Exposure of Human Lung Cells to Polystyrene Microplastics Significantly Retards Cell Proliferation and Triggers Morphological Changes. Chem Res Toxicol 2021; 34:1069-1081. [PMID: 33720697 DOI: 10.1021/acs.chemrestox.0c00486] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Microplastics in the environment produced by decomposition of globally increasing waste plastics have become a dominant component of both water and air pollution. To examine the potential toxicological effects of microplastics on human cells, the cultured human alveolar A549 cells were exposed to polystyrene microplastics (PS-MPs) of 1 and 10 μm diameter as a model of the environmental contaminants. Both sizes caused a significant reduction in cell proliferation but exhibited little cytotoxicity, as measured by the maintenance of cell viabilities determined by trypan blue staining and by Calcein-AM staining. The cell viabilities did not drop below 93% even at concentrations of PS-MPs as high as 100 μg/mL. Despite these high viabilities, further assays revealed a population level decrease in metabolic activity parallel in time with a dramatic decrease in proliferation rate in PS-MP exposed cells. Furthermore, phase contrast imaging of live cells at 72 h revealed major changes in the morphology of cells exposed to microplastics, as well as the uptake of multiple 1 μm PS-MPs into the cells. Confocal fluorescent microscopy at 24 h of exposure confirmed the incorporation of 1 μm PS-MPs. These disturbances at the proliferative and cytoskeletal levels of human cells lead us to propose that airborne polystyrene microplastics may have toxicologic consequences. This is the first report of exposure of human cells to an environmental contaminant resulting in the dual effects of inhibition of cell proliferation and major changes in cell morphology. Our results make clear that human exposure to microplastic pollution has significant consequence and potential for harm to humans.
Collapse
Affiliation(s)
- Kerestin E Goodman
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Joan T Hare
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Zahraa I Khamis
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States.,Department of Chemistry and Biochemistry, Lebanese University, Faculty of Sciences, Hadath-Beirut, Lebanon
| | - Timothy Hua
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306, United States.,Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
42
|
Michalick L, Weidenfeld S, Grimmer B, Fatykhova D, Solymosi PD, Behrens F, Dohmen M, Brack MC, Schulz S, Thomasch E, Simmons S, Müller-Redetzky H, Suttorp N, Kurth F, Neudecker J, Toennies M, Bauer TT, Eggeling S, Corman VM, Hocke AC, Witzenrath M, Hippenstiel S, Kuebler WM. Plasma mediators in patients with severe COVID-19 cause lung endothelial barrier failure. Eur Respir J 2021; 57:13993003.02384-2020. [PMID: 33154030 PMCID: PMC7651836 DOI: 10.1183/13993003.02384-2020] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/11/2020] [Indexed: 01/09/2023]
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 (BIH), Berlin, Germany
| | - Sarah Weidenfeld
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Benjamin Grimmer
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Diana Fatykhova
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Philip Daniel Solymosi
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Felix Behrens
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Melanie Dohmen
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Markus C Brack
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Sabrina Schulz
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Elisa Thomasch
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Szandor Simmons
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Holger Müller-Redetzky
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Norbert Suttorp
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,German Center for Lung Research (DZL), Germany
| | - Florian Kurth
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Jens Neudecker
- Dept of General, Visceral, Vascular and Thoracic Surgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Mario Toennies
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Berlin, Germany
| | - Torsten T Bauer
- Lungenklinik Heckeshorn, HELIOS Klinikum Emil von Behring, Berlin, Germany
| | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin, Germany
| | - Victor Max Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,German Centre for Infection Research (DZIF), Germany
| | - Andreas C Hocke
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,German Center for Lung Research (DZL), Germany
| | - Martin Witzenrath
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,German Center for Lung Research (DZL), Germany.,These authors contributed equally to this study
| | - Stefan Hippenstiel
- Dept of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.,German Center for Lung Research (DZL), Germany.,These authors contributed equally to this study
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany .,German Center for Lung Research (DZL), Germany.,The Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Depts. of Surgery and Physiology, University of Toronto, Toronto, ON, Canada.,German Centre for Cardiovascular Research (DZHK), Germany.,These authors contributed equally to this study
