1
|
Li S, Fu Z, Hong W, Yuan H, Cao W, Xu J, Liu R, Lin Z, Xiang Z, Peng G. Chronic hypoxia promotes pulmonary venous smooth muscle cell proliferation through the CaSR-TRPC6/ROCE pathway. Exp Cell Res 2024; 444:114363. [PMID: 39637934 DOI: 10.1016/j.yexcr.2024.114363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The mechanism underlying chronic hypoxia (CH)-induced pulmonary venous remodeling remains unclear. Cell proliferation is key in vascular remodeling, and the calcium-sensing receptor (CaSR) protein contributes to CH-induced pulmonary venous smooth muscle cell (PVSMC) proliferation. In pulmonary arterial smooth muscle cells, CaSR and transient receptor potential canonical (TRPC) proteins interact, contributing to CH-induced cell proliferation via CaSR-TRPC1/6 signaling. We investigated whether a similar pathway exists in PVSMCs. Rat PVSMCs were isolated and subjected to CH. Cell proliferation was assessed by cell counting, CCK-8, and BrdU incorporation assays. Expression of CaSR and TRPC was analyzed by qPCR and western blotting, while interactions between CaSR and TRPC were detected by co-immunoprecipitation assay. Extracellular Ca2+ restoration was evaluated, to assess store- and receptor-operated Ca2+ entry (SOCE and ROCE, respectively). CH enhanced PVSMC numbers, viability, and DNA synthesis, and upregulated CaSR and TRPC6 expression. Further, CaSR and TRPC6 interacted with one another. CaSR inhibitors (NPS2143, NPS2390) reduced, whereas activators (spermine, R568) enhanced, CH-induced increases in PVSMC numbers, viability, DNA synthesis, and TRPC6 expression. CaSR knockdown using siRNA inhibited CH-induced TRPC6 upregulation and attenuated CH-induced increases in PVSMC numbers, viability, and DNA synthesis. TRPC6 knockdown had no significant effect on CH-induced CaSR upregulation, but significantly attenuated CH-induced increases in PVSMC number, viability, and DNA synthesis. CaSR knockdown reduced ROCE, but not SOCE, enhancement. Overall, CH promotes PVSMC proliferation through the CaSR-TRPC6/ROCE pathway.
Collapse
Affiliation(s)
- Shaoxing Li
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhenli Fu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Geriatric Respiratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wei Hong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong- Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong Yuan
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Weitao Cao
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Juan Xu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Rongmin Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhuandi Lin
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiming Xiang
- The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Zhao E, Wang J, Zhao Y, Xia Q, Wang H, Li Z, Li C, Gai X. Echinacoside inhibits PASMCs calcium overload to prevent hypoxic pulmonary artery remodeling by regulating TRPC1/4/6 and calmodulin. Open Med (Wars) 2024; 19:20241044. [PMID: 39381430 PMCID: PMC11459269 DOI: 10.1515/med-2024-1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 08/11/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024] Open
Abstract
Abstract Research indicates that hypoxic pulmonary hypertension (HPH) potentially stimulates the sympathetic nervous system, which may increase norepinephrine (NE) release and cause excessive Ca2+ influx into pulmonary artery smooth muscle cells (PASMCs), leading to calcium overload and abnormal PASMC proliferation, factors closely associated with pulmonary vascular remodeling (PVR). This study investigates the potential mechanisms underlying echinacoside (ECH) treatment in HPH. Method In the in vitro experiment, NE-induced PASMCs were used to simulate HPH-induced PASMCs' calcium overload and abnormal proliferation. Postincubation with ECH, [Ca2+]cyt changes were detected using Fluo-4 AM. Flow cytometry was employed to ascertain ECH's inhibitory effect on PASMCs proliferation. For in vivo experiments, rats were exposed to a hypoxic and low-pressure oxygen environment to establish the HPH model. Post-ECH treatment, hematoxylin and eosin (HE) staining was conducted to assess PVR, and western blot analysis was used to examine protein expression in the lung tissues of the different groups. Results ECH was observed to inhibit [Ca2+]cyt increase in NE-induced PASMCs in a concentration-dependent manner, effectively reducing abnormal cell proliferation. It also reduced the expression of Transient receptor potential channel (TRPC) 1 (TRPC1), TRPC4, TRPC6, and calmodulin in PASMCs. In vivo studies demonstrated that ECH lowered the expression of these proteins in lung tissues of HPH rats, significantly decreased mean pulmonary artery pressure, and mitigated PVR.
Collapse
Affiliation(s)
- Enqi Zhao
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| | - Jinyu Wang
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| | - Yuefu Zhao
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| | - Qingqing Xia
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| | - Hongmai Wang
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| | - Zhanqiang Li
- Qinghai University Plateau Medicine Research Center, Xining, Qinghai Province, China
| | - Cen Li
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai Province, China
| | - Xiangyun Gai
- School of Pharmacy, Qinghai Minzu University, Xining, Qinghai, 810007, China
| |
Collapse
|
3
|
Wang L, Tang T, Tian X, Peng C, Wu S. Animal models of pulmonary arterial hypertension associated with atrial septal defect. Sci Rep 2024; 14:18287. [PMID: 39112674 PMCID: PMC11306221 DOI: 10.1038/s41598-024-69002-5] [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: 01/05/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a well-known complication of congenital heart disease (CHD). The lack of a satisfactory animal model for PAH associated with CHD (PAH-CHD) has limited progress in understanding the pathogenesis of PAH and the development of therapeutic agents. The development of a rat model for PAH associated with atrial septal defect (ASD) was achieved through atrial septal puncture and thermal ablation. Two and 4 weeks after modeling, hematoxylin and eosin staining showed that the vascular thickness, vascular thickness index, vascular area, and vascular area index in pulmonary arteries with an outer diameter of 50-300 μm in the PAH-ASD 2 and 4 weeks group were higher than those in the sham group (all P < 0.05). Alpha-smooth muscle actin (ɑ-SMA) staining showed that the medial thickness, medial thickness index, medial area, and medial area index in pulmonary arteries with an outer diameter of 50-300 µm at 2 and 4 weeks after modeling were significantly higher than those in the sham group (all P < 0.05). Additionally, mean pulmonary arterial pressure (mPAP) and pulmonary vascular resistance (PVR) in the PAH-ASD 2 and 4 weeks groups were significantly higher than those in the sham group (both P < 0.05). Elastin van Gieson staining showed that the vascular obstruction score in the PAH-ASD 2 and 4 weeks group was significantly higher than that in the sham group (both P < 0.05). The PAH-ASD rats were successfully generated. These findings suggest that our model would be useful for further research into the pathogenesis, prevention, and treatment of PAH-ASD.
Collapse
Affiliation(s)
- Li Wang
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Ting Tang
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Xiaochun Tian
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| | - Chang Peng
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China.
| | - Shuqi Wu
- Department of Pediatrics, Guizhou Children's Hospital, Affiliated Hospital of Zunyi Medical University, 149 Dalian Street, Zunyi, 563000, Guizhou, People's Republic of China
| |
Collapse
|
4
|
Wang B, Xu Y, Huang Y, Shao S, Xu D, Zhang Y, Pang L, Nan Z, Ye Q, Wang Y, Wang W, Jin K, Yuan L. miR-210-5p Promotes Pulmonary Hypertension by Blocking ATP2A2. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07568-y. [PMID: 38656637 DOI: 10.1007/s10557-024-07568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/26/2024]
Abstract
AIM Aberrant expression of ATPase sarcoplasmic/endoplasmic retic Ca2+ transporting 2 (ATP2A2) has attracted attention for its pathophysiologic role in pulmonary hypertension (PH). Several miRNAs, including miR-210-5p, have also been reported to be pathogenic factors in PH, but their exact mechanisms remain unknown. This study aimed to elucidate the potential mechanisms of miR-210-5p and ATP2A2 in MCT-induced PH. METHODS Eighteen Sprague-Dawley rats were randomly divided into two groups-monoclonal (MCT) group and control group-and then administered MCT (60 mg/kg) and saline, respectively. mPAP, PVR, RVHI, WT%, and WA% were significantly increased in PH rats after 3 weeks, confirming that the modeling of PH rats was successful. Subsequently, we determined the expression of ATP2A2 and miR-210-5p in lung tissues using WB and qRT-PCR methods. We established an in vitro model using BMP4 and TGF-β1 treatment of pulmonary artery smooth muscle cells (PASMCs) and examined the expression of ATP2A2 and miR-210-5p using the same method. To further elucidate the regulatory relationship between ATP2A2 and miR-210-5p, we altered the expression level of miR-210-5p and detected the corresponding changes in ATP2A2 levels. In addition, we demonstrated the relationship by dual luciferase experiments. Finally, the effect of silencing ATP2A2 could be confirmed by the level of cell membrane Ca2+ in PAMSCs. RESULTS Up-regulation of miR-210-5p and down-regulation of ATP2A2 were observed in the MCT group compared with the control group, which was confirmed in the in vitro model. In addition, elevated miR-210-5p expression decreased the level of ATP2A2 while increasing the proliferation of PASMCs, and the results of the dual luciferase assay further confirmed that ATP2A2 is a downstream target of miR-210-5p. Additionally, silencing ATP2A2 resulted in increased cytoplasmic Ca2+ levels in PAMSCs. CONCLUSION In MCT-induced PH, miR-210-5p promotes pulmonary vascular remodeling by inhibiting ATP2A2.
Collapse
Affiliation(s)
- Boxiang Wang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yidin Xu
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yilun Huang
- Alberta Institute, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Siming Shao
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Dongshan Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yiying Zhang
- Alberta Institute, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Lingxia Pang
- Functionality Experimental Teaching Center, Basic Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Zhuofan Nan
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
- Department of Urology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Qianxi Ye
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, People's Republic of China
- Department of Cardiovascular Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Yang Wang
- Department of Pathophysiology, Basic Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Wantie Wang
- Department of Pathophysiology, Basic Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Keke Jin
- Department of Pathophysiology, Basic Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Linbo Yuan
- Department of Physiology, Basic Medical School, Wenzhou Medical University, Wenzhou, People's Republic of China.
| |
Collapse
|
5
|
Shan X, Gegentuya, Wang J, Feng H, Zhang Z, Zheng Q, Zhang Q, Yang K, Wang J, Xu L. Aloperine protects pulmonary hypertension via triggering PPARγ signaling and inhibiting calcium regulatory pathway in pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2023; 325:C1058-C1072. [PMID: 37661916 DOI: 10.1152/ajpcell.00286.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/20/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Previous studies have reported the beneficial role of Aloperine (ALO), an active vasodilator purified from the seeds and leaves of the herbal plant Sophora alopecuroides L., on experimental pulmonary hypertension (PH); however, detailed mechanisms remain unclear. In this study, monocrotaline-induced PH (MCT-PH) rat model and primarily cultured rat distal pulmonary arterial smooth muscle cells (PASMCs) were used to investigate the mechanisms of ALO on experimental PH, pulmonary vascular remodeling, and excessive proliferation of PASMCs. Results showed that first, ALO significantly prevented the disease development of MCT-PH by inhibiting right ventricular systolic pressure (RVSP) and right ventricular hypertrophy indexed by the Fulton Index, normalizing the pulmonary arterials (PAs) remodeling and improving the right ventricular function indexed by transthoracic echocardiography. ALO inhibited the excessive proliferation of both PAs and PASMCs. Then, isometric tension measurements showed vasodilation of ALO on precontracted PAs isolated from both control and MCT-PH rats via activating the KCNQ channel, which was blocked by specific KCNQ potassium channel inhibitor linopirdine. Moreover, by using immunofluorescence staining and nuclear/cytosol fractionation, we further observed that ALO significantly enhanced the PPARγ nuclear translocation and activation in PASMCs. Transcriptome analyses also revealed activated PPARγ signaling and suppressed calcium regulatory pathway in lungs from MCT-PH rats treated with ALO. In summary, ALO could attenuate MCT-PH through both transient vasodilation of PAs and chronic activation of PPARγ signaling pathway, which exerted antiproliferative roles on PASMCs and remodeled PAs.NEW & NOTEWORTHY Aloperine attenuates monocrotaline-induced pulmonary hypertension (MCT-PH) in rats by inhibiting the pulmonary vascular remodeling and proliferation of pulmonary arterial smooth muscle cells (PASMCs). In mechanism, Aloperine not only exerts a transient KCNQ-dependent vasodilation in precontracted pulmonary arteries (PAs) from both control and MCT-PH rats but also activates PPARγ nuclear translocation and signaling transduction in PASMCs, which chronically inhibits the calcium regulatory pathway and proliferation of PASMCs.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Calcium/metabolism
- Calcium Signaling/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/prevention & control
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- KCNQ Potassium Channels/metabolism
- KCNQ Potassium Channels/genetics
- Monocrotaline/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Piperidines/pharmacology
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Quinolizidines/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Vascular Remodeling/drug effects
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Xiaoqian Shan
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- 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, China
| | - Gegentuya
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
- 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, China
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Jing Wang
- Department of Scientific Research, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 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, China
| | - Zizhou Zhang
- 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, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Qiuyu Zheng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qing Zhang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - 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, 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, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, China
| | - Lei Xu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
6
|
Balistrieri A, Makino A, Yuan JXJ. Pathophysiology and pathogenic mechanisms of pulmonary hypertension: role of membrane receptors, ion channels, and Ca 2+ signaling. Physiol Rev 2023; 103:1827-1897. [PMID: 36422993 PMCID: PMC10110735 DOI: 10.1152/physrev.00030.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The pulmonary circulation is a low-resistance, low-pressure, and high-compliance system that allows the lungs to receive the entire cardiac output. Pulmonary arterial pressure is a function of cardiac output and pulmonary vascular resistance, and pulmonary vascular resistance is inversely proportional to the fourth power of the intraluminal radius of the pulmonary artery. Therefore, a very small decrease of the pulmonary vascular lumen diameter results in a significant increase in pulmonary vascular resistance and pulmonary arterial pressure. Pulmonary arterial hypertension is a fatal and progressive disease with poor prognosis. Regardless of the initial pathogenic triggers, sustained pulmonary vasoconstriction, concentric vascular remodeling, occlusive intimal lesions, in situ thrombosis, and vascular wall stiffening are the major and direct causes for elevated pulmonary vascular resistance in patients with pulmonary arterial hypertension and other forms of precapillary pulmonary hypertension. In this review, we aim to discuss the basic principles and physiological mechanisms involved in the regulation of lung vascular hemodynamics and pulmonary vascular function, the changes in the pulmonary vasculature that contribute to the increased vascular resistance and arterial pressure, and the pathogenic mechanisms involved in the development and progression of pulmonary hypertension. We focus on reviewing the pathogenic roles of membrane receptors, ion channels, and intracellular Ca2+ signaling in pulmonary vascular smooth muscle cells in the development and progression of pulmonary hypertension.
