1
|
Fließer E, Jandl K, Chen SH, Wang MT, Schupp JC, Kuebler WM, Baker AH, Kwapiszewska G. Transcriptional signatures of endothelial cells shape immune responses in cardiopulmonary health and disease. JCI Insight 2025; 10:e191059. [PMID: 40401523 DOI: 10.1172/jci.insight.191059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
The cardiopulmonary vasculature and its associated endothelial cells (ECs) play an essential role in sustaining life by ensuring the delivery of oxygen and nutrients. Beyond these foundational functions, ECs serve as key regulators of immune responses. Recent advances in single-cell RNA sequencing have revealed that the cardiopulmonary vasculature is composed of diverse EC subpopulations, some of which exhibit specialized immunomodulatory properties. Evidence for immunomodulation includes distinct expression profiles associated with antigen presentation, cytokine secretion, immune cell recruitment, translocation, and clearance - functions critical for maintaining homeostasis in the heart and lungs. In cardiopulmonary diseases, ECs undergo substantial transcriptional reprogramming, leading to a shift from homeostasis to an activated state marked by heightened immunomodulatory activity. This transformation has highlighted the critical role for ECs in disease pathogenesis and their potential as future therapy targets. This Review emphasizes the diverse functions of ECs in the heart and lungs, particularly adaptive and maladaptive immunoregulatory roles in cardiopulmonary health and disease.
Collapse
Affiliation(s)
- Elisabeth Fließer
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| | - Katharina Jandl
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Mei-Tzu Wang
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pulmonary and Infectious Diseases, Hannover Medical School, Hannover, Germany
- Biomedical Research in End-Stage and Obstructive Lung Disease (BREATH), German Center for Lung Research BREATH, Hannover, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- German Center for Lung Research, Associated Partner Site Berlin, Berlin, Germany
- Department of Surgery and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre, St Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Cardiovascular Research Institute Maastricht, School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of German Lung Center, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
2
|
Flores CV, Chan SY. Therapeutic targets for pulmonary arterial hypertension: insights into the emerging landscape. Expert Opin Ther Targets 2025:1-17. [PMID: 40368635 DOI: 10.1080/14728222.2025.2507034] [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: 02/24/2025] [Revised: 04/21/2025] [Accepted: 05/13/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive, life-threatening disease driven by vascular remodeling, right ventricular (RV) dysfunction, and metabolic and inflammatory dysregulation. Current therapies primarily target vasodilation to relieve symptoms but do not reverse disease progression. The recent approval of sotatercept, which modulates BMP/TGF-β signaling, marks a shift toward anti-remodeling therapies. Building on this, recent preclinical advances have identified promising therapeutic targets and potentially disease-modifying treatments. AREAS COVERED This review synthesizes the evolving preclinical landscape of emerging PAH therapeutic targets and drugs, highlighting innovative approaches aimed at addressing the underlying mechanisms of disease progression. Additionally, we discuss novel therapeutic strategies under development. EXPERT OPINION Recent advances in PAH research have identified novel therapeutic targets beyond vasodilators, including modulation of BMP/TGF-β signaling, metabolic programs, epigenetics, cancer-related signaling, the extracellular matrix, and immune pathways, among others. Sotatercept represents a significant advance in therapies that go beyond vasodilation, and long-term safety, efficacy, and durability are being assessed. Future treatment strategies will focus on precision approaches, noninvasive technologies, and regenerative biology to improve outcomes and reverse vascular remodeling.
