1
|
Zhou QY, Pan JQ, Liu W, Jiang ZT, Gao FY, Zhao ZW, Tang CK. Angiotensin II: A novel biomarker in vascular diseases. Clin Chim Acta 2025; 568:120154. [PMID: 39855324 DOI: 10.1016/j.cca.2025.120154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
The renin-angiotensin system (RAS), composed mainly of renin, angiotensin, and aldosterone, is a key endocrine pathway involved in cardiovascular activity regulation. Under physiological conditions, the RAS plays a vital role in water and salt metabolism, blood pressure regulation, and electrolyte balance. Angiotensin II (Ang II) is the most important active component of the RAS, and its receptors are concentrated in vascular, pulmonary, cardiac, and renal tissues in vivo. Moreover, Ang II is closely associated with the development of vascular lesions. Ang II expression is closely associated with atherosclerosis, aortic aneurysm/dissection, ischemic stroke, hypertension, pulmonary hypertension, and type 2 diabetes mellitus. Given the significant pathophysiological role of Ang II in vascular diseases and the availability of advanced detection methods, Ang II holds promise as a reliable biomarker and therapeutic target in clinical settings. This review summarizes the mechanisms through which Ang II contributes to different vascular diseases and discusses its potential application as a biomarker for disease diagnosis.
Collapse
Affiliation(s)
- Qin-Yi Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, PR China; The Affiliated Nanhua Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang 421002 Hunan, PR China
| | - Jin-Qian Pan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, PR China
| | - Wang Liu
- The Affiliated Nanhua Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, China
| | - Zhen-Tao Jiang
- The Affiliated Nanhua Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang 421002 Hunan, PR China
| | - Fang-Ya Gao
- The Affiliated Nanhua Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang 421002 Hunan, PR China
| | - Zhen-Wang Zhao
- School of Basic Medicine, Health Science Center, Hubei University of Arts and Science, Xiangyang, Hubei 441053, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, PR China.
| |
Collapse
|
2
|
Xiong P, Huang Q, Mao Y, Qian H, Yang Y, Mou Z, Deng X, Wang G, He B, You Z. Identification of an immune-related gene panel for the diagnosis of pulmonary arterial hypertension using bioinformatics and machine learning. Int Immunopharmacol 2025; 144:113694. [PMID: 39616855 DOI: 10.1016/j.intimp.2024.113694] [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: 04/07/2024] [Revised: 11/03/2024] [Accepted: 11/20/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE This study aimed to screen an immune-related gene (IRG) panel and develop a novel approach for diagnosing pulmonary arterial hypertension (PAH) utilizing bioinformatics and machine learning (ML). METHODS Gene expression profiles were retrieved from the Gene Expression Omnibus (GEO) database to identify differentially expressed immune-related genes (IRG-DEGs). We employed five machine learning algorithms-LASSO, random forest (RF), boosted regression trees (BRT), XGBoost, and support vector machine recursive feature elimination (SVM-RFE) to identify biomarkers derived from IRG-DEGs associated with the diagnosis of PAH, incorporating them into the IRG-DEGs panel. Validation of these biomarker levels in lung tissue was conducted in a hypoxia-induced mouse model of PAH, investigating the correlation between AIMP1, IL-15, GLRX, SOD1, Fulton's index (RVHI), and the ratio of pulmonary artery medial thickness to external diameter (MT%). Subsequently, we developed a nomogram model based on the IRG-DEGs panel in lung tissue for diagnosing PAH. The expression, distribution, and pseudotime analysis of these biomarkers across various immune cell types were assessed using single-cell sequencing datasets. Finally, we evaluated the diagnostic utility of the nomogram model based on the IRG-DEGs panel in peripheral blood mononuclear cells (PBMCs) for diagnosing PAH. RESULTS A total of 36 upregulated and 17 downregulated IRG-DEGs were identified in lung tissue from patients with PAH. AIMP1, IL-15, GLRX, and SOD1 were subsequently selected as novel immune-related biomarkers for PAH through the aforementioned machine learning algorithms and incorporated into the IRG-DEGs panel. Experimental results from mice with PAH validated that the expression levels of AIMP1, IL-15, and GLRX in lung tissue were elevated, while SOD1 expression was significantly reduced. Additionally, GLRX and AIMP1 exhibited positive correlations with Fulton's index (RVHI). The expression levels of GLRX, IL-15, and AIMP1 showed positive correlations with MT%, whereas SOD1 exhibited negative correlations with MT%. Analysis of single-cell sequencing data further revealed that the levels of IRG-DEG panel members gradually increased during the pseudotime trajectory from PBMCs to macrophages, correlating with macrophage activation. The area under the curve (AUC) for diagnosing PAH using a nomogram model based on the IRG-DEGs panel derived from lung tissue samples and PBMCs was ≥0.969 and 0.900, respectively. CONCLUSIONS We developed an IRG-DEGs panel containing AIMP1, IL-15, GLRX, and SOD1, which may facilitate the diagnosis of pulmonary arterial hypertension (PAH). These findings provide novel insights that may enhance diagnostic and therapeutic approaches for PAH.
Collapse
Affiliation(s)
- Pan Xiong
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Qiuhong Huang
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yang Mao
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Hang Qian
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yi Yang
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Ziye Mou
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiaohui Deng
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Guansong Wang
- Institute of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Binfeng He
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| | - Zaichun You
- Department of General Practice, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China.
| |
Collapse
|
3
|
Rao RJ, Yang J, Jiang S, El-Khoury W, Hafeez N, Okawa S, Tai YY, Tang Y, Aaraj YA, Sembrat JC, Chan SY. Post-transcriptional regulation of IFI16 promotes inflammatory endothelial pathophenotypes observed in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2025; 328:L148-L158. [PMID: 39657959 DOI: 10.1152/ajplung.00048.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease driven by endothelial cell inflammation and dysfunction, resulting in the pathological remodeling of the pulmonary vasculature. Innate immune activation has been linked to PAH development; however, the regulation, propagation, and reversibility of the induction of inflammation in PAH are poorly understood. Here, we demonstrate the role of interferon-inducible protein 16 (IFI16), an innate immune sensor, as a modulator of endothelial inflammation in pulmonary hypertension, using human pulmonary artery endothelial cells (PAECs). Inflammatory stimulus of PAECs with IL-1β upregulates IFI16 expression, inducing proinflammatory cytokine upregulation and cellular apoptosis. IFI16 mRNA stability is regulated by post-transcriptional m6A modification, mediated by Wilms' tumor 1-associated protein (WTAP), a structural stabilizer of the methyltransferase complex, via regulation of m6A methylation of IFI16. In addition, m6A levels are increased in the peripheral blood mononuclear cells of patients with PAH compared with control, indicating that quantifying this epigenetic change in patients may hold potential as a biomarker for disease identification. In summary, our study demonstrates that IFI16 mediates inflammatory endothelial pathophenotypes seen in pulmonary arterial hypertension.NEW & NOTEWORTHY Our work establishes a paradigm of the regulatory role of the Wilms' tumor 1-associated protein (WTAP)-interferon inducible protein 16 (IFI16) axis that uses m6A RNA methylation to drive endothelial inflammatory activation in pulmonary hypertension. Consequently, because m6A epigenetic modifications are both reversible and dynamic, this axis is an attractive diagnostic and therapeutic target in pulmonary hypertension and more broadly in endothelial immune activation.