Collapse
Affiliation(s)
- Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- Harvard University, Cambridge, Massachusetts
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - 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, California
| |
Collapse
|
7
|
Homo sapiens—A Species Not Designed for Space Flight: Health Risks in Low Earth Orbit and Beyond, Including Potential Risks When Traveling beyond the Geomagnetic Field of Earth. Life (Basel) 2023; 13:life13030757. [PMID: 36983912 PMCID: PMC10051707 DOI: 10.3390/life13030757] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Homo sapiens and their predecessors evolved in the context of the boundary conditions of Earth, including a 1 g gravity and a geomagnetic field (GMF). These variables, plus others, led to complex organisms that evolved under a defined set of conditions and define how humans will respond to space flight, a circumstance that could not have been anticipated by evolution. Over the past ~60 years, space flight and living in low Earth orbit (LEO) have revealed that astronauts are impacted to varying degrees by such new environments. In addition, it has been noted that astronauts are quite heterogeneous in their response patterns, indicating that such variation is either silent if one remained on Earth, or the heterogeneity unknowingly contributes to disease development during aging or in response to insults. With the planned mission to deep space, humans will now be exposed to further risks from radiation when traveling beyond the influence of the GMF, as well as other potential risks that are associated with the actual loss of the GMF on the astronauts, their microbiomes, and growing food sources. Experimental studies with model systems have revealed that hypogravity conditions can influence a variety biological and physiological systems, and thus the loss of the GMF may have unanticipated consequences to astronauts’ systems, such as those that are electrical in nature (i.e., the cardiovascular system and central neural systems). As astronauts have been shown to be heterogeneous in their responses to LEO, they may require personalized countermeasures, while others may not be good candidates for deep-space missions if effective countermeasures cannot be developed for long-duration missions. This review will discuss several of the physiological and neural systems that are affected and how the emerging variables may influence astronaut health and functioning.
Collapse
|
8
|
Lazrak A, Song W, Yu Z, Zhang S, Nellore A, Hoopes CW, Woodworth BA, Matalon S. Low molecular weight hyaluronan inhibits lung epithelial ion channels by activating the calcium-sensing receptor. Matrix Biol 2023; 116:67-84. [PMID: 36758905 PMCID: PMC10012407 DOI: 10.1016/j.matbio.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Herein, we tested the hypothesis that low molecular weight hyaluronan (LMW-HA) inhibits lung epithelial ions transport in-vivo, ex-vivo, and in-vitro by activating the calcium-sensing receptor (CaSR). Twenty-four hours post intranasal instillation of 50-150 µg/ml LMW-HA to C57BL/6 mice, there was a 75% inhibition of alveolar fluid clearance (AFC), a threefold increase in the epithelial lining fluid (ELF) depth, and a 20% increase in lung wet/dry (W/D) ratio. Incubation of human and mouse precision cut lung slices with 150 µg/ml LMW-HA reduced the activity and the open probability (Po) of epithelial sodium channel (ENaC) in alveolar epithelial type 2 (ATII) cells, and in mouse tracheal epithelial cells (MTEC) monolayers as early as 4 h. The Cl- current through cystic fibrosis transmembrane conductance regulator (CFTR) and the activity of Na,K-ATPase were both inhibited by more than 66% at 24 h. The inhibitory effects of LMW-HA on ion channels were reversed by 1 µM NPS-2143, or 150 µg/ml high molecular weight hyaluronan (HMW-HA). In HEK-293 cells expressing the calcium-sensitive Cl- channel TMEM16-A, CaSR was required for the activation of the Cl- current by LMW-HA. This is the first demonstration of lung ions and water transport inhibition by LMW-HA, and its mediation through the activation of CaSR.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA.
| | - Weifeng Song
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Shaoyan Zhang
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Anoma Nellore
- Department of Medicine, Division of Infectious Diseases, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Charles W Hoopes
- Division of Cardiothoracic Surgery, Heersink School of Medicine, University of Alabama at Birmingham, AL 35295, USA
| | - Bradford A Woodworth
- Department of Otolaryngology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Gregory Fleming James Cystic Fibrosis Research Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Division of Molecular and Translational Biomedicine, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA; Pulmonary Injury and Repair Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35295, USA
| |
Collapse
|
9
|
Malkmus K, Brosien M, Knoepp F, Schaffelhofer L, Grimminger F, Rummel C, Gudermann T, Dietrich A, Birnbaumer L, Weissmann N, Kraut S. Deletion of classical transient receptor potential 1, 3 and 6 alters pulmonary vasoconstriction in chronic hypoxia-induced pulmonary hypertension in mice. Front Physiol 2022; 13:1080875. [PMID: 36569761 PMCID: PMC9768328 DOI: 10.3389/fphys.2022.1080875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic hypoxia-induced pulmonary hypertension (CHPH) is a severe disease that is characterized by increased proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) leading to pulmonary vascular remodeling. The resulting increase in pulmonary vascular resistance (PVR) causes right ventricular hypertrophy and ultimately right heart failure. In addition, increased PVR can also be a consequence of hypoxic pulmonary vasoconstriction (HPV) under generalized hypoxia. Increased proliferation and migration of PASMCs are often associated with high intracellular Ca2+ concentration. Recent publications suggest that Ca2+-permeable nonselective classical transient receptor potential (TRPC) proteins-especially TRPC1 and 6-are crucially involved in acute and sustained hypoxic responses and the pathogenesis of CHPH. The aim of our study was to investigate whether the simultaneous deletion of TRPC proteins 1, 3 and 6 protects against CHPH-development and affects HPV in mice. We used a mouse model of chronic hypoxia as well as isolated, ventilated and perfused mouse lungs and PASMC cell cultures. Although right ventricular systolic pressure as well as echocardiographically assessed PVR and right ventricular wall thickness (RVWT) were lower in TRPC1, 3, 6-deficient mice, these changes were not related to a decreased degree of pulmonary vascular muscularization and a reduced proliferation of PASMCs. However, both acute and sustained HPV were almost absent in the TRPC1, 3, 6-deficient mice and their vasoconstrictor response upon KCl application was reduced. This was further validated by myographical experiments. Our data revealed that 1) TRPC1, 3, 6-deficient mice are partially protected against development of CHPH, 2) these changes may be caused by diminished HPV and not an altered pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Kathrin Malkmus
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Monika Brosien
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Fenja Knoepp
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Lisa Schaffelhofer
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Friedrich Grimminger
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Thomas Gudermann
- Walther Straub Institute for Pharmacology and Toxicology, Member of the DZL, Ludwig Maximilians University, Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute for Pharmacology and Toxicology, Member of the DZL, Ludwig Maximilians University, Munich, Germany
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina,Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), Durham, United States
| | - Norbert Weissmann
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Simone Kraut
- Cardiopulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany,*Correspondence: Simone Kraut,
| |
Collapse
|
10
|
Chan GHH, Chan E, Kwok CTK, Leung GPH, Lee SMY, Seto SW. The role of p53 in the alternation of vascular functions. Front Pharmacol 2022; 13:981152. [PMID: 36147350 PMCID: PMC9485942 DOI: 10.3389/fphar.2022.981152] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Ageing is a risk factor for many degenerative diseases. Cardiovascular diseases (CVDs) are usually big burdens for elderly, caregivers and the health system. During the aging process, normal functions of vascular cells and tissue progressively lost and eventually develop vascular diseases. Endothelial dysfunction, reduced bioavailability of endothelium-derived nitric oxide are usual phenomena observed in patients with cardiovascular diseases. Myriad of studies have been done to investigate to delay the vascular dysfunction or improve the vascular function to prolong the aging process. Tumor suppressor gene p53, also a transcription factor, act as a gatekeeper to regulate a number of genes to maintain normal cell function including but not limited to cell proliferation, cell apoptosis. p53 also crosstalk with other key transcription factors like hypoxia-inducible factor 1 alpha that contribute to the progression of cardiovascular diseases. Therefore, in recent three decades, p53 has drawn scientists’ attention on its effects in vascular function. Though the role of tumor suppressor gene p53 is still not clear in vascular function, it is found to play regulatory roles and may involve in vascular remodeling, atherosclerosis or pulmonary hypertension. p53 may have a divergent role in endothelial and vascular muscle cells in those conditions. In this review, we describe the different effects of p53 in cardiovascular physiology. Further studies on the effects of endothelial cell-specific p53 deficiency on atherosclerotic plaque formation in common animal models are required before the therapeutic potential can be realized.
Collapse
Affiliation(s)
- Gabriel Hoi-Huen Chan
- Division of Science, Engineering and Health Studies, College of Professional and Continuing Education, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Enoch Chan
- School of Clinical Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Carsten Tsun-Ka Kwok
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, China
| | - Sai-Wang Seto
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
- *Correspondence: Sai-Wang Seto,
| |
Collapse
|
11
|
Mechanistic and therapeutic perspectives of baicalin and baicalein on pulmonary hypertension: A comprehensive review. Biomed Pharmacother 2022; 151:113191. [PMID: 35643068 DOI: 10.1016/j.biopha.2022.113191] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 11/20/2022] Open
Abstract
Pulmonary hypertension (PH) is a chronic and fatal disease, for which new therapeutic drugs and approaches are needed urgently. Baicalein and baicalin, the active compounds of the traditional Chinese medicine, Scutellaria baicalensis Georgi, exhibit a wide range of pharmacological activities. Numerous studies involving in vitro and in vivo models of PH have revealed that the treatment with baicalin and baicalein may be effective. This review summarizes the potential mechanisms driving the beneficial effects of baicalin and baicalein treatment on PH, including anti-inflammatory response, inhibition of pulmonary smooth muscle cell proliferation and endothelial-to-mesenchymal transformation, stabilization of the extracellular matrix, and mitigation of oxidative stress. The pharmacokinetics of these compounds have also been reviewed. The therapeutic potential of baicalin and baicalein warrants their continued study as natural treatments for PH.