Collapse
Affiliation(s)
- Christopher V Flores
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Geng Y, Wang H, Bai Z, Ge R. Iron Replacement Attenuates Hypoxic Pulmonary Hypertension by Remodeling Energy Metabolism via Regulating the HIF2α/Mitochondrial Complex I, III/ROS Axis. Biomolecules 2025; 15:742. [PMID: 40427635 PMCID: PMC12109292 DOI: 10.3390/biom15050742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Iron deficiency is highly prevalent in patients with idiopathic pulmonary hypertension; nevertheless, its role and clinical significance in hypoxic pulmonary hypertension (HPH) remain elusive. Therefore, this study aims to clarify the role and molecular mechanisms of iron in HPH. By means of a retrospective analysis of clinical data from HPH patients and examinations of HPH animal models, we discovered that both HPH patients and animal models exhibit significant iron deficiency, characterized by reduced hepatic iron storage and elevated hepcidin expression. To further explore iron's role in HPH, we modulated iron metabolism through pharmacological and dietary interventions in chronic hypoxic animal models. The results showed that iron deficiency exacerbated chronic hypoxia-induced pulmonary hypertension and right ventricular hypertrophy, while iron supplementation alleviated these conditions. Further investigations revealed that iron regulates HIF2α expression in pulmonary arterial endothelial cells (PAECs) under chronic hypoxia. Therefore, through in vivo and in vitro experiments, we demonstrated that HIF2α inhibition attenuates chronic hypoxia-induced pulmonary hypertension and right ventricular hypertrophy. Mechanistically, chronic hypoxia-mediated iron deficiency enhances HIF2α activation, subsequently suppressing iron/sulfur cluster assembly enzyme (ISCU) expression. This leads to decreased mitochondrial complexes I and III activity, increased reactive oxygen species (ROS) production, and inhibited oxidative phosphorylation. Consequently, metabolic reprogramming in PAECs results in a proliferation/apoptosis imbalance, ultimately exacerbating hypoxia-induced pulmonary hypertension and right ventricular hypertrophy. Collectively, our findings demonstrate that iron supplementation mitigates HPH progression by modulating HIF2α-mediated metabolic reprogramming in PAECs, revealing multiple therapeutic targets for HPH.
Collapse
Affiliation(s)
- Yumei Geng
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (Y.G.); (H.W.)
- Key Laboratory of High Altitude Medicine (Ministry of Education), Qinghai University, Xining 810001, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Huijie Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (Y.G.); (H.W.)
- Key Laboratory of High Altitude Medicine (Ministry of Education), Qinghai University, Xining 810001, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Zhenzhong Bai
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (Y.G.); (H.W.)
- Key Laboratory of High Altitude Medicine (Ministry of Education), Qinghai University, Xining 810001, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (Y.G.); (H.W.)
- Key Laboratory of High Altitude Medicine (Ministry of Education), Qinghai University, Xining 810001, China
- Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| |
Collapse
|
4
|
Zhao Y, Ding C, Su H, Wang A, Tang A, Zhao H, Ma Y, Zhang M, Liu W, Wang R, Zhang Z, Yang S, Liang D, Huang Y, Qian K, Huang L, Fu Q, Cao Y. Single Test-Based Diagnosis and Subtyping of Pulmonary Hypertension Caused by Fibrosing Mediastinitis Using Plasma Metabolic Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416454. [PMID: 40047331 PMCID: PMC12061239 DOI: 10.1002/advs.202416454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/15/2025] [Indexed: 05/10/2025]
Abstract
Pulmonary hypertension (PH) often leads to poor survival outcomes and encompasses diverse subtypes with distinct underlying causes. Specifically, PH resulting from fibrosing mediastinitis (FM-PH) presents significant diagnostic challenges due to nonspecific symptoms and overlap of clinical characterization with other PH subtypes, leading to frequent misdiagnosis and delayed treatment. Moreover, the complex diagnostic procedures impose a significant burden on FM-PH patients, many of whom already experience mobility difficulties. This study represents a single test-based diagnosis of FM-PH, using the plasma metabolites obtained through ferric particle-enhanced laser desorption/ionization mass spectrometry analysis. Distinct metabolic alterations in FM-PH are identified compared to healthy controls and other PH subtypes, achieving an area under the curve (AUC) of 0.987 for FM-PH diagnosis and 0.728 for differentiating FM-PH from other subtypes. By addressing existing gaps in diagnostic strategies, this research highlights the potential of metabolic analysis in elucidating the metabolic landscape of PH.
