1
|
Deng H, Eichmann A, Schwartz MA. Fluid Shear Stress-Regulated Vascular Remodeling: Past, Present, and Future. Arterioscler Thromb Vasc Biol 2025; 45:882-900. [PMID: 40207366 DOI: 10.1161/atvbaha.125.322557] [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] [Indexed: 04/11/2025]
Abstract
The vascular system remodels throughout life to ensure adequate perfusion of tissues as they grow, regress, or change metabolic activity. Angiogenesis, the sprouting of new blood vessels to expand the capillary network, versus regression, in which endothelial cells die or migrate away to remove unneeded capillaries, controls capillary density. In addition, upstream arteries adjust their diameters to optimize blood flow to downstream vascular beds, which is controlled primarily by vascular endothelial cells sensing fluid shear stress (FSS) from blood flow. Changes in capillary density and small artery tone lead to changes in the resistance of the vascular bed, which leads to changes in flow through the arteries that feed these small vessels. The resultant decreases or increases in FSS through these vessels then stimulate their inward or outward remodeling, respectively. This review summarizes our knowledge of endothelial FSS-dependent vascular remodeling, offering insights into potential therapeutic interventions. We first provide a historical overview, then discuss the concept of set point and mechanisms of low-FSS-mediated and high-FSS-mediated inward and outward remodeling. We then cover in vivo animal models, molecular mechanisms, and clinical implications. Understanding the mechanisms underlying physiological endothelial FSS-mediated vascular remodeling and their failure due to mutations or chronic inflammatory and metabolic stresses may lead to new therapeutic strategies to prevent or treat vascular diseases.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Anne Eichmann
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Martin A Schwartz
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale School of Medicine, New Haven, CT (M.A.S.)
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT (M.A.S.)
| |
Collapse
|
2
|
Liao W, Huang Y, Wang X, Hu Z, Zhao C, Wang G. Multidimensional excavation of the current status and trends of mechanobiology in cardiovascular homeostasis and remodeling within 20 years. MECHANOBIOLOGY IN MEDICINE 2025; 3:100127. [PMID: 40395770 PMCID: PMC12067904 DOI: 10.1016/j.mbm.2025.100127] [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: 08/20/2024] [Revised: 02/12/2025] [Accepted: 03/09/2025] [Indexed: 05/22/2025]
Abstract
Mechanobiology is essential for cardiovascular structure and function and regulates the normal physiological and pathological processes of the cardiovascular system. Cells in the cardiovascular system are extremely sensitive to their mechanical environment, and once mechanical stimulation is abnormal, the homeostasis mechanism is damaged or lost, leading to the occurrence of pathological remodeling diseases. In the past 20 years, many articles concerning the mechanobiology of cardiovascular homeostasis and remodeling have been published. To better understand the current development status, research hotspots and future development trends in the field, this paper uses CiteSpace software for bibliometric analysis, quantifies and visualizes the articles published in this field in the past 20 years, and reviews the research hotspots and emerging trends. The regulatory effects of mechanical stimulation on the biological behavior of endothelial cells, smooth muscle cells and the extracellular matrix, as well as the mechanical-related remodeling mechanism in heart failure, have always been research hotspots in this field. This paper reviews the research advances of these research hotspots in detail. This paper also introduces the research status of emerging hotspots, such as those related to cardiac fibrosis, homeostasis, mechanosensitive transcription factors and mechanosensitive ion channels. We hope to provide a systematic framework and new ideas for follow-up research on mechanobiology in the field of cardiovascular homeostasis and remodeling and promote the discovery of more therapeutic targets and novel markers of mechanobiology in the cardiovascular system.