Collapse
Affiliation(s)
- Rashmi J Rao
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Jimin Yang
- Department of Molecular Biology, Jeonbuk National University, Jeonju, South Korea
| | - Siyi Jiang
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Wadih El-Khoury
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Neha Hafeez
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Satoshi Okawa
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Yi Yin Tai
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Ying Tang
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - Yassmin Al Aaraj
- 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| | - John C Sembrat
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - 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 and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
4
|
Huang Y, Zhang J, Zhao Q, Hu X, Zhao H, Wang S, Wang L, Jiang R, Wu W, Liu J, Yuan P, Gong S. Impact of reduced apolipoprotein A-I levels on pulmonary arterial hypertension. Hellenic J Cardiol 2024; 80:31-46. [PMID: 37940001 DOI: 10.1016/j.hjc.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/27/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023] Open
Abstract
OBJECTIVE The significance of apolipoprotein A-I (ApoA-I) is the anti-inflammatory functional component of high-density lipoprotein, which needs to be further studied in relation to pulmonary arterial hypertension (PAH). This study aimed to identify the predictive value of ApoA-1 on the risk and prognosis of PAH, as well as the underlying anti-inflammatory mechanism. METHODS Proteomic analysis was conducted on lung tissue from 6 PAH patients and 4 lung donors. Prediction of risk and mortality risk factors associated with PAH in 343 patients used logistic analysis and Cox regression analysis, respectively. The protective function of ApoA-I was assessed in human pulmonary arterial endothelial cells (HPAEC), while its anti-inflammatory function was evaluated in THP-1 macrophages. RESULTS In the lung tissues of patients with PAH, 168 differentially expressed proteins were associated with lipid metabolism according to GO and KEGG enrichment analysis. A protein-protein interaction network identified ApoA-I as a key protein associated with PAH. Lower ApoA-I levels were independent risk factors for PAH and displayed a stronger predictive value for PAH mortality. Plasma interleukin 6 (IL-6) levels were positively correlated with risk stratification and were higher in PAH patients with lower ApoA-I levels. ApoA-I was downregulated in the lung tissues of monocrotaline (MCT) -induced rats. ApoA-I could reduce the IL-6-induced pro-proliferative and pro-migratory abilities of HPAEC and inhibit the secretion of IL-6 from macrophages, which is compromised under hypoxic conditions. CONCLUSION Our study identified the significance of ApoA-I as a biomarker for predicting the survival outcome of PAH patients, which might relate to its altered anti-inflammatory properties.
Collapse
Affiliation(s)
- Yuxia Huang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Ji Zhang
- Department of Lung Transplantation, First Affiliated Hospital, School of Medical, Zhejiang University, Hangzhou 310000, China
| | - Qinhua Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Xiaoyi Hu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Hui Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China; Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200000, China
| | - Shang Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Wenhui Wu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Jinming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China.
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China.
| | - Sugang Gong
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China.
| |
Collapse
|
5
|
Prasad JD, Williams TJ, Whitford HM. Pulmonary arterial hypertension: updates and perspective with newer therapies. Intern Med J 2024. [PMID: 39440807 DOI: 10.1111/imj.16515] [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: 04/18/2024] [Accepted: 08/15/2024] [Indexed: 10/25/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare condition for which a remarkable change has been witnessed in the epidemiology, assessment and treatment landscape over the last three decades. Well-established registries from the Western world have not only highlighted the shift in the epidemiology to an older, more comorbid cohort but have also identified markers of prognosis that have been validated as part of risk stratification scores in multiple cohorts. The emphasis on early identification through a systematic assessment pathway and the option of upfront combination therapy with serial risk stratification assessment has laid the foundation for the standard of care and improved prognosis. This review provides an update on the assessment and newer therapies for PAH.
Collapse
Affiliation(s)
- Jyotika D Prasad
- Department of Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Trevor J Williams
- Department of Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
| | - Helen M Whitford
- Department of Respiratory Medicine, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Guignabert C, Aman J, Bonnet S, Dorfmüller P, Olschewski AJ, Pullamsetti S, Rabinovitch M, Schermuly RT, Humbert M, Stenmark KR. Pathology and pathobiology of pulmonary hypertension: current insights and future directions. Eur Respir J 2024; 64:2401095. [PMID: 39209474 PMCID: PMC11533988 DOI: 10.1183/13993003.01095-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
In recent years, major advances have been made in the understanding of the cellular and molecular mechanisms driving pulmonary vascular remodelling in various forms of pulmonary hypertension, including pulmonary arterial hypertension, pulmonary hypertension associated with left heart disease, pulmonary hypertension associated with chronic lung disease and hypoxia, and chronic thromboembolic pulmonary hypertension. However, the survival rates for these different forms of pulmonary hypertension remain unsatisfactory, underscoring the crucial need to more effectively translate innovative scientific knowledge into healthcare interventions. In these proceedings of the 7th World Symposium on Pulmonary Hypertension, we delve into recent developments in the field of pathology and pathophysiology, prioritising them while questioning their relevance to different subsets of pulmonary hypertension. In addition, we explore how the latest omics and other technological advances can help us better and more rapidly understand the myriad basic mechanisms contributing to the initiation and progression of pulmonary vascular remodelling. Finally, we discuss strategies aimed at improving patient care, optimising drug development, and providing essential support to advance research in this field.
Collapse
Affiliation(s)
- Christophe Guignabert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Sébastien Bonnet
- Pulmonary Hypertension research group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Dorfmüller
- Department of Pathology, University Hospital Giessen/Marburg, Giessen, Germany
| | - Andrea J Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research Bad Nauheim, Bad Nauheim, Germany
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
- Universities of Giessen and Marburg Lung Centre, Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Marlene Rabinovitch
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ralph T Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
| | - Marc Humbert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver, CO, USA
| |
Collapse
|
7
|
Rao RJ, Yang J, Jiang S, El-Khoury W, Hafeez N, Okawa S, Tai YY, Tang Y, Al Aaraj Y, Sembrat J, Chan SY. Post-transcriptional regulation of IFI16 promotes inflammatory endothelial pathophenotypes observed in pulmonary arterial hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613988. [PMID: 39345560 PMCID: PMC11429958 DOI: 10.1101/2024.09.19.613988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease driven by endothelial cell inflammation and dysfunction, resulting in the pathological remodeling of the pulmonary vasculature. Innate immune activation has been linked to PAH development; however, the regulation, propagation, and reversibility of the induction of inflammation in PAH is poorly understood. Here, we demonstrate a role for interferon inducible protein 16 (IFI16), an innate immune sensor, as a modulator of endothelial inflammation in pulmonary hypertension, utilizing human pulmonary artery endothelial cells (PAECs). Inflammatory stimulus of PAECs with IL-1β up-regulates IFI16 expression, inducing proinflammatory cytokine up-regulation and cellular apoptosis. IFI16 mRNA stability is regulated by post-transcriptional m6A modification, mediated by Wilms' tumor 1-associated protein (WTAP), a structural stabilizer of the methyltransferase complex, via regulation of m6A methylation of IFI16. Additionally, m6A levels are increased in the peripheral blood mononuclear cells of PAH patients compared to control, indicating that quantifying this epigenetic change in patients may hold potential as a biomarker for disease identification. In summary, our study demonstrates IFI16 mediates inflammatory endothelial pathophenotypes seen in pulmonary arterial hypertension.