Collapse
|
12
|
Mayer CA, Roos B, Teske J, Wells N, Martin RJ, Chang W, Pabelick CM, Prakash YS, MacFarlane PM. Calcium-sensing receptor and CPAP-induced neonatal airway hyperreactivity in mice. Pediatr Res 2022; 91:1391-1398. [PMID: 33958714 PMCID: PMC8571113 DOI: 10.1038/s41390-021-01540-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/15/2021] [Accepted: 04/05/2021] [Indexed: 12/03/2022]
Abstract
BACKGROUND Continuous positive airway pressure (CPAP) in preterm infants is initially beneficial, but animal models suggest longer term detrimental airway effects towards asthma. We used a neonatal CPAP mouse model and human fetal airway smooth muscle (ASM) to investigate the role of extracellular calcium-sensing receptor (CaSR) in these effects. METHODS Newborn wild type and smooth muscle-specific CaSR-/- mice were given CPAP for 7 days via a custom device (mimicking CPAP in premature infants), and recovered in normoxia for another 14 days (representing infants at 3-4 years). Airway reactivity was tested using lung slices, and airway CaSR quantified. Role of CaSR was tested using NPS2143 (inhibitor) or siRNA in WT mice. Fetal ASM cells stretched cyclically with/without static stretch mimicking breathing and CPAP were analyzed for intracellular Ca2+ ([Ca2+]i) responses, role of CaSR, and signaling cascades. RESULTS CPAP increased airway reactivity in WT but not CaSR-/- mice, increasing ASM CaSR. NPS2143 or CaSR siRNA reversed CPAP effects in WT mice. CPAP increased fetal ASM [Ca2+]I, blocked by NPS2143, and increased ERK1/2 and RhoA suggesting two mechanisms by which stretch increases CaSR. CONCLUSIONS These data implicate CaSR in CPAP effects on airway function with implications for wheezing in former preterm infants. IMPACT Neonatal CPAP increases airway reactivity to bronchoconstrictor agonist. CPAP increases smooth muscle expression of the extracellular calcium-sensing receptor (CaSR). Inhibition or absence of CaSR blunts CPAP effects on contractility. These data suggest a causal/contributory role for CaSR in stretch effects on the developing airway. These data may impact clinical recognition of the ways that CPAP may contribute to wheezing disorders of former preterm infants.
Collapse
Affiliation(s)
- Catherine A Mayer
- Department of Pediatrics, Division of Neonatology, Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jacob Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Natalya Wells
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Richard J Martin
- Department of Pediatrics, Division of Neonatology, Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Wenhan Chang
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| | - Peter M MacFarlane
- Department of Pediatrics, Division of Neonatology, Rainbow Babies & Children's Hospital, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
13
|
Lu T, Zhang Y, Su Y, Zhou D, Xu Q. Role of store-operated Ca2+ entry in cardiovascular disease. Cell Commun Signal 2022; 20:33. [PMID: 35303866 PMCID: PMC8932232 DOI: 10.1186/s12964-022-00829-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/14/2022] [Indexed: 01/01/2023] Open
Abstract
Store-operated channels (SOCs) are highly selective Ca2+ channels that mediate Ca2+ influx in non-excitable and excitable (i.e., skeletal and cardiac muscle) cells. These channels are triggered by Ca2+ depletion of the endoplasmic reticulum and sarcoplasmic reticulum, independently of inositol 1,4,5-trisphosphate (InsP3), which is involved in cell growth, differentiation, and gene transcription. When the Ca2+ store is depleted, stromal interaction molecule1 (STIM1) as Ca2+ sensor redistributes into discrete puncta near the plasma membrane and activates the protein Ca2+ release activated Ca2+ channel protein 1 (Orai1). Accumulating evidence suggests that SOC is associated with several physiological roles in endothelial dysfunction and vascular smooth muscle proliferation that contribute to the progression of cardiovascular disease. This review mainly elaborates on the contribution of SOC in the vasculature (endothelial cells and vascular smooth muscle cells). We will further retrospect the literature implicating a critical role for these proteins in cardiovascular disease.
Collapse
Affiliation(s)
- Ting Lu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yihua Zhang
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Yong Su
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Dayan Zhou
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China
| | - Qiang Xu
- Department of Cardiology, Chongqing Fifth People's Hospital, No. 24 Renji Road, Chongqing, 400000, China.
| |
Collapse
|
14
|
Zhou Y, Yan H, Li T, Xie M, Li X, Zhao C. New use of old medicine: Nifedipine acts on the TRP family and inflammatory proteins in the treatment of chilblain. Burns 2022; 48:372-380. [PMID: 34103198 DOI: 10.1016/j.burns.2021.05.005] [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: 01/27/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 12/14/2022]
Abstract
As a calcium antagonist, the mechanism of nifedipine for treating chilblain has not been reported. In the present study, we established the chilblain model by using -20 ℃ 95% ethanol to freeze the right back foot of SD rats, and investigated the effects of this drug. Hematoxylin-eosin (HE) examination indicated most of pannus in the skin tissue of chilblain rats had disappeared, and the local inflammatory cells were also greatly reduced when given nifedipine at 15.0 mg/kg/d. The enzyme-linked immunosorbent assay (ELISA) revealed that nifedipine inhibited release of inflammatory factors TNF-α, IL-6, IL-1β and VEGF in serum. The RT-PCR analysis showed that nifedipine down regulated mRNA levels of TRPC-6 and VEGF in skin tissue. Furthermore, immunohistochemical examination showed nifedipine inhibited expression of IL-1β, IL-6, and TNF-α inflammatory protein and further inhibited expression of TRP (transient receptor potential) family proteins TRPM-7, TRPC-1, TRPC-3 and TRPC-6 and reduced expression of VEGF in skin and relieved erythema and oedema. This study demonstrated that nifedipine as an old medicine can be new use for the treatment of chilblain by acting on TRPs family and inflammatory proteins.
Collapse
Affiliation(s)
- Yongqiang Zhou
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Helv Yan
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Ting Li
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Mei Xie
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiuya Li
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Chunli Zhao
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| |
Collapse
|
15
|
Chai T, Qiu C, Xian Z, Lu Y, Zeng Y, Li J. A narrative review of research advances in hypoxic pulmonary hypertension. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:230. [PMID: 35280399 PMCID: PMC8908157 DOI: 10.21037/atm-22-259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/18/2022] [Indexed: 11/23/2022]
Abstract
Background and Objective Hypoxic pulmonary hypertension (HPH) is a pathological syndrome characterized by pulmonary vasoconstriction and pulmonary vascular remodeling caused by hypoxia, which eventually leads to right heart failure or death. There are 2 stages of onset of HPH: hypoxic pulmonary vasoconstriction (HPV) and hypoxic pulmonary vascular remodeling (HPVR). It is an important pathophysiological link in the pathogenesis of chronic obstructive pulmonary disease (COPD) and chronic mountain sickness (CMS), and its severity is closely related to the course and prognosis of COPD and CMS. However, there is a lack of systematic review on the diagnosis, pathogenesis and treatment of HPH. The objective of this paper is to review the diagnosis, pathogenesis, treatment of HPH. Methods In this paper, the method of literature review is adopted to obtain the information about HPH. Based on the literature, comprehensive and systematic review is made. The diagnosis, pathogenesis, treatment of HPH are summarized. Key Content and Findings Right heart catheterization is the gold standard for diagnosing HPH. Hypoxia-inducible factor, oxidative stress, metal metabolism, ion channel, inflammatory cytokines, cell apoptosis and vascular factors are the main pathogenesis of HPH. The treatment of HPH includes long-term oxygen therapy, statins, prostaglandins, phosphodiesterase inhibitor and ET receptor antagonists. Conclusions Although great progress has been made in the pathophysiology and molecular biology of HPH, it is still unclear which factors play a leading role in the pathogenesis of HPH, and no breakthrough has been made in the treatment of HPH. It is believed that the specific mechanism will be revealed as the research continues, and earlier diagnosis and the development of more effective targeted drugs will be the focus of future research.
Collapse
Affiliation(s)
- Tianci Chai
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Chen Qiu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Zhihong Xian
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yongzhen Lu
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Yuwei Zeng
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Jie Li
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
16
|
Association between triglyceride-glucose index and gastric carcinogenesis: a health checkup cohort study. Gastric Cancer 2022; 25:33-41. [PMID: 34355281 DOI: 10.1007/s10120-021-01222-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND We aimed to investigate the association between the triglyceride-glucose (TyG) index and gastric carcinogenesis, including precancerous conditions such as dysplasia, atrophic gastritis, and intestinal metaplasia. METHODS Patients who received an upper endoscopic assessment at a medical center were included. The enrolled patients were divided into four categories according to their TyG index quartile (Q). To evaluate the relationship between increase of TyG index and gastric cancer, we analyzed the patients who received a health checkup twice. Moreover, receiver-operating characteristic curve analysis was used to establish cut-off value of the TyG index for gastric cancer. RESULTS Of 127,564 enrolled patients, 43,525 (34.1%) and 186 (0.1%) were diagnosed with precancerous conditions and gastric cancer, respectively. The odds ratios (ORs) of precancerous conditions given TyG index progressively increased across quartiles: using Q1 as the reference: Q2 (OR = 1.403, P < 0.001), Q3 (OR = 1.646, P < 0.001), and Q4 (OR = 1.656, P < 0.001). The ORs of gastric cancer also increased according to the quartiles: Q2 (OR = 1.619, P = 0.045), Q3 (OR = 2.180, P = 0.004), and Q4 (OR = 2.363, P = 0.001). Moreover, the increase in TyG index between baseline and follow-up tests was more significant in gastric cancer group than in control group (P = 0.001). The optimal cut-off value for predicting gastric cancer was 9.73. CONCLUSIONS The TyG index may be a novel predictive biomarker for gastric carcinogenesis. Notably, increase in the TyG index is significantly associated with gastric cancer.
Collapse
|
17
|
Effect of Dihydropyridine Calcium Channel Blocker on Mortality of Hypertension Patients With Moderate-Severe Pulmonary Acute Respiratory Distress Syndrome: A Multicenter Retrospective Observational Cohort Study. Crit Care Explor 2021; 3:e0506. [PMID: 34514419 PMCID: PMC8425825 DOI: 10.1097/cce.0000000000000506] [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] [Indexed: 11/25/2022] Open
Abstract
The aim was to evaluate the effect of dihydropyridine calcium channel blocker on the prognosis for moderate-severe pulmonary acute respiratory distress syndrome in hypertension patients. DESIGN A retrospective, observational, multicenter cohort study. SETTING A total of 307 patients without propensity score matching and 186 adult inpatients with propensity score matching diagnosed with hypertension and moderate-severe pulmonary acute respiratory distress syndrome in five teaching hospitals in Jiangsu province, China, from December 2015 to December 2020 were enrolled. PATIENTS A total of 307 patients without propensity score matching and 186 patients with propensity score matching diagnosed with hypertension and moderate-severe pulmonary acute respiratory distress syndrome were included in the final analysis. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Demographic characteristics and clinical characteristics were recorded. The propensity score matching method was used to eliminate the difference between group with dihydropyridine calcium channel blocker and group without dihydropyridine calcium channel blocker. The primary outcome was in-hospital mortality. We used univariate and multivariate regression analyses for both patients with or without propensity score matching to assess the effect of these variables on mortality. In the subset of 186 patients with propensity score matching, in-hospital mortality was 53.2%. Inpatient mortality was significantly higher in patients treated with dihydropyridine calcium channel blocker than in those not treated with dihydropyridine calcium channel blocker of patients without propensity score matching (65.4% vs 40.4%; p < 0.01). Multivariate analysis for patients without propensity score matching showed that dihydropyridine calcium channel blocker (hazard ratio, 1.954; 95% CI, 1.415-2.699), lactate dehydrogenase greater than or equal to 600 U/L (hazard ratio, 3.809; 95% CI, 2.106-4.531), and lactate greater than or equal to 2 mmol/L (hazard ratio, 1.454; 95% CI, 1.041-2.029) were independently associated with in-hospital mortality. Based on univariate analysis for patients with propensity score matching, dihydropyridine calcium channel blocker (hazard ratio, 2.021; 95% CI, 1.333-3.064), lactate dehydrogenase greater than or equal to 600 U/L (hazard ratio, 4.379; 95% CI, 2.642-7.257), and lactate greater than or equal to 2 mmol/L (hazard ratio, 2.461; 95% CI, 1.534-3.951) were independently associated with in-hospital mortality. In contrast, patients not treated with dihydropyridine calcium channel blocker had a significant survival advantage over those treated with dihydropyridine calcium channel blocker in both patients without or with propensity score matching (p < 0.001; p = 0.001 by Kaplan-Meier analysis). CONCLUSIONS Dihydropyridine calcium channel blocker, lactate dehydrogenase greater than or equal to 600 U/L, and lactate greater than or equal to 2 mmol/L at admission were independent risk factors for patients with hypertension and moderate-severe pulmonary acute respiratory distress syndrome.