Collapse
Affiliation(s)
- Yating Zhao
- Heart, Lung and Vessels CenterSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
- School of MedicineJiangsu UniversityZhenjiang212000China
- Department of CardiologyPulmonary Vascular Disease Center (PVDC)Gansu Provincial HospitalLanzhou730000China
| | - Chunmeng Ding
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Hongling Su
- Department of CardiologyPulmonary Vascular Disease Center (PVDC)Gansu Provincial HospitalLanzhou730000China
| | - Aqian Wang
- Department of CardiologyPulmonary Vascular Disease Center (PVDC)Gansu Provincial HospitalLanzhou730000China
| | - Aiping Tang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital)Lanzhou730000China
| | - Hongfan Zhao
- The First Clinical Medical SchoolLanzhou UniversityLanzhou730000China
| | - Ya Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital)Lanzhou730000China
| | - Min Zhang
- Clinical Research CenterSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Ruimin Wang
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Ziyue Zhang
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Dingyitai Liang
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Yida Huang
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for CancerSchool of Biomedical EngineeringInstitute of Medical Robotics and Shanghai Academy of Experimental MedicineShanghai Jiao Tong UniversityShanghai200030China
| | - Lin Huang
- Department of Clinical Laboratory MedicineShanghai Chest Hospital, Shanghai Jiao Tong UniversityInstitute of Thoracic OncologyShanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghai200030China
| | - Qihua Fu
- Center for Medical Genetics and Rare DiseasesSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| | - Yunshan Cao
- Heart, Lung and Vessels CenterSichuan Provincial People's HospitalUniversity of Electronic Science and Technology of ChinaChengduSichuan610072China
| |
Collapse
|
5
|
Si Y, Huang H, Pan J, Luo X, Zhang J, Guo Y, Liu D. Identification of potential biomarkers and pathways involved in high-altitude pulmonary edema using GC-MS and LC-MS metabolomic methods. Sci Rep 2024; 14:30978. [PMID: 39730680 PMCID: PMC11680936 DOI: 10.1038/s41598-024-82047-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024] Open
Abstract
High-altitude pulmonary edema (HAPE) is a life-threatening altitude sickness afflicting certain individuals after rapid ascent to high altitude above 2500 m. In the setting of HAPE, an early diagnosis is critical and currently based on clinical evaluation. The aim of this study was to utilize the metabolomics to identify the altered metabolic patterns and potential biomarkers for HAPE. Serum samples from HAPE patients (n = 24) and healthy controls (n = 21) were analyzed by gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS) to profile differential metabolites and explore dysregulated metabolic pathways. The correlation analysis and receiver operating characteristic (ROC) curve analysis were further performed to screen biomarkers for HAPE. A total of 119 differential metabolites between the control and HAPE groups were identified. Top dysregulated pathways included pyrimidine metabolism, citrate cycle, sulfur metabolism, phenylalanine metabolism and purine metabolism. After correlation analysis with clinical indices, 39 differential metabolites were obtained as potential biomarkers related to HAPE. Finally, 7 biomarkers, specifically S-nitroso-N-acetylcysteine, aminocaproic acid, emodin, threo-hydroxyaspartic acid, 6-hydroxynicotinic acid, 3-acetylphenol sulfate and cis-aconitic acid, were screened out through ROC analysis, which displayed high diagnostic accuracy for HAPE. Taken together, the altered serum metabolic profile is associated with the occurrence of HAPE. Diagnostic tests based on the biomarkers from metabolomics may hold promise as a strategy for early detection of HAPE.