Collapse
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
| | - Jimin Yang
- Department of Molecular Biology, Jeonbuk National University, Jeonju, South Korea
| | - Siyi Jiang
- 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
| | - Wadih El-Khoury
- 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
| | - Neha Hafeez
- 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
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Satoshi Okawa
- 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
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yi Yin Tai
- 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
| | - Ying Tang
- 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
| | - Yassmin Al Aaraj
- 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
| | - John Sembrat
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, 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, 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
|
8
|
Ghouleh IA, Rocha AP, Goncharova EA. New Kid on the Block: GLI1+ Cells and Pulmonary Arterioles Neomuscularization. Circ Res 2024; 134:1402-1404. [PMID: 38781304 PMCID: PMC11125530 DOI: 10.1161/circresaha.124.324574] [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: 05/25/2024]
Affiliation(s)
- Imad Al Ghouleh
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute; Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Andres Pulgarin Rocha
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute; Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, UC Davis, Davis, CA, USA
| |
Collapse
|
9
|
Xue H, Lan X, Xue T, Tang X, Yang H, Hu Z, Xu N, Xie B. PD-1 + T lymphocyte proportions and hospitalized exacerbation of COPD: a prospective cohort study. Respir Res 2024; 25:218. [PMID: 38789950 PMCID: PMC11127417 DOI: 10.1186/s12931-024-02847-6] [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: 01/12/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVE To evaluate the predictive value of PD-1 expression in T lymphocytes for rehospitalization due to acute exacerbations of COPD (AECOPD) in discharged patients. METHODS 115 participants hospitalized with COPD (average age 71.8 ± 6.0 years) were recruited at Fujian Provincial Hospital. PD1+T lymphocytes proportions (PD1+T%), baseline demographics and clinical data were recorded at hospital discharge. AECOPD re-admission were collected at 1-year follow-up. Kaplan-Meier analysis compared the time to AECOPD readmissions among groups stratified by PD1+T%. Multivariable Cox proportional hazards regression and stratified analysis determined the correlation between PD1+T%, potential confounders, and AECOPD re-admission. ROC and DCA evaluated PD1+T% in enhancing the clinical predictive values of Cox models, BODE and CODEX. RESULTS 68 participants (59.1%) were AECOPD readmitted, those with AECOPD readmission exhibited significantly elevated baseline PD-1+CD4+T/CD4+T% and PD-1+CD8 + T/CD8 + T% compared to non-readmitted counterparts. PD1+ T lymphocyte levels statistically correlated with BODE and CODEX indices. Kaplan-Meier analysis demonstrated that those in Higher PD1+ T lymphocyte proportions had reduced time to AECOPD readmission (logRank p < 0.05). Cox analysis identified high PD1+CD4+T and PD1+CD8+T ratios as risk factors of AECOPD readmission, with hazard ratios of 1.384(95%CI [1.043-1.725]) and 1.401(95%CI [1.013-1.789]), respectively. Notably, in patients aged < 70 years and with fewer than twice AECOPD episodes in the previous year, high PD1+T lymphocyte counts significantly increased risk for AECOPD readmission(p < 0.05). The AECOPD readmission predictive model, incorporating PD1+T% exhibited superior discrimination to the Cox model, BODE index and CODEX index, AUC of ROC were 0.763(95%CI [0.633-0.893]) and 0.734(95%CI [0.570-0.899]) (DeLong's test p < 0.05).The DCA illustrates that integrating PD1+T% into models significantly enhances the utility in aiding clinical decision-making. CONCLUSION Evaluation of PD1+ lymphocyte proportions offer a novel perspective for identifying high-risk COPD patients, potentially providing insights for COPD management. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR, URL: www.chictr.org.cn/ ), Registration number: ChiCTR2200055611 Date of Registration: 2022-01-14.
Collapse
Affiliation(s)
- Hong Xue
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Xiuyan Lan
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Ting Xue
- Center of Health Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Xuwei Tang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xuefu north Road, Fuzhou, 350122, China
| | - Haitao Yang
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xuefu north Road, Fuzhou, 350122, China
| | - Nengluan Xu
- Department of Infectious Diseases, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No.516 Jinrong South Street, Fuzhou, Fujian, 350001, China.
| | - Baosong Xie
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
10
|
Juaiti M, Feng Y, Tang Y, Liang B, Zha L, Yu Z. Integrated bioinformatics analysis and experimental animal models identify a robust biomarker and its correlation with the immune microenvironment in pulmonary arterial hypertension. Heliyon 2024; 10:e29587. [PMID: 38660271 PMCID: PMC11040037 DOI: 10.1016/j.heliyon.2024.e29587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/26/2024] Open
Abstract
Background Pulmonary arterial hypertension (PAH) represents a substantial global risk to human health. This study aims to identify diagnostic biomarkers for PAH and assess their association with the immune microenvironment through the utilization of sophisticated bioinformatics techniques. Methods Based on two microarray datasets, differentially expressed genes (DEGs) were detected, and hub genes underwent a sequence of machine learning analyses. After pathways associated with PAH were assessed by gene enrichment analysis, the identified genes were validated using external datasets and confirmed in a monocrotaline (MCT)-induced rat model. In addition, three algorithms were employed to estimate the proportions of various immune cell types, and the link between hub genes and immune cells was substantiated. Results Using SVM, LASSO, and WGCNA, we identified seven hub genes, including (BPIFA1, HBA2, HBB, LOC441081, PI15, S100A9, and WIF1), of which only BPIFA1 remained stable in the external datasets and was validated in an MCT-induced rat model. Furthermore, the results of the functional enrichment analysis established a link between PAH and both metabolism and the immune system. Correlation assessment showed that BPIFA1 expression in the MCP-counter algorithm was negatively associated with various immune cell types, positively correlated with macrophages in the ssGSEA algorithm, and correlated with M1 and M2 macrophages in the CIBERSORT algorithm. Conclusion BPIFA1 serves as a modulator of PAH, with the potential to impact the immune microenvironment and disease progression, possibly through its regulatory influence on both M1 and M2 macrophages.
Collapse
Affiliation(s)
- Mukamengjiang Juaiti
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yilu Feng
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yiyang Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Benhui Liang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Lihuang Zha
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zaixin Yu
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| |
Collapse
|
11
|
Wang J, Kuang J, Zhang S, Liu Z, Guo Q, Li S, Qiu L, Fu G, Lin X, Wu J, Tian J, Huang J, Niu Y, Kang K, Zhang Y, Gou D. Comprehensive characterization of small noncoding RNA profiles in hypoxia-induced pulmonary hypertension (HPH) rat tissues. iScience 2024; 27:108815. [PMID: 38322991 PMCID: PMC10844824 DOI: 10.1016/j.isci.2024.108815] [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: 07/24/2023] [Revised: 11/10/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Hypoxia-induced pulmonary hypertension (HPH) is a fatal cardiovascular disease characterized by an elevation in pulmonary artery pressure, resulting in right ventricular dysfunction and eventual heart failure. Exploring the pathogenesis of HPH is crucial, and small noncoding RNAs (sncRNAs) are gaining recognition as potential regulators of cellular responses to hypoxia. In this study, we conducted a comprehensive analysis of sncRNA profiles in eight tissues of male HPH rats using high-throughput sequencing. Our study unveiled several sncRNAs, with the brain, kidney, and spleen exhibiting the highest abundance of microRNA (miRNA), tRNA-derived small RNA (tDR), and small nucleolar RNA (snoRNA), respectively. Moreover, we identified numerous tissue-specific and hypoxia-responsive sncRNAs, particularly miRNAs and tDRs. Interestingly, we observed arm switching in miRNAs under hypoxic conditions and a significant increase in the abundance of 5' tRNA-halves among the total tDRs during hypoxia. Overall, our study provides a comprehensive characterization of the sncRNA profiles in HPH rats.