| |
Collapse
|
43
|
Molecular modeling of three-dimensional structure of hTRPV4 protein and experimental verification of its antagonist binding sites. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2020.129421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Dash P, Mohapatra S, Ghosh S, Nayak B. A Scoping Insight on Potential Prophylactics, Vaccines and Therapeutic Weaponry for the Ongoing Novel Coronavirus (COVID-19) Pandemic- A Comprehensive Review. Front Pharmacol 2021; 11:590154. [PMID: 33815095 PMCID: PMC8015872 DOI: 10.3389/fphar.2020.590154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/29/2020] [Indexed: 12/18/2022] Open
Abstract
The emergence of highly virulent CoVs (SARS-CoV-2), the etiologic agent of novel ongoing "COVID-19" pandemics has been marked as an alarming case of pneumonia posing a large global healthcare crisis of unprecedented magnitude. Currently, the COVID-19 outbreak has fueled an international demand in the biomedical field for the mitigation of the fast-spreading illness, all through the urgent deployment of safe, effective, and rational therapeutic strategies along with epidemiological control. Confronted with such contagious respiratory distress, the global population has taken significant steps towards a more robust strategy of containment and quarantine to halt the total number of positive cases but such a strategy can only delay the spread. A substantial number of potential vaccine candidates are undergoing multiple clinical trials to combat COVID-19 disease, includes live-attenuated, inactivated, viral-vectored based, sub-unit vaccines, DNA, mRNA, peptide, adjuvant, plant, and nanoparticle-based vaccines. However, there are no licensed anti-COVID-19 drugs/therapies or vaccines that have proven to work as more effective therapeutic candidates in open-label clinical trial studies. To counteract the infection (SARS-CoV-2), many people are under prolonged treatment of many chemical drugs that inhibit the PLpro activity (Ribavirin), viral proteases (Lopinavir/Ritonavir), RdRp activity (Favipiravir, Remdesivir), viral membrane fusion (Umifenovir, Chloroquine phosphate (CQ), Hydroxychloroquine phosphate (HCQ), IL-6 overexpression (Tocilizumab, Siltuximab, Sarilumab). Mesenchymal Stem Cell therapy and Convalescent Plasma Therapy have emerged as a promising therapeutic strategy against SARS-CoV-2 virion. On the other hand, repurposing previously designed antiviral agents with tolerable safety profile and efficacy could be the only promising approach and fast response to the novel virion. In addition, research institutions and corporations have commenced the redesign of the available therapeutic strategy to manage the global crisis. Herein, we present succinct information on selected anti-COVID-19 therapeutic medications repurposed to combat SARS-CoV-2 infection. Finally, this review will provide exhaustive detail on recent prophylactic strategies and ongoing clinical trials to curb this deadly pandemic, outlining the major therapeutic areas for researchers to step in.
Collapse
Affiliation(s)
| | | | | | - Bismita Nayak
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, India
| |
Collapse
|
45
|
Abdel Hameid R, Cormet-Boyaka E, Kuebler WM, Uddin M, Berdiev BK. SARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport. Am J Physiol Lung Cell Mol Physiol 2021; 320:L430-L435. [PMID: 33434105 PMCID: PMC7938641 DOI: 10.1152/ajplung.00499.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The tropism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a virus responsible for the ongoing coronavirus disease 2019 (COVID-19) pandemic, toward the host cells is determined, at least in part, by the expression and distribution of its cell surface receptor, angiotensin-converting enzyme 2 (ACE2). The virus further exploits the host cellular machinery to gain access into the cells; its spike protein is cleaved by a host cell surface transmembrane serine protease 2 (TMPRSS2) shortly after binding ACE2, followed by its proteolytic activation at a furin cleavage site. The virus primarily targets the epithelium of the respiratory tract, which is covered by a tightly regulated airway surface liquid (ASL) layer that serves as a primary defense mechanism against respiratory pathogens. The volume and viscosity of this fluid layer is regulated and maintained by a coordinated function of different transport pathways in the respiratory epithelium. We argue that SARS-CoV-2 may potentially alter evolutionary conserved second-messenger signaling cascades via activation of G protein-coupled receptors (GPCRs) or by directly modulating G protein signaling. Such signaling may in turn adversely modulate transepithelial transport processes, especially those involving cystic fibrosis transmembrane conductance regulator (CFTR) and epithelial Na+ channel (ENaC), thereby shifting the delicate balance between anion secretion and sodium absorption, which controls homeostasis of this fluid layer. As a result, activation of the secretory pathways including CFTR-mediated Cl− transport may overwhelm the absorptive pathways, such as ENaC-dependent Na+ uptake, and initiate a pathophysiological cascade leading to lung edema, one of the most serious and potentially deadly clinical manifestations of COVID-19.