Collapse
|
18
|
Hong W, Mo Q, Wang L, Peng F, Zhou Y, Zou W, Sun R, Liang C, Zheng M, Li H, Zhao D, Gao M, Pu J, Li B, Ran P, Peng G. Changes in the gut microbiome and metabolome in a rat model of pulmonary arterial hypertension. Bioengineered 2021; 12:5173-5183. [PMID: 34405758 PMCID: PMC8806624 DOI: 10.1080/21655979.2021.1952365] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The gut microbiota is widely considered to be involved in several diseases, including atherosclerosis, obesity, chronic obstructive pulmonary disease (COPD) and pulmonary arterial hypertension (PAH). This study aimed to determine if changes in the gut microbiome and metabolome play a major role in the early pathogenesis of PAH. Male Wistar rats were injected with monocrotaline (MCT) (55 mg/kg) at day 1 and injected with calcium-sensing receptor (CaSR) antagonist NPS2143 (4.5 mg/kg/d) from days 1 to 21. Fecal samples were obtained. The gut microbiota and metabolome were analyzed by 16S rRNA gene sequencing and mass spectrometry-based analysis to investigate the effect of PAH in this rat model. MCT injection had a marked effect on the composition of the gut microbiota. This finding was further confirmed by metabolomic analysis with identification of several metabolites relevant to the gut microflora. However, NPS2143 partially abrogated this intestinal flora disorder and reversed fecal metabolite abnormalities. In conclusion, our study shows correlations between changes in the gut microbiome and the metabolome in PAH, which are affected by NPS2143.
Collapse
Affiliation(s)
- Wei Hong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiudi Mo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,Department of Respiratory, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Luyao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Fang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China.,Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Yuming Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Weifeng Zou
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, China
| | - Ruiting Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Chunxiao Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Mengning Zheng
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Haiqing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Dongxing Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Mi Gao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jinding Pu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| | - Gongyong Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Romagnoli C, Sharma P, Zonefrati R, Palmini G, Lucattelli E, Ward DT, Ellinger I, Innocenti M, Brandi ML. Study of the Expression and Function of Calcium-Sensing Receptor in Human Skeletal Muscle. Int J Mol Sci 2021; 22:ijms22147282. [PMID: 34298895 PMCID: PMC8304165 DOI: 10.3390/ijms22147282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 01/20/2023] Open
Abstract
Skeletal muscle has an outstanding capacity for regeneration in response to injuries, but there are disorders in which this process is seriously impaired, such as sarcopenia. Pharmacological treatments to restore muscle trophism are not available, therefore, the identification of suitable therapeutic targets that could be useful for the treatment of skeletal reduced myogenesis is highly desirable. In this in vitro study, we explored the expression and function of the calcium-sensing receptor (CaSR) in human skeletal muscle tissues and their derived satellite cells. The results obtained from analyses with various techniques of gene and protein CaSR expression and of its secondary messengers in response to calcium (Ca2+) and CaSR drugs have demonstrated that this receptor is not present in human skeletal muscle tissues, neither in the established satellite cells, nor during in vitro myogenic differentiation. Taken together, our data suggest that, although CaSR is a very important drug target in physiology and pathology, this receptor probably does not have any physiological role in skeletal muscle in normal conditions.
Collapse
Affiliation(s)
- Cecilia Romagnoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (C.R.); (P.S.); (R.Z.); (G.P.)
| | - Preeti Sharma
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (C.R.); (P.S.); (R.Z.); (G.P.)
| | - Roberto Zonefrati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (C.R.); (P.S.); (R.Z.); (G.P.)
- Fondazione Italiana Ricerca sulla Malattie dell’Osso (FIRMO Onlus), 50141 Florence, Italy
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (C.R.); (P.S.); (R.Z.); (G.P.)
| | - Elena Lucattelli
- Plastic and Reconstructive Microsurgery, Careggi University Hospital, 50139 Florence, Italy; (E.L.); (M.I.)
| | - Donald T. Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| | - Isabella Ellinger
- Institute for Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Marco Innocenti
- Plastic and Reconstructive Microsurgery, Careggi University Hospital, 50139 Florence, Italy; (E.L.); (M.I.)
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy; (C.R.); (P.S.); (R.Z.); (G.P.)
- Fondazione Italiana Ricerca sulla Malattie dell’Osso (FIRMO Onlus), 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
20
|
Liao J, Lu W, Chen Y, Duan X, Zhang C, Luo X, Lin Z, Chen J, Liu S, Yan H, Chen Y, Feng H, Zhou D, Chen X, Zhang Z, Yang Q, Liu X, Tang H, Li J, Makino A, Yuan JXJ, Zhong N, Yang K, Wang J. Upregulation of Piezo1 (Piezo Type Mechanosensitive Ion Channel Component 1) Enhances the Intracellular Free Calcium in Pulmonary Arterial Smooth Muscle Cells From Idiopathic Pulmonary Arterial Hypertension Patients. Hypertension 2021; 77:1974-1989. [PMID: 33813851 DOI: 10.1161/hypertensionaha.120.16629] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jing Liao
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yuqin 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xin Duan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (X.D.)
| | - Chenting Zhang
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xiaoyun Luo
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Ziying Lin
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jiyuan 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Yilin 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Dansha Zhou
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xu 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Zizhou Zhang
- 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Qifeng 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Xinyi 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - Jing Li
- Lingnan Medical Research Center, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, China (J. Li)
| | - Ayako Makino
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.)
| | - 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, China (J. Liao, W.L., Yuqin Chen, C.Z., X. Luo, Z.L., J.C., S.L., H.Y., Yilin Chen, H.F., D.Z., X.C., Z.Z., Q.Y., X. Liu, H.T., N.Z., K.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.C., A.M., J.X.-J.Y., J.W.)
| |
Collapse
|
21
|
Roesler AM, Ravix J, Bartman CM, Patel BS, Schiliro M, Roos B, Nesbitt L, Pabelick CM, Martin RJ, MacFarlane PM, Prakash YS. Calcium-Sensing Receptor Contributes to Hyperoxia Effects on Human Fetal Airway Smooth Muscle. Front Physiol 2021; 12:585895. [PMID: 33790802 PMCID: PMC8006428 DOI: 10.3389/fphys.2021.585895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Supplemental O2 (hyperoxia), necessary for maintenance of oxygenation in premature infants, contributes to neonatal and pediatric airway diseases including asthma. Airway smooth muscle (ASM) is a key resident cell type, responding to hyperoxia with increased contractility and remodeling [proliferation, extracellular matrix (ECM) production], making the mechanisms underlying hyperoxia effects on ASM significant. Recognizing that fetal lungs experience a higher extracellular Ca2+ ([Ca2+]o) environment, we previously reported that the calcium sensing receptor (CaSR) is expressed and functional in human fetal ASM (fASM). In this study, using fASM cells from 18 to 22 week human fetal lungs, we tested the hypothesis that CaSR contributes to hyperoxia effects on developing ASM. Moderate hyperoxia (50% O2) increased fASM CaSR expression. Fluorescence [Ca2+]i imaging showed hyperoxia increased [Ca2+]i responses to histamine that was more sensitive to altered [Ca2+]o, and promoted IP3 induced intracellular Ca2+ release and store-operated Ca2+ entry: effects blunted by the calcilytic NPS2143. Hyperoxia did not significantly increase mitochondrial calcium which was regulated by CaSR irrespective of oxygen levels. Separately, fASM cell proliferation and ECM deposition (collagens but not fibronectin) showed sensitivity to [Ca2+]o that was enhanced by hyperoxia, but blunted by NPS2143. Effects of hyperoxia involved p42/44 ERK via CaSR and HIF1α. These results demonstrate functional CaSR in developing ASM that contributes to hyperoxia-induced contractility and remodeling that may be relevant to perinatal airway disease.
Collapse
Affiliation(s)
- Anne M Roesler
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jovanka Ravix
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Colleen M Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Brijeshkumar S Patel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Marta Schiliro
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Benjamin Roos
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Lisa Nesbitt
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States.,Department Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Richard J Martin
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Peter M MacFarlane
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States.,Department Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Liu R, Xu J, Jiang Y, Hong W, Li S, Fu Z, Cao W, Li B, Ran P, Peng G. Platelet-derived growth factor-BB induces pulmonary venous smooth muscle cells proliferation by upregulating calcium sensing receptor under hypoxic conditions. Cytotechnology 2021; 73:189-201. [PMID: 33927476 DOI: 10.1007/s10616-021-00456-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 02/04/2021] [Indexed: 10/22/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling, which exists in both pulmonary arteries and pulmonary veins. Pulmonary vascular remodeling stems from excessive proliferation of pulmonary vascular myocytes. Platelet-derived growth factor-BB (PDGF-BB) is a vital vascular regulator whose level increases in PH human lungs. Although the mechanisms by which pulmonary arterial smooth muscle cells respond to PDGF-BB have been studied extensively, the effects of PDGF-BB on pulmonary venous smooth muscle cells (PVSMCs) remain unknown. We herein examined the involvement of calcium sensing receptor (CaSR) in PDGF-BB-induced PVSMCs proliferation under hypoxic conditions. In PVSMCs isolated from rat intrapulmonary veins, PDGF-BB increased the cell number and DNA synthesis under normoxic and hypoxic conditions, which was accompanied by upregulated CaSR expression. The influences of PDGF-BB on proliferation and CaSR expression in hypoxic PVSMCs were greater than that in normoxic PVSMCs. In hypoxic PVSMCs superfused with Ca2+-free solution, restoration of extracellular Ca2+ induced an increase of [Ca2+]i, which was significantly smaller than that in PDGF-BB-treated hypoxic PVSMCs. The positive CaSR modulator spermine enhanced, whereas the negative CaSR modulator NPS2143 attenuated, the extracellular Ca2+-induced [Ca2+]i increase in PDGF-BB-treated hypoxic PVSMCs. Furthermore, the spermine enhanced, whereas the NPS2143 inhibited, PDGF-BB-induced proliferation in hypoxic PVSMCs. Silencing CaSR with siRNA attenuated the extracellular Ca2+-induced [Ca2+]i increase in PDGF-BB-treated hypoxic PVSMCs and inhibited PDGF-BB-induced proliferation in hypoxic PVSMCs. In conclusion, these results demonstrated that CaSR mediating PDGF-BB-induced excessive PVSMCs proliferation is an important mechanism involved in the initiation and progression of PVSMCs proliferation under hypoxic conditions.
Collapse
Affiliation(s)
- Rongmin Liu
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Juan Xu
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Yongliang Jiang
- Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, China
| | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shaoxing Li
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Zhenli Fu
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Weitao Cao
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Bing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Pixin Ran
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| | - Gongyong Peng
- Guangzhou Institute for Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510120 China
| |
Collapse
|
23
|
mTOR Signaling in Pulmonary Vascular Disease: Pathogenic Role and Therapeutic Target. Int J Mol Sci 2021; 22:ijms22042144. [PMID: 33670032 PMCID: PMC7926633 DOI: 10.3390/ijms22042144] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease without a cure. The exact pathogenic mechanisms of PAH are complex and poorly understood, yet a number of abnormally expressed genes and regulatory pathways contribute to sustained vasoconstriction and vascular remodeling of the distal pulmonary arteries. Mammalian target of rapamycin (mTOR) is one of the major signaling pathways implicated in regulating cell proliferation, migration, differentiation, and protein synthesis. Here we will describe the canonical mTOR pathway, structural and functional differences between mTOR complexes 1 and 2, as well as the crosstalk with other important signaling cascades in the development of PAH. The pathogenic role of mTOR in pulmonary vascular remodeling and sustained vasoconstriction due to its contribution to proliferation, migration, phenotypic transition, and gene regulation in pulmonary artery smooth muscle and endothelial cells will be discussed. Despite the progress in our elucidation of the etiology and pathogenesis of PAH over the two last decades, there is a lack of effective therapeutic agents to treat PAH patients representing a significant unmet clinical need. In this review, we will explore the possibility and therapeutic potential to use inhibitors of mTOR signaling cascade to treat PAH.