Collapse
Affiliation(s)
- Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Jiahao Kuang
- College of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Shasha Zhang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Zixin Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Qianwen Guo
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Shujin Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Lin Qiu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Gaohui Fu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Xinyang Lin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Jiayu Wu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Jinglin Tian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Jinyong Huang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Kang Kang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
- College of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650022, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
12
|
Dai L, Chen Y, Wu J, He Z, Zhang Y, Zhang W, Xie Y, Zeng H, Zhong X. A novel complement C3 inhibitor CP40-KK protects against experimental pulmonary arterial hypertension via an inflammasome NLRP3 associated pathway. J Transl Med 2024; 22:164. [PMID: 38365806 PMCID: PMC10870435 DOI: 10.1186/s12967-023-04741-z] [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: 07/07/2023] [Accepted: 11/20/2023] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a severe cardiopulmonary disease characterized by complement dependent and proinflammatory activation of macrophages. However, effective treatment for complement activation in PAH is lacking. We aimed to explore the effect and mechanism of CP40-KK (a newly identified analog of selective complement C3 inhibitor CP40) in the PAH model. METHODS We used western blotting, immunohistochemistry, and immunofluorescence staining of lung tissues from the monocrotaline (MCT)-induced rat PAH model to study macrophage infiltration, NLPR3 inflammasome activation, and proinflammatory cytokines (IL-1β and IL-18) release. Surface plasmon resonance (SPR), ELISA, and CH50 assays were used to test the affinity between CP40-KK and rat/human complement C3. CP40-KK group rats only received CP40-KK (2 mg/kg) by subcutaneous injection at day 15 to day 28 continuously. RESULTS C3a was significantly upregulated in the plasma of MCT-treated rats. SPR, ELISA, and CH50 assays revealed that CP40-KK displayed similar affinity binding to human and rat complement C3. Pharmacological inhibition of complement C3 cleavage (CP40-KK) could ameliorate MCT-induced NLRP3 inflammasome activity, pulmonary vascular remodeling, and right ventricular hypertrophy. Mechanistically, increased proliferation of pulmonary arterial smooth muscle cells is closely associated with macrophage infiltration, NLPR3 inflammasome activation, and proinflammatory cytokines (IL-1β and IL-18) release. Besides, C3a enhanced IL-1β activity in macrophages and promoted pulmonary arterial smooth muscle cell proliferation in vitro. CONCLUSION Our findings suggest that CP40-KK treatment was protective in the MCT-induced rat PAH model, which might serve as a therapeutic option for PAH.
Collapse
Affiliation(s)
- Lei Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Yu Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Jinhua Wu
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, 530000, Guangxi, China
| | - Zhen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Yueqi Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Wenjun Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Yang Xie
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China
| | - Hesong Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China.
| | - Xiaodan Zhong
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Provincial Engineering Research Center of Vascular Interventional Therapy, Wuhan, 430030, Hubei, China.
| |
Collapse
|
13
|
Ferrian S, Cao A, McCaffrey EF, Saito T, Greenwald NF, Nicolls MR, Bruce T, Zamanian RT, Del Rosario P, Rabinovitch M, Angelo M. Single-Cell Imaging Maps Inflammatory Cell Subsets to Pulmonary Arterial Hypertension Vasculopathy. Am J Respir Crit Care Med 2024; 209:206-218. [PMID: 37934691 PMCID: PMC10806425 DOI: 10.1164/rccm.202209-1761oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/07/2023] [Indexed: 11/09/2023] Open
Abstract
Rationale: Unraveling immune-driven vascular pathology in pulmonary arterial hypertension (PAH) requires a comprehensive understanding of the immune cell landscape. Although patients with hereditary (H)PAH and bone morphogenetic protein receptor type 2 (BMPR2) mutations have more severe pulmonary vascular pathology, it is not known whether this is related to specific immune cell subsets. Objectives: This study aims to elucidate immune-driven vascular pathology by identifying immune cell subtypes linked to severity of pulmonary arterial lesions in PAH. Methods: We used cutting-edge multiplexed ion beam imaging by time of flight to compare pulmonary arteries (PAs) and adjacent tissue in PAH lungs (idiopathic [I]PAH and HPAH) with unused donor lungs, as controls. Measurements and Main Results: We quantified immune cells' proximity and abundance, focusing on those features linked to vascular pathology, and evaluated their impact on pulmonary arterial smooth muscle cells (SMCs) and endothelial cells. Distinct immune infiltration patterns emerged between PAH subtypes, with intramural involvement independently linked to PA occlusive changes. Notably, we identified monocyte-derived dendritic cells within PA subendothelial and adventitial regions, influencing vascular remodeling by promoting SMC proliferation and suppressing endothelial gene expression across PAH subtypes. In patients with HPAH, pronounced immune dysregulation encircled PA walls, characterized by heightened perivascular inflammation involving T cell immunoglobulin and mucin domain-3 (TIM-3)+ T cells. This correlated with an expanded DC subset expressing indoleamine 2,3-dioxygenase 1, TIM-3, and SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1, alongside increased neutrophils, SMCs, and alpha-smooth muscle actin (ACTA2)+ endothelial cells, reinforcing the heightened severity of pulmonary vascular lesions. Conclusions: This study presents the first architectural map of PAH lungs, connecting immune subsets not only with specific PA lesions but also with heightened severity in HPAH compared with IPAH. Our findings emphasize the therapeutic potential of targeting monocyte-derived dendritic cells, neutrophils, cellular interactions, and immune responses to alleviate severe vascular pathology in IPAH and HPAH.