Collapse
Affiliation(s)
- Wei Liao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Yuxi Huang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | | | - Ziqiu Hu
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Chuanrong Zhao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Guixue Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
3
|
Wang Y, Chu T, Meng C, Bian Y, Li J. Piezo1-specific Deletion in Macrophage Protects the Progression of Chronic Inflammatory Bowel Disease in Mice. Cell Mol Gastroenterol Hepatol 2025; 19:101495. [PMID: 40081571 DOI: 10.1016/j.jcmgh.2025.101495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND & AIMS Piezo1, a recently identified mechanically activated nonselective cation channel protein, demonstrates sensitivity to various mechanical stimuli, such as matrix stiffness and shear stress. Although accumulating evidence implicates Piezo1 channels in numerous physiologic and pathophysiologic processes, its involvement in dextran sulfate sodium (DSS)-induced acute and chronic inflammatory bowel disease (IBD) remains incompletely understood. This study aimed to investigate the effect of Piezo1 channels in macrophage polarization and its associated functions in IBD. METHODS DSS-induced inflammatory bowel disease model was established in Piezo1td/Tdt or Piezo1fl/fl and Piezo1△LysM male mice. Additionally, bone marrow-derived macrophages from Piezo1fl/fl and Piezo1△LysM male mice were isolated to elucidate the downstream targets of Piezo1 and the associated underlying molecular mechanisms. RESULTS Our findings revealed that Piezo1 deficiency in macrophages could protect mice from DSS-induced chronic IBD, as evidenced by improved colon length and the preservation of colon structure. The mitigation of inflammation during chronic IBD progression was observed with Piezo1 deficiency in macrophages, characterized by reduced macrophage accumulation, M1 macrophage polarization, T helper 1 infiltration, and decreased inflammatory cytokine secretion. Further investigations unveiled that Piezo1-deficient macrophages inhibit the expression and activity of Nod-like receptor protein 3 and nuclear factor kappa B in colon tissues and bone marrow-derived macrophages while regulating the nuclear translocation of p65. Conversely, macrophage Piezo1 activation enhanced inflammatory cytokine secretion by activating Nod-like receptor protein 3/nuclear factor kappa B pathways. CONCLUSIONS Myeloid Piezo1 mediates colonic immune response, and disrupting Piezo1 inhibits the progression of chronic IBD. This study provides hitherto undocumented evidence of the pivotal role of macrophage Piezo1 channels in regulating the progression of chronic IBD. Targeting macrophage Piezo1 may offer a promising therapeutic strategy against chronic IBD.
Collapse
Affiliation(s)
- Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China
| | - Chengzhen Meng
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China.
| |
Collapse
|
4
|
Li H, Li X, Sun Y, Zhi Z, Song L, Li M, Feng Y, Zhang Z, Liu Y, Chen Y, Zhao F, Zhu T. The Role of Ion Channels in Pulmonary Hypertension: A Review. Pulm Circ 2025; 15:e70050. [PMID: 39958971 PMCID: PMC11830494 DOI: 10.1002/pul2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/16/2024] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
Pulmonary hypertension (PH) constitutes a critical challenge in cardiopulmonary medicine with a pathogenesis that is multifaceted and intricate. Ion channels, crucial determinants of cellular electrochemical gradient modulation, have emerged as significant participants in the pathophysiological progression of PH. These channels, abundant on the membranes of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), pivotally navigate the nuanced interplay of cell proliferation, migration, and endothelial function, each vital to the pulmonary vascular remodeling (PVR) hallmark of PH. Our review delves into the mechanistic insights of potassium, calcium, magnesium, zinc, and chloride ion channels in relation to their involvement in PH. It not only emphasizes the notable advances and discoveries that cast these ion channels as underlying factors in the etiology and exacerbation of PH but also highlights their potential as innovative therapeutic targets.
Collapse
Affiliation(s)
- Han‐Fei Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Xin‐Yao Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Qing Sun
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Ze‐Ying Zhi
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Liao‐Fan Song
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Meng Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yi‐Ming Feng
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Zhi‐Hao Zhang
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yan‐Feng Liu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Jing Chen
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Fan‐Rong Zhao
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Tian‐Tian Zhu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
- Department of PharmacyThe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| |
Collapse
|
5
|
Bi Y, Li H, Diao M, Liu Q, Huang L, Tao Y, Wan Y, Lin X. Piezo1 overexpression in the uterus contributes to myometrium contraction and inflammation-associated preterm birth. J Transl Med 2024; 22:1140. [PMID: 39716206 DOI: 10.1186/s12967-024-05978-y] [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: 07/29/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Preterm birth, a leading cause of perinatal mortality and morbidity, is often associated with inflammation and aberrant myometrial contractions. This study investigates the role of Piezo1, a mechanosensitive ion channel, in myometrium contraction and inflammation-associated preterm birth. METHODS We employed Western blotting, Immunofluorescence, and Quantitative real-time PCR techniques to examine Piezo1 expression in uterine tissues. Functional assays, including myometrial contractility studies and cell contraction assays, were conducted to elucidate the effects of Piezo1 on myometrial contractions. Piezo1 inhibitors and gene knockdown techniques were used to investigate the impact of Piezo1 on inflammation-associated preterm birth, complemented by inflammatory cytokine profiling and calcium imaging to investigate the mechanism. RESULTS Our findings reveal that Piezo1 is the predominant mechanosensitive channel in mouse myometrium tissue and mouse primary uterine smooth muscle (pUSMCs), with increased expression during mouse and human pregnancy. Following lipopolysaccharide (LPS) intrauterine injection, Piezo1 mRNA and protein levels were elevated in the mouse uterine smooth muscle layer. Direct pharmacologic activation of Piezo1 by Yoda1 increased the contraction of pUSMCs and shortened the pregnancy duration. In contrast, inhibition with Gsmtx4 or siRNA knockdown of Piezo1 attenuated LPS-induced pUSMCs contraction and spontaneous uterine myometrium contraction. Additionally, blocking or knocking down Piezo1 prolonged the pregnancy in an LPS-induced preterm birth model. Yoda1 stimulation increased intracellular Ca2+ levels in pUSMCs, while Gsmtx4 reduced these levels. Gsmtx4 decreased cox-2 expression and inflammation factors in LPS-stimulated pUSMCs. CONCLUSIONS These results suggest that Piezo1 acts as a critical regulator of uterine function, and its overexpression may predispose to preterm labor through heightened myometrial activity and inflammation. The study underscores the potential of targeting Piezo1 as a therapeutic strategy to mitigate preterm birth associated with uterine inflammation.