Collapse
|
24
|
The transmembrane peptide DWORF activates SERCA2a via dual mechanisms. J Biol Chem 2021; 296:100412. [PMID: 33581112 PMCID: PMC7988493 DOI: 10.1016/j.jbc.2021.100412] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/01/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The Ca-ATPase isoform 2a (SERCA2a) pumps cytosolic Ca2+ into the sarcoplasmic reticulum (SR) of cardiac myocytes, enabling muscle relaxation during diastole. Abnormally high cytosolic [Ca2+] is a central factor in heart failure, suggesting that augmentation of SERCA2a Ca2+ transport activity could be a promising therapeutic approach. SERCA2a is inhibited by the protein phospholamban (PLB), and a novel transmembrane peptide, dwarf open reading frame (DWORF), is proposed to enhance SR Ca2+ uptake and myocyte contractility by displacing PLB from binding to SERCA2a. However, establishing DWORF’s precise physiological role requires further investigation. In the present study, we developed cell-based FRET biosensor systems that can report on protein–protein interactions and structural changes in SERCA2a complexes with PLB and/or DWORF. To test the hypothesis that DWORF competes with PLB to occupy the SERCA2a-binding site, we transiently transfected DWORF into a stable HEK cell line expressing SERCA2a labeled with a FRET donor and PLB labeled with a FRET acceptor. We observed a significant decrease in FRET efficiency, consistent with a decrease in the fraction of SERCA2a bound to PLB. Surprisingly, we also found that DWORF also activates SERCA’s enzymatic activity directly in the absence of PLB at subsaturating calcium levels. Using site-directed mutagenesis, we generated DWORF variants that do not activate SERCA, thus identifying residues P15 and W22 as necessary for functional SERCA2a–DWORF interactions. This work advances our mechanistic understanding of the regulation of SERCA2a by small transmembrane proteins and sets the stage for future therapeutic development in heart failure research.
Collapse
|
25
|
Zhou MY, Cheng L, Chen L, Gu YJ, Wang Y. Calcium-sensing receptor in the development and treatment of pulmonary hypertension. Mol Biol Rep 2021; 48:975-981. [PMID: 33394231 DOI: 10.1007/s11033-020-06065-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 12/03/2020] [Indexed: 12/22/2022]
Abstract
Calcium-sensing receptor (CaSR) is widely involved in the cell proliferation, differentiation, migration, adhesion and apoptosis, which can affect the vascular remodeling in the humanbody. The main ligand of CaSR is extracellular Ca2+. CaSR has the physiological significance in Ca2+ homeostasis. Pulmonary vascular remodeling is one of the main histopathological changes of pulmonary hypertension (PH). The abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) results in the pulmonary vascular remodeling. CaSR is an important regulator of [Ca2+]i. [Ca2+]i is the main cause of the excessive pulmonary vascular remodeling in patients with PH. In this review, it was conclued that the structure of CaSR was prone to explore the devolopment or the treatment of PH. It was found that the regulation of CaSR with some miRNA could inhibit the proliferation of PASMCs, and that CaSR could affect the occurrence of autophagy in PH. Therefore, CaSR would become a new therapeutic target to PH.
Collapse
MESH Headings
- Adamantane/analogs & derivatives
- Adamantane/therapeutic use
- Animals
- Autophagy/drug effects
- Autophagy/genetics
- Calcium/metabolism
- Calcium Channel Blockers/therapeutic use
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Quinoxalines/therapeutic use
- Receptors, Calcium-Sensing/antagonists & inhibitors
- Receptors, Calcium-Sensing/genetics
- Receptors, Calcium-Sensing/metabolism
- Signal Transduction
- Vascular Remodeling/drug effects
- Vascular Remodeling/genetics
Collapse
Affiliation(s)
- Ming-Yuan Zhou
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Lin Cheng
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Lei Chen
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Ying-Jian Gu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning, 110122, People's Republic of China.
| |
Collapse
|
26
|
Wang R, Chen J, Ding F, Zhang L, Wu X, Wan Y, Hu J, Zhang X, Wu Q. Renal tubular injury induced by glyphosate combined with hard water: the role of cytosolic phospholipase A2. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:130. [PMID: 33569432 PMCID: PMC7867956 DOI: 10.21037/atm-20-7739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background The combined effects of glyphosate and hard water on chronic kidney disease of unknown etiology (CDKu) have attracted much interest, but the mechanisms remain unknown. Cytoplasmic phospholipase A2 (cPLA2) plays a key role in the acute and chronic inflammatory reactions. This study explored the effect of glyphosate combined with hard water on renal tubules and the possible targets and mechanisms involved. Methods In vivo experiments were conducted to investigate the synergistic effects and potential mechanisms of glyphosate and hard water on renal tubular injury in mice. Results Administration of glyphosate in mice resulted in elevated levels of β2-microglobulin (β2-MG), albumin (ALB), and serum creatinine (SCr) compared to control mice. This increase was more pronounce when glyphosate was combined with hard water. In the glyphosate-treated mice, small areas of the kidney revealed fibroblast proliferation and vacuolar degeneration, particularly at the higher dose of 400 mg/kg glyphosate. However, the combination of glyphosate and hard water induced an even greater degree of pathological changes in the kidney. Immunofluorescence and western blot analyses showed that glyphosate and hard water had a coordinated effect on calcium ions (Ca2+)-activated phospholipase A2 and the activation may play a key role in inflammation and renal tubular injury. Exposure to glyphosate alone or glyphosate plus hard water increased the levels of oxidative stress markers and inflammatory biomarkers, namely, thromboxane A2 (TX-A2), leukotriene B4 (LTB4), prostaglandin E2 (PGE2), nitric oxide synthase (NOS), and nitric oxide (NO). Parameters of oxidative stress, including the levels of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were decreased. Further analysis showed that the levels of these biomarkers were significantly different between the mice treated with glyphosate plus hard water and the mice treated with glyphosate alone. Conclusions These findings suggested that hard water combined with glyphosate can induce renal tubular injury in mice, and this may involve mitogen-activated protein kinases (MAPK)/cytosolic phospholipase A2 (cPLA2)/arachidonic acid (AA) and its downstream factors.
Collapse
Affiliation(s)
- Ruojing Wang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fan Ding
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Lin Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Xuan Wu
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Jianying Hu
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Xiaoyan Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Wu
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Yang Y, Yin L, Zhu M, Song S, Sun C, Han X, Xu Y, Zhao Y, Qi Y, Xu L, Peng JY. Protective effects of dioscin on vascular remodeling in pulmonary arterial hypertension via adjusting GRB2/ERK/PI3K-AKT signal. Biomed Pharmacother 2021; 133:111056. [PMID: 33378960 DOI: 10.1016/j.biopha.2020.111056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/05/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and lethal cardiopulmonary. Pulmonary vascular remodeling (PVR) caused by excessive proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs) is the chief pathological feature of PAH. Dioscin is a natural product that possesses multiple pharmacological activities, but its effect on PAH remains unclear. In this study, effect of dioscin on vascular remodeling in PAH was assessed in hypoxia-induced PASMCs, hypoxia-induced and monocrotaline (MCT)-induced rats. Western blot, Real-time PCR and siRNA transfection tests were applied to evaluate the possible mechanisms of dioscin. In vitro experiments, results showed dioscin markedly inhibited the proliferation and migration, and promoted apoptosis of hypoxic PASMCs. In vivo, dioscin significantly decreased the right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI), and improved pulmonary vascular stenosis in rats induced by hypoxia or MCT. Molecular mechanism studies showed that dioscin significantly reduced the expression of growth factor receptor-bound protein 2 (GRB2). Subsequently, dioscin reduced the expressions of Ras, Cyclin D1, CDK4, c-Fos, PCNA and p-ERK to inhibit proliferation and migration of PASMCs, inhibited p-PI3K and p-AKT levels and increased Bax/Bcl2 ratio to promote cell apoptosis. GRB2 siRNA transfection in PASMCs further confirmed that the inhibitory action of dioscin in PAH was evoked by adjusting GRB2/ERK/PI3K-AKT signal. Taken together, our study indicated that dioscin attenuates PAH through adjusting GRB2/ERK/PI3K-AKT signal to inhibit PASMCs proliferation and migration, and promote apoptosis, and dioscin may be developed as a therapeutic strategy for treating PAH in the future.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Diosgenin/analogs & derivatives
- Diosgenin/pharmacology
- Disease Models, Animal
- Extracellular Signal-Regulated MAP Kinases/metabolism
- GRB2 Adaptor Protein/genetics
- GRB2 Adaptor Protein/metabolism
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/enzymology
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Signal Transduction
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Yueyue Yang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Manning Zhu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Changjie Sun
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing 100081, China.
| | - J-Y Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning Province, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
28
|
Wang RX, He RL, Jiao HX, Zhang RT, Guo JY, Liu XR, Gui LX, Lin MJ, Wu ZJ. Preventive treatment with ginsenoside Rb1 ameliorates monocrotaline-induced pulmonary arterial hypertension in rats and involves store-operated calcium entry inhibition. PHARMACEUTICAL BIOLOGY 2020; 58:1055-1063. [PMID: 33096951 PMCID: PMC7592893 DOI: 10.1080/13880209.2020.1831026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
CONTEXT Ginsenoside Rb1, the main active ingredient of ginseng, exhibits ex vivo depression of store-operated calcium entry (SOCE) and related vasoconstriction in pulmonary arteries derived from pulmonary hypertension (PH) rats. However, the in vivo effects of ginsenoside Rb1 on PH remain unclear. OBJECTIVE This study explored the possibility of using ginsenoside Rb1 as an in vivo preventive medication for type I PH, i.e., pulmonary arterial hypertension (PAH), and potential mechanisms involving SOCE. MATERIALS AND METHODS Male Sprague-Dawley rats (170-180 g) were randomly divided into Control, MCT, and MCT + Rb1 groups (n = 20). Control rats received only saline injection. Rats in the MCT + Rb1 and MCT groups were intraperitoneally administered single doses of 50 mg/kg monocrotaline (MCT) combined with 30 mg/kg/day ginsenoside Rb1 or equivalent volumes of saline for 21 consecutive days. Subsequently, comprehensive parameters related to SOCE, vascular tone, histological changes and hemodynamics were measured. RESULTS Ginsenoside Rb1 reduced MCT-induced STIM1, TRPC1, and TRPC4 expression by 35.00, 31.96, and 32.24%, respectively, at the protein level. SOCE-related calcium entry and pulmonary artery contraction decreased by 162.6 nM and 71.72%. The mean pulmonary artery pressure, right ventricle systolic pressure, and right ventricular mass index decreased by 19.5 mmHg, 21.6 mmHg, and 39.50%. The wall thickness/radius ratios decreased by 14.67 and 17.65%, and the lumen area/total area ratios increased by 18.55 and 15.60% in intrapulmonary vessels with 51-100 and 101-150 μm o.d. CONCLUSION Ginsenoside Rb1, a promising candidate for PH prevention, inhibited SOCE and related pulmonary vasoconstriction, and relieved MCT-induced PAH in rats.