Collapse
Affiliation(s)
- Selena Ferrian
- Department of Pathology
- Early Clinical Development Informatics, Genentech Inc., South San Francisco, California
| | - Aiqin Cao
- Department of Pediatrics
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
- Basic Science and Engineering (BASE) Initiative, Betty Irene Moore Children’s Heart Center, Stanford, California
| | | | | | | | - Mark R. Nicolls
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California
| | | | - Roham T. Zamanian
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California
| | - Patricia Del Rosario
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Marlene Rabinovitch
- Department of Pediatrics
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
- Basic Science and Engineering (BASE) Initiative, Betty Irene Moore Children’s Heart Center, Stanford, California
- Stanford Cardiovascular Institute, Stanford University, Palo Alto, California
| | | |
Collapse
|
14
|
Jutant EM, Chelgham MK, Ottaviani M, Thuillet R, Le Vely B, Humbert M, Guignabert C, Tu L, Huertas A. Central role of ubiquitin-specific protease 8 in leptin signaling pathway in pulmonary arterial hypertension. J Heart Lung Transplant 2024; 43:120-133. [PMID: 37704159 DOI: 10.1016/j.healun.2023.09.003] [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: 02/10/2023] [Revised: 07/31/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Leptin receptor (ObR-b) is overexpressed in pulmonary artery smooth muscle cells (PA-SMCs) from patients with pulmonary arterial hypertension (PAH) and is implicated in both mechanisms that contribute to pulmonary vascular remodeling: hyperproliferation and inflammation. Our aim was to investigate the role of ubiquitin-specific peptidase 8 (USP8) in ObR-b overexpression in PAH. METHODS We performed in situ and in vitro experiments in human lung specimens and isolated PA-SMCs combined with 2 different in vivo models in rodents and we generated a mouse with an inducible USP8 deletion specifically in smooth muscles. RESULTS Our results showed an upregulation of USP8 in the smooth muscle layer of distal pulmonary arteries from patients with PAH, and upregulation of USP8 expression in PAH PA-SMCs, compared to controls. USP8 inhibition in PAH PA-SMCs significantly blocked both ObR-b protein expression level at the cell surface as well as ObR-b-dependant intracellular signaling pathway as shown by a significant decrease in pSTAT3 expression. USP8 was required for ObR-b activation in PA-SMCs and its inhibition prevented Ob-mediated cell proliferation through STAT3 pathway. USP8 inhibition by the chemical inhibitor DUBs-IN-2 protected against the development of experimental PH in the 2 established experimental models of PH. Targeting USP8 specifically in smooth muscle cells in a transgenic mouse model also protected against the development of experimental PH. CONCLUSIONS Our findings highlight the role of USP8 in ObR-b overexpression and pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Etienne-Marie Jutant
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Mustapha K Chelgham
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Mina Ottaviani
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Benjamin Le Vely
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France
| | - Alice Huertas
- INSERM UMR_S 999, Hôpital Marie Lannelongue, 133 Avenue de la Résistance, 92350 Le Plessis-Robinson, France; Université Paris-Saclay, School of Medicine, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, 78 Rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France.
| |
Collapse
|
15
|
Boucetta H, Zhang L, Sosnik A, He W. Pulmonary arterial hypertension nanotherapeutics: New pharmacological targets and drug delivery strategies. J Control Release 2024; 365:236-258. [PMID: 37972767 DOI: 10.1016/j.jconrel.2023.11.012] [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: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, serious, and incurable disease characterized by high lung pressure. PAH-approved drugs based on conventional pathways are still not exhibiting favorable therapeutic outcomes. Drawbacks like short half-lives, toxicity, and teratogenicity hamper effectiveness, clinical conventionality, and long-term safety. Hence, approaches like repurposing drugs targeting various and new pharmacological cascades and/or loaded in non-toxic/efficient nanocarrier systems are being investigated lately. This review summarizes the status of conventional, repurposed, either in vitro, in vivo, and/or in clinical trials of PAH treatment. In-depth description, discussion, and classification of the new pharmacological targets and nanomedicine strategies with a description of all the nanocarriers that showed promising efficiency in delivering drugs are discussed. Ultimately, an illustration of the different nucleic acids tailored and nanoencapsulated within different types of nanocarriers to restore the pathways affected by this disease is presented.
Collapse
Affiliation(s)
- Hamza Boucetta
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
16
|
Santos EW, Khatoon S, Di Mise A, Zheng YM, Wang YX. Mitochondrial Dynamics in Pulmonary Hypertension. Biomedicines 2023; 12:53. [PMID: 38255160 PMCID: PMC10813473 DOI: 10.3390/biomedicines12010053] [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: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Mitochondria are essential organelles for energy production, calcium homeostasis, redox signaling, and other cellular responses involved in pulmonary vascular biology and disease processes. Mitochondrial homeostasis depends on a balance in mitochondrial fusion and fission (dynamics). Mitochondrial dynamics are regulated by a viable circadian clock. Hypoxia and nicotine exposure can cause dysfunctions in mitochondrial dynamics, increases in mitochondrial reactive oxygen species generation and calcium concentration, and decreases in ATP production. These mitochondrial changes contribute significantly to pulmonary vascular oxidative stress, inflammatory responses, contractile dysfunction, pathologic remodeling, and eventually pulmonary hypertension. In this review article, therefore, we primarily summarize recent advances in basic, translational, and clinical studies of circadian roles in mitochondrial metabolism in the pulmonary vasculature. This knowledge may not only be crucial to fully understanding the development of pulmonary hypertension, but also greatly help to create new therapeutic strategies for treating this devastating disease and other related pulmonary disorders.
Collapse
Affiliation(s)
- Ed Wilson Santos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Subika Khatoon
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona, 4, 70125 Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; (E.W.S.); (S.K.); (A.D.M.)
| |
Collapse
|
17
|
Chen CN, Hajji N, Yeh FC, Rahman S, Ali S, Wharton J, Baxan N, Zhao L, Xie CY, Chen YG, Frid MG, Chelladurai P, Pullamsetti SS, Stenmark KR, Wilkins MR, Zhao L. Restoration of Foxp3 + Regulatory T Cells by HDAC-Dependent Epigenetic Modulation Plays a Pivotal Role in Resolving Pulmonary Arterial Hypertension Pathology. Am J Respir Crit Care Med 2023; 208:879-895. [PMID: 37676930 DOI: 10.1164/rccm.202301-0181oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023] Open
Abstract
Rationale: Immune dysregulation is a common feature of pulmonary arterial hypertension (PAH). Histone deacetylase (HDAC)-dependent transcriptional reprogramming epigenetically modulates immune homeostasis and is a novel disease-oriented approach in modern times. Objectives: To identify a novel functional link between HDAC and regulatory T cells (Tregs) in PAH, aiming to establish disease-modified biomarkers and therapeutic targets. Methods: Peripheral blood mononuclear cells were isolated from patients with idiopathic PAH (IPAH) and rodent models of pulmonary hypertension (PH): monocrotaline rats, Sugen5416-hypoxia rats, and Treg-depleted mice. HDAC inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) was used to examine the immune modulatory effects in vivo, ex vivo, and in vitro. Measurements and Main Results: Increased HDAC expression was associated with reduced Foxp3+ Tregs and increased PD-1 (programmed cell death-1) signaling in peripheral blood mononuclear cells from patients with IPAH. SAHA differentially modified a cluster of epigenetic-sensitive genes and induced Foxp3+ Treg conversion in IPAH T cells. Rodent models recapitulated these epigenetic aberrations and T-cell dysfunction. SAHA attenuated PH phenotypes and restored FOXP3 transcription and Tregs in PH rats; interestingly, the effects were more profound in female rats. Selective depletion of CD25+ Tregs in Sugen5416-hypoxia mice neutralized the effects of SAHA. Furthermore, SAHA inhibited endothelial cytokine/chemokine release upon stimulation and subsequent immune chemotaxis. Conclusions: Our results indicated HDAC aberration was associated with Foxp3+ Treg deficiency and demonstrated an epigenetic-mediated mechanism underlying immune dysfunction in PAH. Restoration of Foxp3+ Tregs by HDAC inhibitors is a promising approach to resolve pulmonary vascular pathology, highlighting the potential benefit of developing epigenetic therapies for PAH.