Collapse
Affiliation(s)
- Yanmei Bi
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Min Diao
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Qian Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Lu Huang
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Yunkai Tao
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Yantong Wan
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Xuemei Lin
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, No. 20, Section 3, South of Renmin Road, Chengdu, 610041, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Shen YH, Ding D, Lian TY, Qiu BC, Yan Y, Wang PW, Zhang WH, Jing ZC. Panorama of artery endothelial cell dysfunction in pulmonary arterial hypertension. J Mol Cell Cardiol 2024; 197:61-77. [PMID: 39437884 DOI: 10.1016/j.yjmcc.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal lung disease characterized by progressive pulmonary vascular remodeling. The initial cause of pulmonary vascular remodeling is the dysfunction of pulmonary arterial endothelial cells (PAECs), manifested by changes in the categorization of cell subtypes, endothelial programmed cell death, such as apoptosis, necroptosis, pyroptosis, ferroptosis, et al., overproliferation, senescence, metabolic reprogramming, endothelial-to-mesenchymal transition, mechanosensitivity, and regulation ability of peripheral cells. Therefore, it is essential to explore the mechanism of endothelial dysfunction in the context of PAH. This review aims to provide a comprehensive understanding of the molecular mechanisms underlying endothelial dysfunction in PAH. We highlight the developmental process of PAECs and changes in PAH and summarise the latest classification of endothelial dysfunction. Our review could offer valuable insights into potential novel EC-specific targets for preventing and treating PAH.
Collapse
Affiliation(s)
- Ying-Huizi Shen
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Wen Wang
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei-Hua Zhang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Mote N, Kubik S, Polacheck WJ, Baker BM, Trappmann B. A nanoporous hydrogel-based model to study chemokine gradient-driven angiogenesis under luminal flow. LAB ON A CHIP 2024; 24:4892-4906. [PMID: 39308400 DOI: 10.1039/d4lc00460d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The growth of new blood vessels through angiogenesis is a highly coordinated process, which is initiated by chemokine gradients that activate endothelial cells within a perfused parent vessel to sprout into the surrounding 3D tissue matrix. While both biochemical signals from pro-angiogenic factors, as well as mechanical cues originating from luminal fluid flow that exerts shear stress on the vessel wall, have individually been identified as major regulators of endothelial cell sprouting, it remains unclear whether and how both types of cues synergize. To fill this knowledge gap, here, we created a 3D biomimetic model of chemokine gradient-driven angiogenic sprouting, in which a micromolded tube inside a hydrogel matrix is seeded with endothelial cells and connected to a perfusion system to control fluid flow rates and resulting shear forces on the vessel wall. To allow for the formation of chemokine gradients despite the presence of luminal flow, a nanoporous synthetic hydrogel that supports angiogenesis but limits the interstitial flow proved crucial. Using this system, we find that luminal flow and resulting shear stress is a major regulator of the speed and morphogenesis of angiogenic sprouting, whose action is mediated through changes in vascular permeability.