Collapse
Affiliation(s)
- Yachen Si
- Department of Intensive Care Unit, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China
- Department of Internal Medicine, The 944 Hospital of Joint Logistics Support Force of Chinese PLA, Jiuquan, China
| | - He Huang
- Department of Neurology, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China
| | - Jing Pan
- Department of Health Service, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China
| | - Xiaozheng Luo
- Department of Intensive Care Unit, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China
| | - Jiangming Zhang
- Department of Intensive Care Unit, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China
| | - Yan Guo
- Department of Stomatology, Western Theater Command General Hospital, Chengdu, China.
| | - Dongmei Liu
- Department of Intensive Care Unit, The 940 Hospital of Joint Logistics Support Force of Chinese PLA, Lanzhou, China.
| |
Collapse
|
6
|
Luo L, Zhuang X, Fu L, Dong Z, Yi S, Wang K, Jiang Y, Zhao J, Yang X, Hei F. The role of the interplay between macrophage glycolytic reprogramming and NLRP3 inflammasome activation in acute lung injury/acute respiratory distress syndrome. Clin Transl Med 2024; 14:e70098. [PMID: 39623879 PMCID: PMC11612265 DOI: 10.1002/ctm2.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a severe respiratory condition associated with elevated morbidity and mortality. Understanding their complex pathophysiological mechanisms is crucial for developing new preventive and therapeutic strategies. Recent studies highlight the significant role of inflammation involved in ALI/ARDS, particularly the hyperactivation of the NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) inflammasome in macrophages. This activation drives pulmonary inflammation by releasing inflammatory signalling molecules and is linked to metabolic reprogramming, marked by increased glycolysis and reduced oxidative phosphorylation. However, the relationship between NLRP3 inflammasome activation and macrophage glycolytic reprogramming in ALI/ARDS, as well as the molecular mechanisms regulating these processes, remain elusive. This review provides a detailed description of the interactions and potential mechanisms linking NLRP3 inflammasome activation with macrophage glycolytic reprogramming, proposing that glycolytic reprogramming may represent a promising therapeutic target for mitigating inflammatory responses in ALI/ARDS. KEY POINTS: NLRP3 inflammasome activation is pivotal in mediating the excessive inflammatory response in ALI/ARDS. Glycolytic reprogramming regulates NLRP3 inflammasome activation. Therapeutic potential of targeting glycolytic reprogramming to inhibit NLRP3 inflammasome activation in ALI/ARDS.
Collapse
Affiliation(s)
- Lan Luo
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaoli Zhuang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Lin Fu
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ziyuan Dong
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Shuyuan Yi
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Kan Wang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yu Jiang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ju Zhao
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Xiaofang Yang
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Feilong Hei
- Department of Extracorporeal Circulation and Mechanical Circulation AssistantsCenter for Cardiac Intensive CareBeijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
7
|
Ge Q, Zhang T, Yu J, Lu X, Xiao S, Zhang T, Qing T, Xiao Z, Zeng L, Luo L. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed Pharmacother 2024; 181:117706. [PMID: 39581144 DOI: 10.1016/j.biopha.2024.117706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiovascular disease characterized by elevated pulmonary vascular resistance, progressive increases in pulmonary artery pressures, ultimately leading to right-sided heart failure, and potentially mortality. Pulmonary vascular remodeling is pivotal in PAH onset and progression. While targeted drug therapies have notably ameliorated PAH prognosis, current medications primarily focus on vascular vasodilation, with limited ability to reverse pulmonary vascular remodeling fundamentally, resulting in suboptimal patient prognoses. Cellular death in pulmonary vasculature, once thought to be confined to apoptosis and necrosis, has evolved with the identification of pyroptosis, autophagy, and ferroptosis, revealing their association with vascular injury in PAH. These novel forms of regulated cellular death impact reactive oxygen species (ROS) generation, calcium stress, and inflammatory cascades, leading to pulmonary vascular cell loss, exacerbating vascular injury, and mediating adverse remodeling, inflammation, immune anomalies, and current emerging mechanisms (such as endothelial-mesenchymal transition, abnormal energy metabolism, and epigenetic regulation) in the pathogenesis of PAH. This review comprehensively delineates the roles of autophagy, pyroptosis, and ferroptosis in PAH, elucidating recent advances in their involvement and regulation of vascular injury. It juxtaposes their distinct functions in PAH and discusses the interplay of these programmed cell deaths in pulmonary vascular injury, highlighting the benefits of combined targeted therapies in mitigating pulmonary arterial hypertension-induced vascular injury, providing novel insights into targeted treatments for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Jiangbiao Yu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Xuelin Lu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Sijie Xiao
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Ting Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tao Qing
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China.