Collapse
Affiliation(s)
- Reem Abdel Hameid
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | | | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mohammed Uddin
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Bakhrom K Berdiev
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
46
|
Zhou X, Chen D, Wang L, Zhao Y, Wei L, Chen Z, Yang B. Low serum calcium: a new, important indicator of COVID-19 patients from mild/moderate to severe/critical. Biosci Rep 2020; 40:BSR20202690. [PMID: 33252122 PMCID: PMC7755121 DOI: 10.1042/bsr20202690] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) virus is still spreading, finding out the initial hits of viral infection is important to minimize the mild/moderate population, prevent disease aggravation and organs dysfunction. Objective: We investigated COVID-19 patients with different serum calcium levels. DESIGN We checked the serum calcium level of the patients based on days after symptom onset as well as the severity of COVID-19. We also checkeed multi-organ injuries and immune cytokines level in their blood. RESULTS Both mild/moderate and severe critical cases we observed showed low calcium level in the early stage of viral infection, while the severe/critical cases showed significant lower calcium level than mild/moderate cases in the early stage. We also found that low calcium level related to severe/critical multi-organ injuries especially in the mild/moderate population. Proinflammatory cytokine IL-6 also correlated to calcium change in both mild/moderate and severe/critical cases. CONCLUSIONS Our finding indicates that calcium balance is a primal hit of COVID-19 and a biomarker of clinical severity at the beginning of symptom onset. Calcium is closely associated with virus-associated multiple organ injuryes and the increase of inflammatory cytokines. Our results provide a new, important indicator of COVID-19 patients from mild/moderate to severe/critical: serum calcium.
Collapse
Affiliation(s)
- Xi Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Dong Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Lan Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| |
Collapse
|
47
|
Zubcevic L. Temperature‐sensitive transient receptor potential vanilloid channels: structural insights into ligand‐dependent activation. Br J Pharmacol 2020; 179:3542-3559. [DOI: 10.1111/bph.15310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/19/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Lejla Zubcevic
- Department of Biochemistry and Molecular Biology The University of Kansas School of Medicine Kansas City KS USA
| |
Collapse
|
48
|
Design, Synthesis and In Vitro Experimental Validation of Novel TRPV4 Antagonists Inspired by Labdane Diterpenes. Mar Drugs 2020; 18:md18100519. [PMID: 33081023 PMCID: PMC7594054 DOI: 10.3390/md18100519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023] Open
Abstract
Labdane diterpenes are widespread classes of natural compounds present in variety of marine and terrestrial organisms and plants. Many of them represents “natural libraries” of compounds with interesting biological activities due to differently functionalized drimane nucleus exploitable for potential pharmacological applications. The transient receptor potential channel subfamily V member 4 (TRPV4) channel has recently emerged as a pharmacological target for several respiratory diseases, including the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Inspired by the labdane-like bicyclic core, a series of homodrimane-derived esters and amides was designed and synthesized by modifying the flexible tail in position 1 of (+)-sclareolide, an oxidized derivative of the bioactive labdane-type diterpene sclareol. The potency and selectivity towards rTRPV4 and hTRPV1 receptors were assessed by calcium influx cellular assays. Molecular determinants critical for eliciting TRPV4 antagonism were identified by structure-activity relationships. Among the selective TRPV4 antagonists identified, compound 6 was the most active with an IC50 of 5.3 μM. This study represents the first report of semisynthetic homodrimane TRPV4 antagonists, selective over TRPV1, and potentially useful as pharmacological tools for the development of novel TRPV4 channel modulators.
Collapse
|
49
|
Witzenrath M, Kuebler WM. Pneumonia in the face of COVID-19. Am J Physiol Lung Cell Mol Physiol 2020; 319:L863-L866. [PMID: 32996786 PMCID: PMC7839244 DOI: 10.1152/ajplung.00447.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Martin Witzenrath
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,Division of Pulmonary Inflammation, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Partner site Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of the Freie Universität Berlin, Humboldt Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.,German Center for Lung Research (DZL), Partner site Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,Departments of Physiology and Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Swayne LA, Johnstone SR, Ng CS, Sanchez-Arias JC, Good ME, Penuela S, Lohman AW, Wolpe AG, Laubach VE, Koval M, Isakson BE. Consideration of Pannexin 1 channels in COVID-19 pathology and treatment. Am J Physiol Lung Cell Mol Physiol 2020; 319:L121-L125. [PMID: 32519892 PMCID: PMC7347959 DOI: 10.1152/ajplung.00146.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion Center for Heart and Reparative Medicine Research, Virginia Tech, Roanoke, Virginia.,Department of Biological Sciences, Virginia Tech, Roanoke, Virginia
| | - Chen Seng Ng
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Juan C Sanchez-Arias
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Alexander W Lohman
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Michael Koval
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|