Collapse
Affiliation(s)
- Nidhi Mote
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Sarah Kubik
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - William J Polacheck
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, 27514 USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, 2174 Lurie BME Building, 1101 Beal Avenue, Ann Arbor, MI, 48109 USA
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Straße 6, 44227 Dortmund, Germany.
| |
Collapse
|
8
|
Wang YM, Chu TJ, Wan RT, Niu WP, Bian YF, Li J. Quercetin ameliorates atherosclerosis by inhibiting inflammation of vascular endothelial cells via Piezo1 channels. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155865. [PMID: 39004029 DOI: 10.1016/j.phymed.2024.155865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Natural antioxidants, exemplified by quercetin (Qu), have been shown to exert a protective effect against atherosclerosis (AS). However, the precise pharmacological mechanisms of Qu also remain elusive. PURPOSE Here, we aimed to uncover the anti-atherosclerotic mechanisms of Qu. METHODS/STUDY DESIGNS The inflammatory cytokine expression, activity of NLRP3 inflammasome and NF-κB, as well as mechanically activated currents and intracellular calcium levels were measured in endothelial cells (ECs). In addition, to explore whether Qu inhibited atherosclerotic plaque formation via Piezo1 channels, Ldlr-/- and Piezo1 endothelial-specific knockout mice (Piezo1△EC) were established. RESULTS Our findings revealed that Qu significantly inhibited Yoda1-evoked calcium response in human umbilical vein endothelial cells (HUVECs), underscoring its role as a selective modulator of Piezo1 channels. Additionally, Qu effectively reduced mechanically activated currents in HUVECs. Moreover, Qu exhibited a substantial inhibitory effect on inflammatory cytokine expression and reduced the activity of NF-κB/NLRP3 in ECs exposed to ox-LDL or mechanical stretch, and these effects remained unaffected after Piezo1 genetic depletion. Furthermore, our study demonstrated that Qu substantially reduced the formation of atherosclerotic plaques, and this effect remained consistent even after Piezo1 genetic depletion. CONCLUSION These results collectively provide compelling evidence that Qu ameliorates atherosclerosis by inhibiting the inflammatory response in ECs by targeting Piezo1 channels. In addition, Qu modulated atherosclerosis via inhibiting Piezo1 mediated NFκB/IL-1β and NLRP3/caspase1/ IL-1β axis to suppress the inflammation. Overall, this study reveals the potential mechanisms by which natural antioxidants, such as Qu, protect against atherosclerosis.
Collapse
Affiliation(s)
- Yu-Man Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Tian-Jiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Ren-Tao Wan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei-Pin Niu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China
| | - Yi-Fei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, 4655 Daxue Road Changqing District, Ji'nan, Shandong 250355, China.
| |
Collapse
|
9
|
Jin C, Su S, Yu S, Zhang Y, Chen K, Xiang M, Ma H. Essential Roles of PIEZO1 in Mammalian Cardiovascular System: From Development to Diseases. Cells 2024; 13:1422. [PMID: 39272994 PMCID: PMC11394449 DOI: 10.3390/cells13171422] [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: 07/15/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Mechanical force is the basis of cardiovascular development, homeostasis, and diseases. The perception and response of mechanical force by the cardiovascular system are crucial. However, the molecular mechanisms mediating mechanotransduction in the cardiovascular system are not yet understood. PIEZO1, a novel transmembrane mechanosensitive cation channel known for its regulation of touch sensation, has been found to be widely expressed in the mammalian cardiovascular system. In this review, we elucidate the role and mechanism of PIEZO1 as a mechanical sensor in cardiovascular development, homeostasis, and disease processes, including embryo survival, angiogenesis, cardiac development repair, vascular inflammation, lymphangiogenesis, blood pressure regulation, cardiac hypertrophy, cardiac fibrosis, ventricular remodeling, and heart failure. We further summarize chemical molecules targeting PIEZO1 for potential translational applications. Finally, we address the controversies surrounding emergent concepts and challenges in future applications.