Collapse
Affiliation(s)
- Rui-Xing Wang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Rui-Lan He
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Hai-Xia Jiao
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Run-Tian Zhang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Jing-Yi Guo
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Xiao-Ru Liu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Biochemistry and Molecular biology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Long-Xin Gui
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Mo-Jun Lin
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Mo-Jun Lin Department of Physiology and Pathophysiology, Fujian Medical University, 1 Xueyuan Road, Shangjie Zhen, Minhou County, Fuzhou350108, P.R. China
| | - Zhi-Juan Wu
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, People’s Republic of China
- CONTACT Zhi-Juan Wu Department of Physiology and Pathophysiology, Fujian Medical University, 1 Xueyuan Road, Shangjie Zhen, Minhou County, Fuzhou350108, P.R. China
| |
Collapse
|
29
|
Jain PP, Hosokawa S, Xiong M, Babicheva A, Zhao T, Rodriguez M, Rahimi S, Pourhashemi K, Balistrieri F, Lai N, Malhotra A, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Yuan JXJ. Revisiting the mechanism of hypoxic pulmonary vasoconstriction using isolated perfused/ventilated mouse lung. Pulm Circ 2020; 10:2045894020956592. [PMID: 33282184 PMCID: PMC7691930 DOI: 10.1177/2045894020956592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxic Pulmonary Vasoconstriction (HPV) is an important physiological mechanism of the lungs that matches perfusion to ventilation thus maximizing O2 saturation of the venous blood within the lungs. This study emphasizes on principal pathways in the initiation and modulation of hypoxic pulmonary vasoconstriction with a primary focus on the role of Ca2+ signaling and Ca2+ influx pathways in hypoxic pulmonary vasoconstriction. We used an ex vivo model, isolated perfused/ventilated mouse lung to evaluate hypoxic pulmonary vasoconstriction. Alveolar hypoxia (utilizing a mini ventilator) rapidly and reversibly increased pulmonary arterial pressure due to hypoxic pulmonary vasoconstriction in the isolated perfused/ventilated lung. By applying specific inhibitors for different membrane receptors and ion channels through intrapulmonary perfusion solution in isolated lung, we were able to define the targeted receptors and channels that regulate hypoxic pulmonary vasoconstriction. We show that extracellular Ca2+ or Ca2+ influx through various Ca2+-permeable channels in the plasma membrane is required for hypoxic pulmonary vasoconstriction. Removal of extracellular Ca2+ abolished hypoxic pulmonary vasoconstriction, while blockade of L-type voltage-dependent Ca2+ channels (with nifedipine), non-selective cation channels (with 30 µM SKF-96365), and TRPC6/TRPV1 channels (with 1 µM SAR-7334 and 30 µM capsazepine, respectively) significantly and reversibly inhibited hypoxic pulmonary vasoconstriction. Furthermore, blockers of Ca2+-sensing receptors (by 30 µM NPS2143, an allosteric Ca2+-sensing receptors inhibitor) and Notch (by 30 µM DAPT, a γ-secretase inhibitor) also attenuated hypoxic pulmonary vasoconstriction. These data indicate that Ca2+ influx in pulmonary arterial smooth muscle cells through voltage-dependent, receptor-operated, and store-operated Ca2+ entry pathways all contribute to initiation of hypoxic pulmonary vasoconstriction. The extracellular Ca2+-mediated activation of Ca2+-sensing receptors and the cell-cell interaction via Notch ligands and receptors contribute to the regulation of hypoxic pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Pritesh P. Jain
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Susumu Hosokawa
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Pediatrics, Tokyo Medical
and Dental University, Tokyo, Japan
| | - Mingmei Xiong
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
- Department of Critical Medicine, The
Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Tengteng Zhao
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Marisela Rodriguez
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Shamin Rahimi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Kiana Pourhashemi
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Francesca Balistrieri
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Ning Lai
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - Atul Malhotra
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| | - John Y.-J. Shyy
- Division of Cardiovascular Medicine,
Department of Medicine, University of California, San Diego, USA
| | | | | | - Ayako Makino
- Division of Endocrinology and
Metabolism, University of California, San Diego, CA, USA
| | - Jason X.-J. Yuan
- Section of Physiology, Division of
Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego,
CA, USA
| |
Collapse
|
30
|
Liu JJ, Tang MM, Zhu ML, Xie CX, Kang PF, Ling X, Zhang H, Wang XJ, Tang B. Docosahexaenoic acid inhibits Ca 2+ influx and downregulates CaSR by upregulating microRNA-16 in pulmonary artery smooth muscle cells. J Biochem Mol Toxicol 2020; 34:e22573. [PMID: 32659049 DOI: 10.1002/jbt.22573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/25/2020] [Accepted: 06/23/2020] [Indexed: 11/10/2022]
Abstract
Docosahexaenoic acid (DHA) is reported to have the potential to ameliorate pulmonary arterial hypertension (PAH), while the specific mechanism is still obscure. This study aims to investigate the function of DHA in pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism. In our study, DHA was used to incubate PASMCs. Cytosolic-free Ca2+ concentration ([Ca2+ ]cyt) was measured using Fluo-3 AM method. Real-time polymerase chain reaction was used to detect microRNA-16 (miR-16) and calcium-sensing receptor (CaSR) messenger RNA expression levels. CCK-8 assay, BrdU assay, and Transwell assay were employed to detect the effects of DHA on proliferation and migration of PASMCs. CaSR was confirmed as a direct target of miR-16 using dual-luciferase assay, polymerase chain reaction, and Western blot analysis. It was found that DHA significantly inhibited PASMC proliferation and migration and decreased [Ca2+ ]cyt. After transfection of miR-16 mimics, proliferation and migration ability of PASMCs were significantly inhibited, whereas opposite effects were observed after miR-16 inhibition. [Ca2+ ]cyt was also inhibited by miR-16 transfection. DHA then promoted the expression of miR-16, and the effects of DHA on PASMCs were annulled when miR-16 was inhibited. CaSR was identified as a direct target of miR-16. CaSR was inhibited directly by miR-16 and indirectly by DHA. In conclusion, DHA inhibits the proliferation and migration of PASMCs, and probably ameliorates PAH via regulating miR-16/CaSR axis.
Collapse
Affiliation(s)
- Jin-Jun Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Ming-Ming Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Ming-Li Zhu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Cai-Xia Xie
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Pin-Fang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Xuan Ling
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Heng Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Xiao-Jing Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Bi Tang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| |
Collapse
|
31
|
Patel BS, Ravix J, Pabelick C, Prakash YS. Class C GPCRs in the airway. Curr Opin Pharmacol 2020; 51:19-28. [PMID: 32375079 DOI: 10.1016/j.coph.2020.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/17/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Understanding and targeting of GPCRs remain a critical aspect of airway pharmacology and therapeutics for diseases such as asthma or COPD. Most attention has been on the large Class A GPCRs towards improved bronchodilation and blunting of remodeling. Better known in the central or peripheral nervous system, there is increasing evidence that Class C GPCRs which include metabotropic glutamate and GABA receptors, the calcium sensing receptor, sweet/umami taste receptors and a number of orphan receptors, can contribute to airway structure and function. In this review, we will summarize current state of knowledge regarding the pharmacology of Class C GPCRs, their expression and potential functions in the airways, and the application of pharmacological agents targeting this group in the context of airway diseases.
Collapse
Affiliation(s)
- Brijeshkumar S Patel
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jovanka Ravix
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Christina Pabelick
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiologyand Perioperative Medicine, Mayo Clinic, Rochester, MN, United States; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
32
|
Lazrak A, Yu Z, Doran S, Jian MY, Creighton J, Laube M, Garantziotis S, Prakash YS, Matalon S. Upregulation of airway smooth muscle calcium-sensing receptor by low-molecular-weight hyaluronan. Am J Physiol Lung Cell Mol Physiol 2020; 318:L459-L471. [PMID: 31913654 PMCID: PMC7099432 DOI: 10.1152/ajplung.00429.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/19/2022] Open
Abstract
We investigated the mechanisms involved in the development of airway hyperresponsiveness (AHR) following exposure of mice to halogens. Male mice (C57BL/6; 20-25 g) exposed to either bromine (Br2) or Cl2 (600 or 400 ppm, respectively, for 30 min) developed AHR 24 h after exposure. Nifedipine (5 mg/kg body wt; an L-type calcium channel blocker), administered subcutaneously after Br2 or Cl2 exposure, produced higher AHR compared with Br2 or Cl2 alone. In contrast, diltiazem (5 mg/kg body wt; a nondihydropyridine L-type calcium channel blocker) decreased AHR to control (air) values. Exposure of immortalized human airway smooth muscle cells (hASMC) to Br2 resulted in membrane potential depolarization (Vm Air: 62 ± 3 mV; 3 h post Br2:-45 ± 5 mV; means ± 1 SE; P < 0.001), increased intracellular [Ca2+]i, and increased expression of the calcium-sensing receptor (Ca-SR) protein. Treatment of hASMC with a siRNA against Ca-SR significantly inhibited the Br2 and nifedipine-induced Vm depolarization and [Ca2+]i increase. Intranasal administration of an antagonist to Ca-SR in mice postexposure to Br2 reversed the effects of Br2 and nifedipine on AHR. Incubation of hASMC with low-molecular-weight hyaluronan (LMW-HA), generated by exposing high-molecular-weight hyaluronan (HMW-HA) to Br2, caused Vm depolarization, [Ca2+]i increase, and Ca-SR expression to a similar extent as exposure to Br2 and Cl2. The addition of HMW-HA to cells or mice exposed to Br2, Cl2, or LMW-HA reversed these effects in vitro and improved AHR in vivo. We conclude that detrimental effects of halogen exposure on AHR are mediated via activation of the Ca-SR by LMW-HA.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen Doran
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ming-Yuan Jian
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Judy Creighton
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mandy Laube
- Department of Pediatrics, Division of Neonatology, Leipzig University, Leipzig, Germany
| | - Stavros Garantziotis
- Matrix Biology Group, Immunity, Inflammation, and Disease Laboratory, National Institutes of Health/National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Y S Prakash
- Department of Physiology and Biomedical Engineering and Anesthesiology, Mayo Clinic Alix School of Medicine and Science, Rochester, Minnesota
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
33
|
Wang H, Cheng X, Tian J, Xiao Y, Tian T, Xu F, Hong X, Zhu MX. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol Ther 2020; 209:107497. [PMID: 32004513 DOI: 10.1016/j.pharmthera.2020.107497] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a group of receptor-operated calcium-permeable nonselective cation channels of the TRP superfamily. The seven mammalian TRPC members, which can be further divided into four subgroups (TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7) based on their amino acid sequences and functional similarities, contribute to a broad spectrum of cellular functions and physiological roles. Studies have revealed complexity of their regulation involving several components of the phospholipase C pathway, Gi and Go proteins, and internal Ca2+ stores. Recent advances in cryogenic electron microscopy have provided several high-resolution structures of TRPC channels. Growing evidence demonstrates the involvement of TRPC channels in diseases, particularly the link between genetic mutations of TRPC6 and familial focal segmental glomerulosclerosis. Because TRPCs were discovered by the molecular identity first, their pharmacology had lagged behind. This is rapidly changing in recent years owning to great efforts from both academia and industry. A number of potent tool compounds from both synthetic and natural products that selective target different subtypes of TRPC channels have been discovered, including some preclinical drug candidates. This review will cover recent advancements in the understanding of TRPC channel regulation, structure, and discovery of novel TRPC small molecular probes over the past few years, with the goal of facilitating drug discovery for the study of TRPCs and therapeutic development.
Collapse
Affiliation(s)
- Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaoding Cheng
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Jinbin Tian
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuling Xiao
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China
| | - Tian Tian
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Fuchun Xu
- Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China
| | - Xuechuan Hong
- State Key Laboratory of Virology, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China; Innovation Center for Traditional Tibetan Medicine Modernization and Quality Control, Medical College, Department of Chemistry and Environmental Science, School of Science, Tibet University, Lhasa 850000, China.
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
34
|
King MM, Kayastha BB, Franklin MJ, Patrauchan MA. Calcium Regulation of Bacterial Virulence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:827-855. [PMID: 31646536 DOI: 10.1007/978-3-030-12457-1_33] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+) is a universal signaling ion, whose major informational role shaped the evolution of signaling pathways, enabling cellular communications and responsiveness to both the intracellular and extracellular environments. Elaborate Ca2+ regulatory networks have been well characterized in eukaryotic cells, where Ca2+ regulates a number of essential cellular processes, ranging from cell division, transport and motility, to apoptosis and pathogenesis. However, in bacteria, the knowledge on Ca2+ signaling is still fragmentary. This is complicated by the large variability of environments that bacteria inhabit with diverse levels of Ca2+. Yet another complication arises when bacterial pathogens invade a host and become exposed to different levels of Ca2+ that (1) are tightly regulated by the host, (2) control host defenses including immune responses to bacterial infections, and (3) become impaired during diseases. The invading pathogens evolved to recognize and respond to the host Ca2+, triggering the molecular mechanisms of adhesion, biofilm formation, host cellular damage, and host-defense resistance, processes enabling the development of persistent infections. In this review, we discuss: (1) Ca2+ as a determinant of a host environment for invading bacterial pathogens, (2) the role of Ca2+ in regulating main events of host colonization and bacterial virulence, and (3) the molecular mechanisms of Ca2+ signaling in bacterial pathogens.