Collapse
Affiliation(s)
- Chien-Nien Chen
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Nabil Hajji
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Fu-Chiang Yeh
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sunniyat Rahman
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Haematology, University College London Cancer Institute, University College London, London, United Kingdom
| | - Souad Ali
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Nicoleta Baxan
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Lin Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Chong-Yang Xie
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Yi-Guan Chen
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Maria G Frid
- Division of Critical Care Medicine and Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado, Denver, Colorado
| | - Prakash Chelladurai
- Max-Planck Institute for Heart and Lung Research, Member of German Center for Lung Research, Giessen, Germany; and
| | - Soni Savai Pullamsetti
- Max-Planck Institute for Heart and Lung Research, Member of German Center for Lung Research, Giessen, Germany; and
- Institute of Molecular Biology and Tumor Research, Marburg, Germany
| | - Kurt R Stenmark
- Division of Critical Care Medicine and Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado, Denver, Colorado
| | - Martin R Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
18
|
Solinas S, Boucly A, Beurnier A, Kularatne M, Grynblat J, Eyries M, Dorfmüller P, Sitbon O, Humbert M, Montani D. Diagnosis and management of pulmonary veno-occlusive disease. Expert Rev Respir Med 2023; 17:635-649. [PMID: 37578057 DOI: 10.1080/17476348.2023.2247989] [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: 04/26/2023] [Revised: 07/08/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Pulmonary veno-occlusive disease (PVOD) is an orphan disease and uncommon etiology of pulmonary arterial hypertension (PAH) characterized by substantial small pulmonary vein and capillary involvement. AREAS COVERED PVOD, also known as 'PAH with features of venous/capillary involvement' in the current ESC/ERS classification. EXPERT OPINION In recent years, particular risk factors for PVOD have been recognized, including genetic susceptibilities and environmental factors (such as exposure to occupational organic solvents, chemotherapy, and potentially tobacco). The discovery of biallelic mutations in the EIF2AK4 gene as the cause of heritable PVOD has been a breakthrough in understanding the molecular basis of PVOD. Venous and capillary involvement (PVOD-like) has also been reported to be relatively common in connective tissue disease-associated PAH (especially systemic sclerosis), and in rare pulmonary diseases like sarcoidosis and pulmonary Langerhans cell granulomatosis. Although PVOD and pulmonary arterial hypertension (PAH) exhibit similarities, including severe precapillary PH, it is essential to differentiate between them since PVOD has a worse prognosis and requires specific management. Indeed, PVOD patients are characterized by poor response to PAH-approved drugs, which can lead to pulmonary edema and clinical deterioration. Due to the lack of effective treatments, early referral to a lung transplantation center is crucial.
Collapse
Affiliation(s)
- Sabina Solinas
- School of Medicine, Université Paris- Saclay, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires, AP-HP, Hopital Bicetre, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Athénaïs Boucly
- School of Medicine, Université Paris- Saclay, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires, AP-HP, Hopital Bicetre, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Antoine Beurnier
- School of Medicine, Université Paris- Saclay, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, ERN-LUNG, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Mithum Kularatne
- Division of Respiratory Medicine, Department of Medicine, University of Calgary, Calgary, Canada
| | - Julien Grynblat
- School of Medicine, Université Paris- Saclay, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Mélanie Eyries
- Sorbonne Université, Departement de genetique, Assistance Publique- Hopitaux de Paris, Hopital Pitié-Salpetriere, Paris, France
- INSERM UMRS 1166, ICAN- Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Peter Dorfmüller
- Department of Pathology, University of Giessen and Marburg Lung Center, Justus-Liebig University Giessen, Giessen, Germany
| | - Olivier Sitbon
- School of Medicine, Université Paris- Saclay, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires, AP-HP, Hopital Bicetre, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- School of Medicine, Université Paris- Saclay, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires, AP-HP, Hopital Bicetre, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- School of Medicine, Université Paris- Saclay, Paris, France
- Service de Pneumologie et Soins Intensifs Respiratoires, AP-HP, Hopital Bicetre, Paris, France
- INSERM UMRS 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
19
|
Koudstaal T, Boomars KA. Inflammatory biomarkers in pulmonary arterial hypertension: ready for clinical implementation? Eur Respir J 2023; 61:61/3/2300018. [PMID: 36958746 DOI: 10.1183/13993003.00018-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 03/25/2023]
Affiliation(s)
- Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Karin A Boomars
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Boucly A, Tu L, Guignabert C, Rhodes C, De Groote P, Prévot G, Bergot E, Bourdin A, Beurnier A, Roche A, Jevnikar M, Jaïs X, Montani D, Wilkins MR, Humbert M, Sitbon O, Savale L. Cytokines as prognostic biomarkers in pulmonary arterial hypertension. Eur Respir J 2023; 61:2201232. [PMID: 36549710 DOI: 10.1183/13993003.01232-2022] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/12/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Risk stratification and assessment of disease progression in patients with pulmonary arterial hypertension (PAH) are challenged by the lack of accurate disease-specific and prognostic biomarkers. To date, brain natriuretic peptide (BNP) and/or its N-terminal fragment (NT-proBNP) are the only markers for right ventricular dysfunction used in clinical practice, in association with echocardiographic and invasive haemodynamic variables to predict outcome in patients with PAH. METHODS This study was designed to identify an easily measurable biomarker panel in the serum of 80 well-phenotyped PAH patients with idiopathic, heritable or drug-induced PAH at baseline and at first follow-up. The prognostic value of identified cytokines of interest was secondly analysed in an external validation cohort of 125 PAH patients. RESULTS Among the 20 biomarkers studied with the multiplex Ella platform, we identified a three-biomarker panel composed of β-NGF, CXCL9 and TRAIL that were independently associated with prognosis both at the time of PAH diagnosis and at the first follow-up after initiation of PAH therapy. β-NGF and CXCL9 were predictors of death or transplantation, whereas high levels of TRAIL were associated with a better prognosis. Furthermore, the prognostic value of the three cytokines was more powerful for predicting survival than usual non-invasive variables (New York Heart Association Functional Class, 6-min walk distance and BNP/NT-proBNP). The results were validated in a fully independent external validation cohort. CONCLUSION The monitoring of β-NGF, CXCL9 and TRAIL levels in serum should be considered in the management and treatment of patients with PAH to objectively guide therapeutic options.
Collapse
Affiliation(s)
- Athénaïs Boucly
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| | | | - Pascal De Groote
- Université de Lille, Service de Cardiologie, CHU Lille, Institut Pasteur de Lille, Inserm U1167, Lille, France
| | - Grégoire Prévot
- CHU de Toulouse, Hôpital Larrey, Service de Pneumologie, Toulouse, France
| | - Emmanuel Bergot
- Unicaen, UFR Santé, Service de Pneumologie & Oncologie Thoracique, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Arnaud Bourdin
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR_9214, Montpellier, France
| | - Antoine Beurnier
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne Roche
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Mitja Jevnikar
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Xavier Jaïs
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - David Montani
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Marc Humbert
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Olivier Sitbon
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| | - Laurent Savale
- INSERM UMR_S999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- These authors contributed equally to this work
| |
Collapse
|
21
|
Tsigkou V, Oikonomou E, Anastasiou A, Lampsas S, Zakynthinos GE, Kalogeras K, Katsioupa M, Kapsali M, Kourampi I, Pesiridis T, Marinos G, Vavuranakis MA, Tousoulis D, Vavuranakis M, Siasos G. Molecular Mechanisms and Therapeutic Implications of Endothelial Dysfunction in Patients with Heart Failure. Int J Mol Sci 2023; 24:ijms24054321. [PMID: 36901752 PMCID: PMC10001590 DOI: 10.3390/ijms24054321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Heart failure is a complex medical syndrome that is attributed to a number of risk factors; nevertheless, its clinical presentation is quite similar among the different etiologies. Heart failure displays a rapidly increasing prevalence due to the aging of the population and the success of medical treatment and devices. The pathophysiology of heart failure comprises several mechanisms, such as activation of neurohormonal systems, oxidative stress, dysfunctional calcium handling, impaired energy utilization, mitochondrial dysfunction, and inflammation, which are also implicated in the development of endothelial dysfunction. Heart failure with reduced ejection fraction is usually the result of myocardial loss, which progressively ends in myocardial remodeling. On the other hand, heart failure with preserved ejection fraction is common in patients with comorbidities such as diabetes mellitus, obesity, and hypertension, which trigger the creation of a micro-environment of chronic, ongoing inflammation. Interestingly, endothelial dysfunction of both peripheral vessels and coronary epicardial vessels and microcirculation is a common characteristic of both categories of heart failure and has been associated with worse cardiovascular outcomes. Indeed, exercise training and several heart failure drug categories display favorable effects against endothelial dysfunction apart from their established direct myocardial benefit.