Collapse
Affiliation(s)
- Chengjiang Jin
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Sheng’an Su
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Shuo Yu
- Department of Anesthesiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yue Zhang
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Kaijie Chen
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Meixiang Xiang
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Hong Ma
- Cardiovascular Key Laboratory of Zhejiang Province, National Key Laboratory of Vascular Implantable Devices, Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
10
|
Yang L, Peng Z, Gong F, Yan W, Shi Y, Li H, Zhou C, Yao H, Yuan M, Yu F, Feng L, Wan N, Liu G. TRPC4 aggravates hypoxic pulmonary hypertension by promoting pulmonary endothelial cell apoptosis. Free Radic Biol Med 2024; 219:141-152. [PMID: 38636714 DOI: 10.1016/j.freeradbiomed.2024.04.224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/31/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Pulmonary hypertension (PH) is a devastating disease that lacks effective treatment options and is characterized by severe pulmonary vascular remodeling. Pulmonary arterial endothelial cell (PAEC) dysfunction drives the initiation and pathogenesis of pulmonary arterial hypertension. Canonical transient receptor potential (TRPC) channels, a family of Ca2+-permeable channels, play an important role in various diseases. However, the effect and mechanism of TRPCs on PH development have not been fully elucidated. Among the TRPC family members, TRPC4 expression was markedly upregulated in PAECs from hypoxia combined with SU5416 (HySu)-induced PH mice and monocrotaline (MCT)-treated PH rats, as well as in hypoxia-exposed PAECs, suggesting that TRPC4 in PAECs may participate in the occurrence and development of PH. In this study, we aimed to investigate whether TRPC4 in PAECs has an aggravating effect on PH and elucidate the molecular mechanisms. We observed that hypoxia treatment promoted PAEC apoptosis through a caspase-12/endoplasmic reticulum stress (ERS)-dependent pathway. Knockdown of TRPC4 attenuated hypoxia-induced apoptosis and caspase-3/caspase-12 activity in PAECs. Accordingly, adeno-associated virus (AAV) serotype 6-mediated pulmonary endothelial TRPC4 silencing (AAV6-Tie-shRNA-TRPC4) or TRPC4 antagonist suppressed PH progression as evidenced by reduced right ventricular systolic pressure (RVSP), pulmonary vascular remodeling, PAEC apoptosis and reactive oxygen species (ROS) production. Mechanistically, unbiased RNA sequencing (RNA-seq) suggested that TRPC4 deficiency suppressed the expression of the proapoptotic protein sushi domain containing 2 (Susd2) in hypoxia-exposed mouse PAECs. Moreover, TRPC4 activated hypoxia-induced PAEC apoptosis by promoting Susd2 expression. Therefore, inhibiting TRPC4 ameliorated PAEC apoptosis and hypoxic PH in animals by repressing Susd2 signaling, which may serve as a therapeutic target for the management of PH.
Collapse
Affiliation(s)
- Liu Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zeyu Peng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fanpeng Gong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - WenXin Yan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yi Shi
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hanyi Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chang Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hong Yao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Menglu Yuan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Naifu Wan
- Department of Vascular & Cardiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guizhu Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
11
|
Luo T, Wu H, Zhu W, Zhang L, Huang Y, Yang X. Emerging therapies: Potential roles of SGLT2 inhibitors in the management of pulmonary hypertension. Respir Med 2024; 227:107631. [PMID: 38631526 DOI: 10.1016/j.rmed.2024.107631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
Pulmonary hypertension (PH) is a pathophysiological disorder that may involve multiple clinical conditions and may be associated with a variety of cardiovascular and respiratory diseases. Pulmonary hypertension due to left heart disease (PH-LHD) currently lacks targeted therapies, while Pulmonary arterial hypertension (PAH), despite approved treatments, carries considerable residual risk. Metabolic dysfunction has been linked to the pathogenesis and prognosis of PH through various studies, with emerging metabolic agents offering a potential avenue for improving patient outcomes. Sodium-glucose cotransporter 2 inhibitor (SGLT-2i), a novel hypoglycemic agent, could ameliorate metabolic dysfunction and exert cardioprotective effects. Recent small-scale studies suggest SGLT-2i treatment may improve pulmonary artery pressure in patients with PH-LHD, and the PAH animal model shows that SGLT-2i can reduce pulmonary vascular remodeling and prevent progression in PAH, suggesting potential benefits for patients with PH-LHD and perhaps PAH. This review aims to succinctly review PH's pathophysiology, and the connection between metabolic dysfunction and PH, and investigate the prospective mechanisms of action of SGLT-2i in PH-LHD and PAH management.
Collapse
Affiliation(s)
- Taimin Luo
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, 610000, China
| | - Hui Wu
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wanlong Zhu
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Pharmacy, Panzhihua Second People's Hospital, Panzhihua, 617000, China
| | - Liaoyun Zhang
- Department of Pharmacy, Sichuan Provincial Maternity and Child Health Care Hospital & Women's and Children's Hospital, Chengdu, 610000, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
12
|
Karkempetzaki AI, Ravid K. Piezo1 and Its Function in Different Blood Cell Lineages. Cells 2024; 13:482. [PMID: 38534326 PMCID: PMC10969519 DOI: 10.3390/cells13060482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Mechanosensation is a fundamental function through which cells sense mechanical stimuli by initiating intracellular ion currents. Ion channels play a pivotal role in this process by orchestrating a cascade of events leading to the activation of downstream signaling pathways in response to particular stimuli. Piezo1 is a cation channel that reacts with Ca2+ influx in response to pressure sensation evoked by tension on the cell lipid membrane, originating from cell-cell, cell-matrix, or hydrostatic pressure forces, such as laminar flow and shear stress. The application of such forces takes place in normal physiological processes of the cell, but also in the context of different diseases, where microenvironment stiffness or excessive/irregular hydrostatic pressure dysregulates the normal expression and/or activation of Piezo1. Since Piezo1 is expressed in several blood cell lineages and mutations of the channel have been associated with blood cell disorders, studies have focused on its role in the development and function of blood cells. Here, we review the function of Piezo1 in different blood cell lineages and related diseases, with a focus on megakaryocytes and platelets.