Collapse
Affiliation(s)
- Michelle M King
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, USA
| | - Biraj B Kayastha
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, USA
| | - Michael J Franklin
- Department of Microbiology and Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Marianna A Patrauchan
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK, USA.
| |
Collapse
|
35
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
36
|
Robinson BL, Gu Q, Tryndyak V, Ali SF, Dumas M, Kanungo J. Nifedipine toxicity is exacerbated by acetyl l-carnitine but alleviated by low-dose ketamine in zebrafish in vivo. J Appl Toxicol 2019; 40:257-269. [PMID: 31599005 DOI: 10.1002/jat.3901] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/22/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022]
Abstract
Calcium channel blocker (CCB) poisoning is a common and sometimes life-threatening emergency. Our previous studies have shown that acetyl l-carnitine (ALCAR) prevents cardiotoxicity and developmental toxicity induced by verapamil, a CCB used to treat patients with hypertension. Here, we tested whether toxicities of nifedipine, a dihydropyridine CCB used to treat hypertension, can also be mitigated by co-treatment with ALCAR. In the zebrafish embryos at three different developmental stages, nifedipine induced developmental toxicity with pericardial sac edema in a dose-dependent manner, which were surprisingly exacerbated with ALCAR co-treatment. Even with low-dose nifedipine (5 μm), when the pericardial sac looked normal, ALCAR co-treatment showed pericardial sac edema. We hypothesized that toxicity by nifedipine, a vasodilator, may be prevented by ketamine, a known vasoconstrictor. Nifedipine toxicity in the embryos was effectively prevented by co-treatment with low (subanesthetic) doses (25-100 μm added to the water) of ketamine, although a high dose of ketamine (2 mm added to the water) partially prevented the toxicity.As expected of a CCB, nifedipine either in the presence or absence of ketamine-reduced metabolic reactive oxygen species (ROS), a downstream product of calcium signaling, in the rapidly developing digestive system. However, nifedipine induced ROS in the trunk region that showed significantly stunted growth indicating that the tissues under stress potentially produced pathologic ROS. To the best of our knowledge, these studies for the first time show that nifedipine and the dietary supplement ALCAR together induce adverse effects while providing evidence on the therapeutic efficacy of subanesthetic doses of ketamine against nifedipine toxicity in vivo.
Collapse
Affiliation(s)
- Bonnie L Robinson
- Division of Neurotoxicology, US Food and Drug Administration, Jefferson, Arkansas
| | - Qiang Gu
- Division of Neurotoxicology, US Food and Drug Administration, Jefferson, Arkansas
| | - Volodymyr Tryndyak
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Syed F Ali
- Division of Neurotoxicology, US Food and Drug Administration, Jefferson, Arkansas
| | | | - Jyotshna Kanungo
- Division of Neurotoxicology, US Food and Drug Administration, Jefferson, Arkansas
| |
Collapse
|
37
|
TRPP2 associates with STIM1 to regulate cerebral vasoconstriction and enhance high salt intake-induced hypertensive cerebrovascular spasm. Hypertens Res 2019; 42:1894-1904. [PMID: 31541223 DOI: 10.1038/s41440-019-0324-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/05/2019] [Accepted: 08/14/2019] [Indexed: 11/08/2022]
Abstract
Cerebrovascular spasm is a life-threatening event in salt-sensitive hypertension. The relationship between store-operated calcium entry (SOCE) and vasoconstriction in hypertension has not been fully clarified. This study investigated the changes in cerebrovascular contractile responses in high salt intake-induced hypertension and the functional roles of the main components of SOCE, namely, polycystin-2 (TRPP2), stromal interaction molecule 1 (STIM1), and Orai3. Polycystic kidney disease 2 (which encodes TRPP2) knockout mice displayed decreased cerebrovascular SOCE-induced contraction. The blood pressure of age-matched rats fed a normal or high-salt diet for 4 weeks was monitored weekly using noninvasive tail-cuff plethysmography. The systolic blood pressure of the rats fed a high-salt diet was significantly higher than that of controls. Western blotting and immunohistochemical results showed that these hypertensive rats expressed higher levels of cerebrovascular TRPP2, STIM1, and Orai3 than controls. Cerebrovascular tension measurements of the basilar artery indicated that SOCE-mediated contraction was significantly increased in hypertensive rats compared with control rats. In addition, SOCE-mediated contraction was decreased in the basilar arteries of rats pretreated with the SOCE inhibitor BTP-2 (10 μM) or transfected with TRPP2-specific or STIM1-specific small interfering RNA. Staining with 2,3,5-triphenyltetrazolium chloride (TTC) was used to quantify the infarcted brain area 24 h after middle cerebral artery occlusion, a model of ischemic stroke, in rodents. The infarcted brain area was significantly greater in hypertensive rats and significantly lower in BTP-2-treated rats than in controls. Taken together, these findings indicate that SOCE-induced contraction may be overactive in the basilar arteries of salt-sensitive hypertensive rats, suggesting the dysregulation of TRPP2 and SOCE and its other components.
Collapse
|
38
|
Magnesium Sulfate Mitigates the Progression of Monocrotaline Pulmonary Hypertension in Rats. Int J Mol Sci 2019; 20:ijms20184622. [PMID: 31540416 PMCID: PMC6770589 DOI: 10.3390/ijms20184622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022] Open
Abstract
We investigated whether magnesium sulfate (MgSO4) mitigated pulmonary hypertension progression in rats. Pulmonary hypertension was induced by a single intraperitoneal injection of monocrotaline (60 mg/kg). MgSO4 (100 mg/kg) was intraperitoneally administered daily for 3 weeks, from the seventh day after monocrotaline injection. Adult male rats were randomized into monocrotaline (MCT) or monocrotaline plus MgSO4 (MM) groups (n = 15 per group); control groups were maintained simultaneously. For analysis, surviving rats were euthanized on the 28th day after receiving monocrotaline. The survival rate was higher in the MM group than in the MCT group (100% versus 73.3%, p = 0.043). Levels of pulmonary artery wall thickening, α-smooth muscle actin upregulation, right ventricular systolic pressure increase, and right ventricular hypertrophy were lower in the MM group than in the MCT group (all p < 0.05). Levels of lipid peroxidation, mitochondrial injury, inflammasomes and cytokine upregulation, and apoptosis in the lungs and right ventricle were lower in the MM group than in the MCT group (all p < 0.05). Notably, the mitigation effects of MgSO4 on pulmonary artery wall thickening and right ventricular hypertrophy were counteracted by exogenous calcium chloride. In conclusion, MgSO4 mitigates pulmonary hypertension progression, possibly by antagonizing calcium.
Collapse
|
39
|
A Combined Targeted and Whole Exome Sequencing Approach Identified Novel Candidate Genes Involved in Heritable Pulmonary Arterial Hypertension. Sci Rep 2019; 9:753. [PMID: 30679663 PMCID: PMC6345742 DOI: 10.1038/s41598-018-37277-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 11/29/2018] [Indexed: 01/08/2023] Open
Abstract
The pathogenesis of idiopathic and heritable forms of pulmonary arterial hypertension is still not completely understood, even though several causative genes have been proposed, so that a third of patients remains genetically unresolved. Here we applied a multistep approach to extend identification of the genetic bases of such a disease by searching for novel candidate genes/pathways. Twenty-eight patients belonging to 18 families were screened for BMPR2 mutations and BMPR2-negative samples were tested for 12 additional candidate genes by means of a specific massive parallel sequencing-based assay. Finally, whole exome sequencing was performed on four patients showing no mutations at known disease genes, as well as on their unaffected parents. In addition to EIF2AK4, which has been already suggested to be associated with pulmonary veno-occlusive disease, we identified the novel candidate genes ATP13A3, CD248, EFCAB4B, involved in lung vascular remodeling that represent reliable drivers contributing to the disease according to their biological functions/inheritance patterns. Therefore, our results suggest that combining gene panel and whole exome sequencing provides new insights useful for the genetic diagnosis of familial and idiopathic pulmonary arterial hypertension, as well as for the identification of biological pathways that will be potentially targeted by new therapeutic strategies.
Collapse
|
40
|
Roesler AM, Wicher SA, Ravix J, Britt RD, Manlove L, Teske JJ, Cummings K, Thompson MA, Farver C, MacFarlane P, Pabelick CM, Prakash YS. Calcium sensing receptor in developing human airway smooth muscle. J Cell Physiol 2019; 234:14187-14197. [PMID: 30624783 DOI: 10.1002/jcp.28115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022]
Abstract
Airway smooth muscle (ASM) regulation of airway structure and contractility is critical in fetal/neonatal physiology in health and disease. Fetal lungs experience higher Ca2+ environment that may impact extracellular Ca2+ ([Ca2+ ]o ) sensing receptor (CaSR). Well-known in the parathyroid gland, CaSR is also expressed in late embryonic lung mesenchyme. Using cells from 18-22 week human fetal lungs, we tested the hypothesis that CaSR regulates intracellular Ca2+ ([Ca2+ ]i ) in fetal ASM (fASM). Compared with adult ASM, CaSR expression was higher in fASM, while fluorescence Ca2+ imaging showed that [Ca2+ ]i was more sensitive to altered [Ca2+ ]o . The fASM [Ca2+ ]i responses to histamine were also more sensitive to [Ca2+ ]o (0-2 mM) compared with an adult, enhanced by calcimimetic R568 but blunted by calcilytic NPS2143. [Ca2+ ]i was enhanced by endogenous CaSR agonist spermine (again higher sensitivity compared with adult). Inhibition of phospholipase C (U73122; siRNA) or inositol 1,4,5-triphosphate receptor (Xestospongin C) blunted [Ca2+ ]o sensitivity and R568 effects. NPS2143 potentiated U73122 effects. Store-operated Ca2+ entry was potentiated by R568. Traction force microscopy showed responsiveness of fASM cellular contractility to [Ca2+ ]o and NPS2143. Separately, fASM proliferation showed sensitivity to [Ca2+ ]o and NPS2143. These results demonstrate functional CaSR in developing ASM that modulates airway contractility and proliferation.
Collapse
Affiliation(s)
- Anne M Roesler
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sarah A Wicher
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jovanka Ravix
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Rodney D Britt
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Logan Manlove
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jacob J Teske
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Katelyn Cummings
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael A Thompson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Carol Farver
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Peter MacFarlane
- Division of Neonatology, Case Western University, Cleveland, Ohio
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
41
|
Inoue R, Kurahara LH, Hiraishi K. TRP channels in cardiac and intestinal fibrosis. Semin Cell Dev Biol 2018; 94:40-49. [PMID: 30445149 DOI: 10.1016/j.semcdb.2018.11.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023]
Abstract
It is now widely accepted that advanced fibrosis underlies many chronic inflammatory disorders and is the main cause of morbidity and mortality of the modern world. The pathogenic mechanism of advanced fibrosis involves diverse and intricate interplays between numerous extracellular and intracellular signaling molecules, among which the non-trivial roles of a stress-responsive Ca2+/Na+-permeable cation channel superfamily, the transient receptor potential (TRP) protein, are receiving growing attention. Available evidence suggests that several TRP channels such as TRPC3, TRPC6, TRPV1, TRPV3, TRPV4, TRPA1, TRPM6 and TRPM7 may play central roles in the progression and/or prevention of fibroproliferative disorders in vital visceral organs such as lung, heart, liver, kidney, and bowel as well as brain, blood vessels and skin, and may contribute to both acute and chronic inflammatory processes involved therein. This short paper overviews the current knowledge accumulated in this rapidly growing field, with particular focus on cardiac and intestinal fibrosis, which are tightly associated with the pathogenesis of atrial fibrillation and inflammatory bowel diseases such as Crohn's disease.
Collapse
Affiliation(s)
- Ryuji Inoue
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Lin-Hai Kurahara
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan
| | - Keizo Hiraishi
- Department of Physiology, Fukuoka University School of medicine, Nanakuma 7-451, Jonan-ku, Fukuoka 814-0180, Japan
| |
Collapse
|
42
|
Hua C, Zhao J, Wang H, Chen F, Meng H, Chen L, Zhang Q, Yan J, Yuan L. Apple polyphenol relieves hypoxia-induced pulmonary arterial hypertension via pulmonary endothelium protection and smooth muscle relaxation: In vivo and in vitro studies. Biomed Pharmacother 2018; 107:937-944. [DOI: 10.1016/j.biopha.2018.08.080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
|
43
|
Lambert M, Capuano V, Olschewski A, Sabourin J, Nagaraj C, Girerd B, Weatherald J, Humbert M, Antigny F. Ion Channels in Pulmonary Hypertension: A Therapeutic Interest? Int J Mol Sci 2018; 19:ijms19103162. [PMID: 30322215 PMCID: PMC6214085 DOI: 10.3390/ijms19103162] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 12/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.
Collapse
Affiliation(s)
- Mélanie Lambert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Véronique Capuano
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
- Department of Physiology, Medical University Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Jessica Sabourin
- Signalisation et Physiopathologie Cardiovasculaire, UMRS 1180, Univ. Paris-Sud, INSERM, Université Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Stiftingtalstrasse 24, Graz 8010, Austria.
| | - Barbara Girerd
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Jason Weatherald
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB T1Y 6J4, Canada.
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB T1Y 6J4, Canada.
| | - Marc Humbert
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| | - Fabrice Antigny
- Univ. Paris-Sud, Faculté de Médecine, 94270 Kremlin-Bicêtre, France.