| |
Collapse
|
8
|
Wang MY, Mo XY, Yi MX, Lu HY. Visualization of the relationship between metabolism and lung diseases from the perspective of bibliometric analysis: research trends and future prospects. Front Med (Lausanne) 2024; 11:1443926. [PMID: 39664315 PMCID: PMC11631585 DOI: 10.3389/fmed.2024.1443926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024] Open
Abstract
Background Extensive research has examined the role of metabolism in lung disease development, yet a comprehensive literature review remains absent despite numerous publications. Objective This study aims to visualize and assess the advancements in research on metabolism and its role in lung diseases. Methods Publications from January 1, 1991, to April 30, 2024, related to lung diseases and metabolism were sourced from the Web of Science Core Collection and analyzed using CiteSpace 6.2.R4, VOSviewer 1.6.19, Bibliometrix, R Studio, and various online tools. Results A total of 1,542 studies were collected and processed through these platforms for literature analysis and data visualization. The analysis revealed a sharp increase in annual publications on metabolism and lung diseases, with the United States and China emerging as leading contributors. Current research trends highlight a shift toward investigating metabolic reprogramming of immune cells in the context of lung diseases. Moreover, genes such as TNF, DIF, AKT1, INS, IL-6, CXCL8, IL-1β, TP53, NF-κB1, MTOR, IFNG, TGF-β1, HIF1α, VEGFA, IL-10, NFE2L2, PPARG, AKT, CRP, STAT3, and CD4 have received significant attention in this research domain. Employing a bibliometric approach, this study offers a comprehensive and objective examination of the knowledge landscape, shedding light on the evolving trends in this field. The findings serve as a valuable resource for researchers, offering a clearer perspective on the advancements in metabolism-related lung disease studies.
Collapse
Affiliation(s)
| | | | | | - Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
9
|
Wang H, Gao Y, Bai J, Liu H, Li Y, Zhang J, Ma C, Zhao X, Zhang L, Wan K, Zhu D. CircLMBR1 inhibits phenotypic transformation of hypoxia-induced pulmonary artery smooth muscle via the splicing factor PUF60. Eur J Pharmacol 2024; 980:176855. [PMID: 39059570 DOI: 10.1016/j.ejphar.2024.176855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
Phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs) contributes to vascular remodeling in hypoxic pulmonary hypertension (PH). Recent studies have suggested that circular RNAs (circRNAs) may play important roles in the vascular remodeling of hypoxia-induced PH. However, whether circRNAs cause pulmonary vascular remodeling by regulating the phenotypic transformation in PH has not been investigated. Microarray and RT-qPCR analysis identified that circLMBR1, a novel circRNA, decreased in mouse lung tissues of the hypoxia-SU5416 PH model, as well as in human PASMCs and mouse PASMCs exposed to hypoxia. Overexpression of circLMBR1 in the Semaxinib (SU5416) mouse model ameliorated hypoxia-induced PH and vascular remodeling in the lungs. Notably, circLMBR1 was mainly distributed in the nucleus and bound to the splicing factor PUF60. CircLMBR1 suppressed the phenotypic transformation of human PASMCs and vascular remodeling by inhibiting PUF60 expression. Furthermore, we identified U2AF65 as the downstream regulatory factor of PUF60. U2AF65 directly interacted with the pre-mRNA of the contractile phenotype marker smooth muscle protein 22-α (SM22α) and inhibited its splicing. Meanwhile, hypoxia exposure increased the formation of the PUF60-U2AF65 complex, thereby inhibiting SM22α production and inducing the transition of human PASMCs from a contractile phenotype to a synthetic phenotype. Overall, our results verified the important role of circLMBR1 in the pathological process of PH. We also proposed a new circLMBR1/PUF60-U2AF65/pre-SM22α pathway that could regulate the phenotypic transformation and proliferation of human PASMCs. This study may provide new perspectives for the diagnosis and treatment of PH.