Collapse
Affiliation(s)
- Anastasia Iris Karkempetzaki
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Katya Ravid
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA;
- Whitaker Cardiovascular Institute, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| |
Collapse
|
13
|
Coste B, Delmas P. PIEZO Ion Channels in Cardiovascular Functions and Diseases. Circ Res 2024; 134:572-591. [PMID: 38422173 DOI: 10.1161/circresaha.123.322798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The cardiovascular system provides blood supply throughout the body and as such is perpetually applying mechanical forces to cells and tissues. Thus, this system is primed with mechanosensory structures that respond and adapt to changes in mechanical stimuli. Since their discovery in 2010, PIEZO ion channels have dominated the field of mechanobiology. These have been proposed as the long-sought-after mechanosensitive excitatory channels involved in touch and proprioception in mammals. However, more and more pieces of evidence point to the importance of PIEZO channels in cardiovascular activities and disease development. PIEZO channel-related cardiac functions include transducing hemodynamic forces in endothelial and vascular cells, red blood cell homeostasis, platelet aggregation, and arterial blood pressure regulation, among others. PIEZO channels contribute to pathological conditions including cardiac hypertrophy and pulmonary hypertension and congenital syndromes such as generalized lymphatic dysplasia and xerocytosis. In this review, we highlight recent advances in understanding the role of PIEZO channels in cardiovascular functions and diseases. Achievements in this quickly expanding field should open a new road for efficient control of PIEZO-related diseases in cardiovascular functions.
Collapse
Affiliation(s)
- Bertrand Coste
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| | - Patrick Delmas
- Centre de Recherche en CardioVasculaire et Nutrition, Aix-Marseille Université - INSERM 1263 - INRAE 1260, Marseille, France
| |
Collapse
|
14
|
Zhang M, Wang QR, Hou X, Wang Q, Yang X, Zhou T, Liu X, Wu L, Wang J, Jin X, Liu Z, Huang B. Blockage of mechanosensitive Piezo1 channel alleviates the severity of experimental malaria-associated acute lung injury. Parasit Vectors 2024; 17:46. [PMID: 38303078 PMCID: PMC10832208 DOI: 10.1186/s13071-024-06144-5] [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: 10/29/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Malaria-associated acute lung injury (MA-ALI) is a well-recognized clinical complication of severe, complicated malaria that is partly driven by sequestrations of infected red blood cells (iRBCs) on lung postcapillary induced impaired blood flow. In earlier studies the mechanosensitive Piezo1 channel emerged as a regulator of mechanical stimuli, but the function and underlying mechanism of Piezo1 impacting MA-ALI severity via sensing the impaired pulmonary blood flow are still not fully elucidated. Thus, the present study aimed to explore the role of Piezo1 in the severity of murine MA-ALI. METHODS Here, we utilized a widely accepted murine model of MA-ALI using C57BL/6 mice with Plasmodium berghei ANKA infection and then added a Piezo1 inhibitor (GsMTx4) to the model. The iRBC-stimulated Raw264.7 macrophages in vitro were also targeted with GsMTx4 to further explore the potential mechanism. RESULTS Our data showed an elevation in the expression of Piezo1 and number of Piezo1+-CD68+ macrophages in lung tissues of the experimental MA-ALI mice. Compared to the infected control mice, the blockage of Piezo1 with GsMTx4 dramatically improved the survival rate but decreased body weight loss, peripheral blood parasitemia/lung parasite burden, experimental cerebral malaria incidence, total protein concentrations in bronchoalveolar lavage fluid, lung wet/dry weight ratio, vascular leakage, pathological damage, apoptosis and number of CD68+ and CD86+ macrophages in lung tissues. This was accompanied by a dramatic increase in the number of CD206+ macrophages (M2-like subtype), upregulation of anti-inflammatory cytokines (e.g. IL-4 and IL-10) and downregulation of pro-inflammatory cytokines (e.g. TNF-α and IL-1β). In addition, GsMTx4 treatment remarkably decreased pulmonary intracellular iron accumulation, protein level of 4-HNE (an activator of ferroptosis) and the number of CD68+-Piezo1+ and CD68+-4-HNE+ macrophages but significantly increased protein levels of GPX4 (an inhibitor of ferroptosis) in experimental MA-ALI mice. Similarly, in vitro study showed that the administration of GsMTx4 led to a remarkable elevation in the mRNA levels of CD206, IL-4, IL-10 and GPX-4 but to a substantial decline in CD86, TNF-α, IL-1β and 4-HNE in the iRBC-stimulated Raw264.7 cells. CONCLUSIONS Our findings indicated that blockage of Piezo1 with GsMTx4 alleviated the severity of experimental MA-ALI in mice partly by triggering pulmonary macrophage M2 polarization and subsequent anti-inflammatory responses but inhibited apoptosis and ferroptosis in lung tissue. Our data suggested that targeting Piezo1 in macrophages could be a promising therapeutic strategy for treating MA-ALI.