Collapse
Affiliation(s)
- Vasiliki Tsigkou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-69-4770-1299
| | - Artemis Anastasiou
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Stamatios Lampsas
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - George E. Zakynthinos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Katsioupa
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Maria Kapsali
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Islam Kourampi
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Theodoros Pesiridis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration General Hospital, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Sotiria Chest Disease Hospital, 11527 Athens, Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Lao TT. The obstetric implications of pulmonary hypertension and lung transplant. Best Pract Res Clin Obstet Gynaecol 2022; 85:70-82. [PMID: 35868979 DOI: 10.1016/j.bpobgyn.2022.06.002] [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: 05/31/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
Pulmonary hypertension (PH) is present when the mean pulmonary artery pressure is elevated to >20 mmHg as determined by right heart catheterization. It is categorized into five groups according to the underlying causes. Its presence is considered a contraindication to pregnancy for which therapeutic termination is usually advised due to grave maternal prognosis and pregnancy outcome. Nevertheless, pregnancies in affected women are increasingly reported, about half of which were unplanned, and two-thirds resulted in live births without increased foetal anomalies, notwithstanding increased risks of preterm birth, low birth weight, pre-eclampsia, other obstetric complications, and medical comorbidities, when managed under a multidisciplinary team in specialized centres. Successful lung transplant, the ultimate treatment for PH and other progressive lung diseases, restores fertility, and pregnancy is increasingly encountered, but there is a higher risk of graft rejection and mortality, compared with recipients of other organs. Preconception assessment is vital in optimizing maternal and pregnancy outcomes.
Collapse
Affiliation(s)
- Terence T Lao
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| |
Collapse
|
23
|
Medrek S, Melendres-Groves L. Evolving nonvasodilator treatment options for pulmonary arterial hypertension. Curr Opin Pulm Med 2022; 28:361-368. [PMID: 35838352 DOI: 10.1097/mcp.0000000000000887] [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: 11/26/2022]
Abstract
PURPOSE OF REVIEW With the establishment of vasodilator therapy as a mainstay of treatment for pulmonary arterial hypertension (PAH), new therapeutic approaches are needed to prevent the development of the vasculopathy associated with this disease. Many studies are currently underway to investigate nonvasodilator treatment options. RECENT FINDINGS Modulation of bone morphogenic protein receptor type 2 (BMPR2) signaling with sotatercept showed promising results in phase 2 studies. Rituximab, an anti-CD20 monoclonal antibody, showed some signal for beneficial effect in patients with scleroderma-associated PAH. Studies evaluating agents including tocilizumab, selonsertib, bardoxolone, 10-nitro-9(E)-enoic acid (CXA-10) and intravenous iron have not shown acceptable efficacy in treating PAH. SUMMARY Pharmacologic approaches for the treatment of PAH include altering of transforming growth factor β/BMPR2 signaling, proliferation via growth factors, immune response, oxidative stress, estrogen signaling, metabolism, and neurohormonal modulation. Other treatment modalities including pulmonary artery nerve denervation, stem cell therapy, and inter-atrial shunt formation are also being explored.
Collapse
Affiliation(s)
- Sarah Medrek
- Division of Pulmonary Critical Care Medicine, University of New Mexico, Albuquerque, New Mexico, USA
| | | |
Collapse
|
24
|
New progress in diagnosis and treatment of pulmonary arterial hypertension. J Cardiothorac Surg 2022; 17:216. [PMID: 36038916 PMCID: PMC9422157 DOI: 10.1186/s13019-022-01947-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/15/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease. Although great progress has been made in its diagnosis and treatment in recent years, its mortality rate is still very significant. The pathophysiology and pathogenesis of PAH are complex and involve endothelial dysfunction, chronic inflammation, smooth muscle cell proliferation, pulmonary arteriole occlusion, antiapoptosis and pulmonary vascular remodeling. These factors will accelerate the progression of the disease, leading to poor prognosis. Therefore, accurate etiological diagnosis, treatment and prognosis judgment are particularly important. Here, we systematically review the pathophysiology, diagnosis, genetics, prognosis and treatment of PAH.
Collapse
|
25
|
Single-Cell RNA-Sequencing Reveals the Active Involvement of Macrophage Polarizations in Pulmonary Hypertension. DISEASE MARKERS 2022; 2022:5398157. [PMID: 36246557 PMCID: PMC9553540 DOI: 10.1155/2022/5398157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022]
Abstract
Background. Sustained hypoxia can trigger a progressive rise in pulmonary artery pressure and cause serious pulmonary diseases. Macrophages play important roles along the progression of pulmonary hypertension. However, the state of macrophage polarization during the early stage of pulmonary hypertension is unclear. Methods. Unlike traditional sequencing method, single-cell sequencing can accurately distinguish among cell types and better understand cell-to-cell relationships. In this study, we investigated the polarization of macrophages in pulmonary hypertension via single-cell RNA-sequencing in a mice hypoxia model, which was then validated in patients with pulmonary hypertension. Results. We identified that the intermittent exposure to hypoxic conditions could lead to the production of more M2-type macrophages than M1-type macrophages in a mouse model. Further validation analysis was performed by analyzing lung tissue of patients with pulmonary hypertension, revealing that the number of disease-associated M2 macrophages was substantially increased. Conclusions. In this study, the active anti-inflammatory response of macrophage involved in pulmonary hypertension has been identified, suggesting that intervention against the polarization of macrophages to the M2 type may be a potential way to reduce chronic pulmonary inflammation, pulmonary vascular remodeling, and artery pressure. Thus, investigation of macrophage polarization associated with hypoxia could help us better understand disease mechanism and craft effective prevention strategies and approaches.