Collapse
MESH Headings
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Animals
- Humans
- Mice
- Vascular Remodeling/drug effects
- Vascular Remodeling/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Phenotype
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Male
- Splicing Factor U2AF/genetics
- Splicing Factor U2AF/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia/genetics
- Mice, Inbred C57BL
- Cell Hypoxia
- Indoles/pharmacology
- Pyrroles
Collapse
Affiliation(s)
- Hongdan Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yupei Gao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Huiyu Liu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yiying Li
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Junting Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Xijuan Zhao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Kuiyu Wan
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, 150081, PR China.
| |
Collapse
|
10
|
Liu R, Zhou T, Li X, Zou Q, Yu J, Ye J, Wang W, Zhou Y, Sun SK. A Non-Metallic Nanozyme Ameliorates Pulmonary Hypertension Through Inhibiting ROS/TGF-β1 Signaling. Adv Healthc Mater 2024:e2401909. [PMID: 39155419 DOI: 10.1002/adhm.202401909] [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: 05/23/2024] [Revised: 08/01/2024] [Indexed: 08/20/2024]
Abstract
Pulmonary hypertension (PH) is a life-threatening cardiovascular disease with a lack of effective treatment options. Nanozymes, though promising for PH therapy, pose safety risks due to their metallic nature. Here, a non-metallic nanozyme is reported for the treatment of monocrotaline (MCT)-induced PH with a therapeutic mechanism involving the ROS/TGF-β1 signaling. The synthesized melanin-polyvinylpyrrolidone-polyethylene glycol (MPP) nanoparticles showcase ultra-small size, excellent water solubility, high biocompatibility, and remarkable antioxidant capacity. The MPP nanoparticles are capable of effectively eliminating ROS in isolated pulmonary artery smooth muscle cells (PASMCs) from PH rats, and significantly reduce PASMC proliferation and migration. In vivo results from a PH model demonstrate that MPP nanoparticles significantly increase pulmonary artery acceleration time, decrease wall thickening and PCNA expression in lung tissues, as evidenced by echocardiograpy, histology and immunoblot analysis. Additionally, MPP nanoparticles treatment improve running capacity, decrease Fulton index, and attenuate right ventricular fibrosis in MCT-PH rats by using treadmill test, picrosirius red, and trichrome Masson staining. Further transcriptomic and biochemical analyses reveal that inhibiting ROS-driven activation of TGF-β1 in the PA is the mechanism by which MPP nanoparticles exert their therapeutic effect. This study provides a novel approach for treating PH with non-metallic nanozymes based on a well-understood mechanism.
Collapse
Affiliation(s)
- Ruxia Liu
- Department of Rehabilitation, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ting Zhou
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of CT, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, 710068, China
| | - Xinsheng Li
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Quan Zou
- School of Medical Imaging, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, 300203, China
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jiaojiao Yu
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| | - Jingjing Ye
- Trauma Treatment Center, Peking University People's Hospital, Key Laboratory of Trauma Treatment and Neural Regeneration (Peking University) Ministry of Education, National Center for Trauma Medicine, Beijing, 100044, China
| | - Wenhui Wang
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yan Zhou
- Department of Ultrasound, Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, 300203, China
| |
Collapse
|
11
|
Rao RJ, Chan SY. Mediating Metabolism: Inhibition of Malic Enzyme 1 (ME1) Restores Endothelial Bioenergetics and Adenosine Signaling in Pulmonary Hypertension. Circulation 2024; 149:1372-1374. [PMID: 38648276 PMCID: PMC11045165 DOI: 10.1161/circulationaha.124.068738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Affiliation(s)
- Rashmi J. Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|