Collapse
Affiliation(s)
- Min Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Qian Ru Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xinpeng Hou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Qi Wang
- Guangzhou Chest Hospital, Guangzhou, 510095, People's Republic of China
| | - Xiaoyan Yang
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, 511400, People's Republic of China
| | - Tingting Zhou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xiaobo Liu
- School of Basic Medical Science, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Lirong Wu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Jie Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China
| | - Zhenlong Liu
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
| | - Bo Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
- School of Basic Medical Science, Guangdong Pharmaceutical University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
15
|
Kelley B, Zhang EY, Khalfaoui L, Schiliro M, Wells N, Pabelick CM, Prakash YS, Vogel ER. Piezo channels in stretch effects on developing human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2023; 325:L542-L551. [PMID: 37697925 PMCID: PMC11068394 DOI: 10.1152/ajplung.00008.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
The use of respiratory support strategies such as continuous positive airway pressure in premature infants can substantially stretch highly compliant perinatal airways, leading to airway hyperreactivity and remodeling in the long term. The mechanisms by which stretch detrimentally affects the airway are unknown. Airway smooth muscle cells play a critical role in contractility and remodeling. Using 18-22-wk gestation human fetal airway smooth muscle (fASM) as an in vitro model, we tested the hypothesis that mechanosensitive Piezo (PZ) channels contribute to stretch effects. We found that PZ1 and PZ2 channels are expressed in the smooth muscle of developing airways and that their expression is influenced by stretch. PZ activation via agonist Yoda1 or stretch results in significant [Ca2+]i responses as well as increased extracellular matrix production. These data suggest that functional PZ channels may play a role in detrimental stretch-induced airway changes in the context of prematurity.NEW & NOTEWORTHY Piezo channels were first described just over a decade ago and their function in the lung is largely unknown. We found that piezo channels are present and functional in the developing airway and contribute to intracellular calcium responses and extracellular matrix remodeling in the setting of stretch. This may improve our understanding of the mechanisms behind development of chronic airway diseases, such as asthma, in former preterm infants exposed to respiratory support, such as continuous positive airway pressure (CPAP).
Collapse
Affiliation(s)
- Brian Kelley
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Emily Y Zhang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Latifa Khalfaoui
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Marta Schiliro
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Natalya Wells
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Elizabeth R Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
16
|
Arenas GA, Valenzuela JG, Peñaloza E, Paz AA, Iturriaga R, Saez CG, Krause BJ. Transcriptional Profiling of Human Endothelial Cells Unveils PIEZO1 and Mechanosensitive Gene Regulation by Prooxidant and Inflammatory Inputs. Antioxidants (Basel) 2023; 12:1874. [PMID: 37891953 PMCID: PMC10604317 DOI: 10.3390/antiox12101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/15/2023] [Accepted: 09/24/2023] [Indexed: 10/29/2023] Open
Abstract
PIEZO1 is a mechanosensitive cation channel implicated in shear stress-mediated endothelial-dependent vasorelaxation. Since altered shear stress patterns induce a pro-inflammatory endothelial environment, we analyzed transcriptional profiles of human endothelial cells to determine the effect of altered shear stress patterns and subsequent prooxidant and inflammatory conditions on PIEZO1 and mechanosensitive-related genes (MRG). In silico analyses were validated in vitro by assessing PIEZO1 transcript levels in both the umbilical artery (HUAEC) and vein (HUVEC) endothelium. Transcriptional profiling showed that PIEZO1 and some MRG associated with the inflammatory response were upregulated in response to high (15 dyn/cm2) and extremely high shear stress (30 dyn/cm2) in HUVEC. Changes in PIEZO1 and inflammatory MRG were paralleled by p65 but not KLF or YAP1 transcription factors. Similarly, PIEZO1 transcript levels were upregulated by TNF-alpha (TNF-α) in diverse endothelial cell types, and pre-treatment with agents that prevent p65 translocation to the nucleus abolished PIEZO1 induction. ChIP-seq analysis revealed that p65 bonded to the PIEZO1 promoter region, an effect increased by the stimulation with TNF-α. Altogether this data showed that NF-kappa B activation via p65 signaling regulates PIEZO1 expression, providing a new molecular link for prooxidant and inflammatory responses and mechanosensitive pathways in the endothelium.