- AP-HP, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire (DHU) Thorax Innovation, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, 94270 Le Kremlin-Bicêtre, France.
- UMRS 999, INSERM and Univ. Paris⁻Sud, Laboratoire d'Excellence (LabEx) en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Hôpital-Marie-Lannelongue, 92350 Le Plessis Robinson, France.
| |
Collapse
|
44
|
Lu M, Leng B, He X, Zhang Z, Wang H, Tang F. Calcium Sensing Receptor-Related Pathway Contributes to Cardiac Injury and the Mechanism of Astragaloside IV on Cardioprotection. Front Pharmacol 2018; 9:1163. [PMID: 30364197 PMCID: PMC6193074 DOI: 10.3389/fphar.2018.01163] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/24/2018] [Indexed: 11/29/2022] Open
Abstract
Activation of calcium sensing receptor (CaSR) contributes to cardiac injury, but the underlying mechanism has not yet been examined. Astragaloside IV (AsIV) was previously reported to exhibit protective effects against various myocardial injuries. The aim of the present study was to investigate the underlying mechanism of CaSR in cardiac hypertrophy and apoptosis and to evaluate whether the protective effect of AsIV against myocardial injury is associated with CaSR and its related signaling pathway. In vivo and in vitro myocardial injury was induced by isoproterenol (Iso) or GdCl3 (a CaSR agonist) in rats and heart H9C2 cells. Cardiac cell hypertrophy, apoptosis, function, Mitochondrial Membrane Potential (MMP), mitochondrial ultrastructure, and [Ca2+]i, as well as the protein expression of CaSR, calcium/calmodulin-dependent protein kinase II (CaMKII), calcineurin (CaN), sarcoplasmic reticulum Ca2+-ATPase2a (SERCA2a), and the inositol 1,4,5-trisphosphate receptor (IP3R), were measured in vivo and/or in vitro. The results showed that AsIV attenuated cardiac hypertrophy and apoptosis and attenuated impairments in cardiac function, mitochondrial structure, and MMP induced by Iso or GdCl3 in rat myocardial tissue and H9C2 cells. Importantly, AsIV treatment inhibited the enhancement of [Ca2+]i and CaSR expression induced by Iso or GdCl3, an effect similar to that of the CaSR antagonist NPS2143. In addition, AsIV treatment repressed CaSR, CaMKII, and CaN activation and inhibited NFAT-3 nuclear translocation. Mechanistic analysis using lentivirus infection showed that CaSR overexpression activated the CaMKII and CaN signaling pathways and that this response was enhanced by Iso. The results suggested that CaSR-mediated changes in [Ca2+]i and CaMKII and CaN signaling pathways contribute to cardiac hypertrophy and apoptosis and are involved in the protective effect of astragaloside IV against cardiac injury.
Collapse
Affiliation(s)
- Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Bin Leng
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Xin He
- Internal Medicine-Cardiovascular Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zhen Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| | - Futian Tang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
45
|
Bone Marrow-Derived Endothelial Progenitor Cells Contribute to Monocrotaline-Induced Pulmonary Arterial Hypertension in Rats via Inhibition of Store-Operated Ca 2+ Channels. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4892349. [PMID: 30320134 PMCID: PMC6167576 DOI: 10.1155/2018/4892349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/19/2018] [Indexed: 11/17/2022]
Abstract
Purpose This study aimed to explore whether bone marrow- (BM-) derived endothelial progenitor cells (EPCs) contributing to monocrotaline- (MCT-) induced pulmonary arterial hypertension (PAH) in rats via modulating store-operated Ca2+ channels (SOC). Methods Sprague Dawley (SD) rats were assigned into MCT group (n = 30) and control group (n = 20). Rats in MCT group were subcutaneously administered with 60 mg/kg MCT solution, and rats in control group were injected with equal amount of vehicle. After 3 weeks of treatment, right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) of two groups were measured, and BM-derived EPCs were isolated. Immunochemistry identification and vasculogenesis detection of EPCs were then performed. [Ca2+]cyt measurement was performed to detect store-operated calcium entry (SOCE) in two groups, followed by determination of Orai and canonical transient receptor potential (TRPC) channels expression. Results After 3 weeks of treatment, there were significant increases in RVSP and RVHI in MCT group compared with control group, indicating that MCT successfully induced PAH in rats. Moreover, the SOCE ([Ca2+]cyt rise) in BM-derived EPCs of MCT group was lower than that of control group. Furthermore, the expression levels of Orai3, TRPC1, TRPC3, and TRPC6 in BM-derived EPCs were decreased in MCT group in comparison with control group. Conclusions The SOC activities were inhibited in BM-derived EPCs of MCT-treated rats. These results may be associated with the depressed expression of Orai3, TRPC1, TRPC3, and TRPC6, which are major mediators of SOC.
Collapse
|
46
|
Strassheim D, Karoor V, Stenmark K, Verin A, Gerasimovskaya E. A current view of G protein-coupled receptor - mediated signaling in pulmonary hypertension: finding opportunities for therapeutic intervention. ACTA ACUST UNITED AC 2018; 2. [PMID: 31380505 PMCID: PMC6677404 DOI: 10.20517/2574-1209.2018.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathological vascular remodeling is observed in various cardiovascular diseases including pulmonary hypertension (PH), a disease of unknown etiology that has been characterized by pulmonary artery vasoconstriction, right ventricular hypertrophy, vascular inflammation, and abnormal angiogenesis in pulmonary circulation. G protein-coupled receptors (GPCRs) are the largest family in the genome and widely expressed in cardiovascular system. They regulate all aspects of PH pathophysiology and represent therapeutic targets. We overview GPCRs function in vasoconstriction, vasodilation, vascular inflammation-driven remodeling and describe signaling cross talk between GPCR, inflammatory cytokines, and growth factors. Overall, the goal of this review is to emphasize the importance of GPCRs as critical signal transducers and targets for drug development in PH.
Collapse
Affiliation(s)
- Derek Strassheim
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Vijaya Karoor
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.,Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
47
|
Guo Y, Yang X, He J, Liu J, Yang S, Dong H. Important roles of the Ca 2+-sensing receptor in vascular health and disease. Life Sci 2018; 209:217-227. [PMID: 30098342 DOI: 10.1016/j.lfs.2018.08.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Ca2+-sensing receptor (CaSR), a member of G protein-coupled receptor family, is widely expressed in the vascular system, including perivascular neurons, vascular endothelial cells (VECs) and vascular smooth muscle cells (VSMCs). When stimulated, CaSR can further increase the cytosolic Ca2+ concentration ([Ca2+]cyt) in two ways: intracellular Ca2+ release from endo/sarcoplasmic reticulum (ER/SR) and extracellular Ca2+ entry through Ca2+-permeable cation channels. In endothelium, increased Ca2+ subsequently activate nitric oxide synthase (NOS) and intermediate conductance Ca2+-activated K+ channels (IKCa), resulting in vasodilation through NOS-mediated NO release or membrane hyperpolarization. In VSMCs, CaSR-induced intracellular Ca2+ increase causes blood vessel constriction. CaSR activation predominantly induces vasorelaxation of whole vascular tissues through VECs-dependent mechanisms; however, CaSR-induced Ca2+ signaling in VSMCs may play a braking role in CaSR-mediated vasorelaxation. Emerging evidence reveals the importance of CaSR in the regulation of vascular tone and blood pressure. Here, we summarized recent advances in CaSR-mediated vascular reaction and the underlying mechanisms in different species, including humans. In addition, several studies have demonstrated that CaSR dysfunction may be associated with some fatal vascular diseases, such as pulmonary arterial hypertension, primary hypertension, diabetes, acute myocardial infarction and vascular calcification. With the advance of studies on CaSR in vascular health and disease, it is expected positive modulators or negative modulators of CaSR used for the treatment of specific diseases may be promising therapeutic options for the prevention and/or treatment of vascular diseases.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xin Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jialin He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jingjing Liu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
48
|
Reyes RV, Castillo-Galán S, Hernandez I, Herrera EA, Ebensperger G, Llanos AJ. Revisiting the Role of TRP, Orai, and ASIC Channels in the Pulmonary Arterial Response to Hypoxia. Front Physiol 2018; 9:486. [PMID: 29867539 PMCID: PMC5949889 DOI: 10.3389/fphys.2018.00486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary arteries are exquisitely responsive to oxygen changes. They rapidly and proportionally contract as arterial PO2 decrease, and they relax as arterial PO2 is re-established. The hypoxic pulmonary vasoconstriction (HPV) is intrinsic since it does not require neural or endocrine factors, as evidenced in isolated vessels. On the other hand, pulmonary arteries also respond to sustained hypoxia with structural and functional remodeling, involving growth of smooth muscle medial layer and later recruitment of adventitial fibroblasts, secreted mitogens from endothelium and changes in the response to vasoconstrictor and vasodilator stimuli. Hypoxic pulmonary arterial vasoconstriction and remodeling are relevant biological responses both under physiological and pathological conditions, to explain matching between ventilation and perfusion, fetal to neonatal transition of pulmonary circulation and pulmonary artery over-constriction and thickening in pulmonary hypertension. Store operated channels (SOC) and receptor operated channels (ROC) are plasma membrane cationic channels that mediate calcium influx in response to depletion of internal calcium stores or receptor activation, respectively. They are involved in both HPV and pathological remodeling since their pharmacological blockade or genetic suppression of several of the Stim, Orai, TRP, or ASIC proteins in SOC or ROC complexes attenuate the calcium increase, the tension development, the pulmonary artery smooth muscle proliferation, and pulmonary arterial hypertension. In this Mini Review, we discussed the evidence obtained in in vivo animal models, at the level of isolated organ or cells of pulmonary arteries, and we identified and discussed the questions for future research needed to validate these signaling complexes as targets against pulmonary hypertension.
Collapse
Affiliation(s)
- Roberto V Reyes
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Sebastián Castillo-Galán
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ismael Hernandez
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Germán Ebensperger
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| | - Aníbal J Llanos
- Unidad de Fisiología y Fisiopatología Perinatal, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies, Universidad de Chile, Santiago, Chile
| |
Collapse
|
49
|
Increased CaSR and TRPC6 pulmonary vascular expression in the nitrofen-induced model of congenital diaphragmatic hernia. Pediatr Surg Int 2018; 34:211-215. [PMID: 28983729 DOI: 10.1007/s00383-017-4191-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2017] [Indexed: 11/27/2022]
Abstract
AIMS AND OBJECTIVES The high morbidity and mortality rates in congenital diaphragmatic hernia (CDH) are attributed primarily to severe lung hypoplasia and/or persistent pulmonary hypertension (PPH). PPH in CDH is characterized by abnormal vascular remodeling with thickening of medial and adventitial layers and extension of smooth muscle into previously nonmuscularized arteries. Excessive proliferation of pulmonary arterial smooth muscle cells (PASMC) is an important contributor to the concentric pulmonary arterial remodeling. An increase in cytosolic-free Ca2+ concentration in PASMC is a major trigger for pulmonary vasoconstriction and a key stimulus for PASMC proliferation and migration. Calcium-sensing receptor (CaSR), a member of the G-protein coupled receptor family, is activated by cations (e.g., Ca2+, Mg2+) and polyamines. Under normal physiological conditions, the expression levels of CaSR in the pulmonary vasculature are very low. Canonical transient receptor potential channels (TRPCs) constitute a series of nonselective cation channels with variable degree of Ca2+ selectivity. TRPC6 has been reported to play a crucial role in the regulation of neo-muscularization, vasoreactivity, and vasomotor tone in the pulmonary vasculature. We hypothesized that CaSR and TRPC6 expression is upregulated in the pulmonary vasculature of nitrofen-induced CDH rats. MATERIALS AND METHODS Following ethical approval (REC1103), time-pregnant Sprague Dawley rats received nitrofen or vehicle on gestational day (D) 9. D21 fetuses were divided into CDH and control (n = 12). Quantitative real-time polymerase chain reaction (QRT-PCR), western blotting, and confocal-immunofluorescence microscopy were performed to detect lung gene and protein expression of CaSR and TRPC6. RESULTS QRT-PCR and western blot analysis revealed that CaSR and TPRC6 expression was significantly increased in the CDH group compared to controls (p < 0.05). Confocal-immunofluorescence microscopy revealed that CaSR and TRPC6 lung expression was markedly increased in CDH group compared to controls. CONCLUSION Increased CaSR and TRPC6 expression in CDH lung suggests that CaSR interacting with TRPC6 may contribute to abnormal vascular remodeling resulting in pulmonary vasoconstriction and development of PPH.
Collapse
|