Collapse
Affiliation(s)
- German A. Arenas
- Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua 2841959, Chile;
| | - Jose G. Valenzuela
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile (C.G.S.)
| | - Estefanía Peñaloza
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile
| | - Adolfo A. Paz
- Instituto de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, Santiago 7500000, Chile
| | - Rodrigo Iturriaga
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Investigación en Fisiología y Medicina en Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta 1271155, Chile
| | - Claudia G. Saez
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile (C.G.S.)
| | - Bernardo J. Krause
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile
| |
Collapse
|
17
|
Xu Y, Huang Y, Cheng X, Hu B, Jiang D, Wu L, Peng S, Hu J. Mechanotransductive receptor Piezo1 as a promising target in the treatment of fibrosis diseases. Front Mol Biosci 2023; 10:1270979. [PMID: 37900917 PMCID: PMC10602816 DOI: 10.3389/fmolb.2023.1270979] [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: 08/11/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Fibrosis could happen in every organ, leading to organic malfunction and even organ failure, which poses a serious threat to global health. Early treatment of fibrosis has been reported to be the turning point, therefore, exploring potential correlates in the pathogenesis of fibrosis and how to reverse fibrosis has become a pressing issue. As a mechanism-sensitive cationic calcium channel, Piezo1 turns on in response to changes in the lipid bilayer of the plasma membrane. Piezo1 exerts multiple biological roles, including inhibition of inflammation, cytoskeletal stabilization, epithelial-mesenchymal transition, stromal stiffness, and immune cell mechanotransduction, interestingly enough. These processes are closely associated with the development of fibrotic diseases. Recent studies have shown that deletion or knockdown of Piezo1 attenuates the onset of fibrosis. Therefore, in this paper we comprehensively describe the biology of this gene, focusing on its potential relevance in pulmonary fibrosis, renal fibrosis, pancreatic fibrosis, and cardiac fibrosis diseases, except for the role of drugs (agonists), increased intracellular calcium and mechanical stress using this gene in alleviating fibrosis.
Collapse
Affiliation(s)
- Yi Xu
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Yiqian Huang
- The Second Affiliated Hospital of Nanchang University, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Xiaoqing Cheng
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bin Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Danling Jiang
- Department of Ultrasound Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
18
|
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
|
19
|
Wang LJ, Feng F, Li JC, Chen TT, Liu LP. Role of heparanase in pulmonary hypertension. Front Pharmacol 2023; 14:1202676. [PMID: 37637421 PMCID: PMC10450954 DOI: 10.3389/fphar.2023.1202676] [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: 04/09/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Pulmonary hypertension (PH) is a pathophysiological condition of increased pulmonary circulation vascular resistance due to various reasons, which mainly leads to right heart dysfunction and even death, especially in critically ill patients. Although drug interventions have shown some efficacy in improving the hemodynamics of PH patients, the mortality rate remains high. Hence, the identification of new targets and treatment strategies for PH is imperative. Heparanase (HPA) is an enzyme that specifically cleaves the heparan sulfate (HS) side chains in the extracellular matrix, playing critical roles in inflammation and tumorigenesis. Recent studies have indicated a close association between HPA and PH, suggesting HPA as a potential therapeutic target. This review examines the involvement of HPA in PH pathogenesis, including its effects on endothelial cells, inflammation, and coagulation. Furthermore, HPA may serve as a biomarker for diagnosing PH, and the development of HPA inhibitors holds promise as a targeted therapy for PH treatment.
Collapse
Affiliation(s)
- Lin-Jun Wang
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Fei Feng
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Jian-Chun Li
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Li-Ping Liu
- The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
- Departments of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
20
|
Endothelial mechanosensing: A forgotten target to treat vascular remodeling in hypertension? Biochem Pharmacol 2022; 206:115290. [DOI: 10.1016/j.bcp.2022.115290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/23/2022]
|