Collapse
|
26
|
Serum of Post-COVID-19 Syndrome Patients with or without ME/CFS Differentially Affects Endothelial Cell Function In Vitro. Cells 2022; 11:cells11152376. [PMID: 35954219 PMCID: PMC9367589 DOI: 10.3390/cells11152376] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
A proportion of COVID-19 reconvalescent patients develop post-COVID-19 syndrome (PCS) including a subgroup fulfilling diagnostic criteria of Myalgic encephalomyelitis/Chronic Fatigue Syndrome (PCS/CFS). Recently, endothelial dysfunction (ED) has been demonstrated in these patients, but the mechanisms remain elusive. Therefore, we investigated the effects of patients’ sera on endothelia cells (ECs) in vitro. PCS (n = 17), PCS/CFS (n = 13), and healthy controls (HC, n = 14) were screened for serum anti-endothelial cell autoantibodies (AECAs) and dysregulated cytokines. Serum-treated ECs were analysed for the induction of activation markers and the release of small molecules by flow cytometry. Moreover, the angiogenic potential of sera was measured in a tube formation assay. While only marginal differences between patient groups were observed for serum cytokines, AECA binding to ECs was significantly increased in PCS/CFS patients. Surprisingly, PCS and PCS/CFS sera reduced surface levels of several EC activation markers. PCS sera enhanced the release of molecules associated with vascular remodelling and significantly promoted angiogenesis in vitro compared to the PCS/CFS and HC groups. Additionally, sera from both patient cohorts induced the release of molecules involved in inhibition of nitric oxide-mediated endothelial relaxation. Overall, PCS and PCS/CFS patients′ sera differed in their AECA content and their functional effects on ECs, i.e., secretion profiles and angiogenic potential. We hypothesise a pro-angiogenic effect of PCS sera as a compensatory mechanism to ED which is absent in PCS/CFS patients.
Collapse
|
27
|
Yang Z, Yu M, Li X, Tu Y, Wang C, Lei W, Song M, Wang Y, Huang Y, Ding F, Hao K, Han X, Ni X, Qu L, Shen Z, Hu S. Retinoic acid inhibits the angiogenesis of human embryonic stem cell-derived endothelial cells by activating FBP1-mediated gluconeogenesis. Stem Cell Res Ther 2022; 13:239. [PMID: 35672803 PMCID: PMC9171939 DOI: 10.1186/s13287-022-02908-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endothelial cells are located in the inner lumen of blood and lymphatic vessels and exhibit the capacity to form new vessel branches from existing vessels through a process called angiogenesis. This process is energy intensive and tightly regulated. Glycolysis is the main energy source for angiogenesis. Retinoic acid (RA) is an active metabolite of vitamin A and exerts biological effects through its receptor retinoic acid receptor (RAR). In the clinic, RA is used to treat acne vulgaris and acute promyelocytic leukemia. Emerging evidence suggests that RA is involved in the formation of the vasculature; however, its effect on endothelial cell angiogenesis and metabolism is unclear. METHODS Our study was designed to clarify the abovementioned effect with human embryonic stem cell-derived endothelial cells (hESC-ECs) employed as a cell model. RESULTS We found that RA inhibits angiogenesis, as manifested by decreased proliferation, migration and sprouting activity. RNA sequencing revealed general suppression of glycometabolism in hESC-ECs in response to RA, consistent with the decreased glycolytic activity and glucose uptake. After screening glycometabolism-related genes, we found that fructose-1,6-bisphosphatase 1 (FBP1), a key rate-limiting enzyme in gluconeogenesis, was significantly upregulated after RA treatment. After silencing or pharmacological inhibition of FBP1 in hESC-ECs, the capacity for angiogenesis was enhanced, and the inhibitory effect of RA was reversed. ChIP-PCR demonstrated that FBP1 is a target gene of RAR. When hESC-ECs were treated with the RAR inhibitor BMS493, FBP1 expression was decreased and the effect of RA on angiogenesis was partially blocked. CONCLUSIONS The inhibitory role of RA in glycometabolism and angiogenesis is RAR/FBP1 dependent, and FBP1 may be a novel therapeutic target for pathological angiogenesis.
Collapse
Affiliation(s)
- Zhuangzhuang Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xuechun Li
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yuanyuan Tu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Min Song
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Yong Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Ying Huang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Fengyue Ding
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Kaili Hao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xinglong Han
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Xuan Ni
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Lina Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
28
|
Huang L, Li H, Huang S, Wang S, Liu Q, Luo L, Gan S, Fu G, Zou P, Chen G, Wu Z. Notopterol Attenuates Monocrotaline-Induced Pulmonary Arterial Hypertension in Rat. Front Cardiovasc Med 2022; 9:859422. [PMID: 35722110 PMCID: PMC9203832 DOI: 10.3389/fcvm.2022.859422] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Current targeted pulmonary arterial hypertension (PAH) therapies have improved lung hemodynamics, cardiac function, and quality of life; however, none of these have reversed the ongoing remodeling of blood vessels. Considering notopterol, a linear furocoumarin extracted from the root of traditional Chinese medicine Qiang-Huo (Notopterygium incisum), had shown the antiproliferative and anti-inflammatory properties in previous studies, we hypothesized that it could play a role in ameliorating PAH. Methods In vivo, we conducted monocrotaline (MCT) induced PAH rats and treated them with notopterol for 3 weeks. Then, the rats were examined by echocardiography and RV catheterization. The heart and lung specimens were harvested for the detection of gross examination, histological examination and expression of inflammatory molecules. In vitro, human pulmonary arterial smooth muscle cells (HPASMCs) were treated with notopterol after hypoxia; then, cell proliferation was assessed by cell counting kit-8 and Edu assay, and cell migration was detected by wound healing assays. Results We found that notopterol improved mortality rate and RV function while reducing right ventricular systolic pressure in MCT-induced PAH rats. Furthermore, notopterol reduced right ventricular hypertrophy and fibrosis, and it also eased pulmonary vascular remodeling and MCT-induced muscularization. In addition, notopterol attenuated the pro-inflammatory factor (IL-1β, IL-6) and PCNA in the lungs of PAH rats. For the cultured HPASMCs subjected to hypoxia, we found that notopterol can inhibit the proliferation and migration of HPASMCs. Conclusion Our studies show that notopterol exerts anti-inflammatory and anti-proliferative effects in the pulmonary arteries, which may contribute to prevention of PAH.
Collapse
Affiliation(s)
- Lin Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huayang Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Suiqing Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shunjun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Quan Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Li Luo
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuangjiao Gan
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Guangguo Fu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - PeiYun Zou
- GuangZhou Janus Biotechnology Co. Ltd., Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Guangxian Chen
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Zhongkai Wu
| |
Collapse
|
29
|
Flores K, Siques P, Brito J, Arribas SM. AMPK and the Challenge of Treating Hypoxic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms23116205. [PMID: 35682884 PMCID: PMC9181235 DOI: 10.3390/ijms23116205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary artery pressure produced by vasoconstriction and hyperproliferative remodeling of the pulmonary artery and subsequent right ventricular hypertrophy (RVH). The search for therapeutic targets for cardiovascular pathophysiology has extended in many directions. However, studies focused on mitigating high-altitude pulmonary hypertension (HAPH) have been rare. Because AMP-activated protein kinase (AMPK) is involved in cardiovascular and metabolic pathology, AMPK is often studied as a potential therapeutic target. AMPK is best characterized as a sensor of cellular energy that can also restore cellular metabolic homeostasis. However, AMPK has been implicated in other pathways with vasculoprotective effects. Notably, cellular metabolic stress increases the intracellular ADP/ATP or AMP/ATP ratio, and AMPK activation restores ATP levels by activating energy-producing catabolic pathways and inhibiting energy-consuming anabolic pathways, such as cell growth and proliferation pathways, promoting cardiovascular protection. Thus, AMPK activation plays an important role in antiproliferative, antihypertrophic and antioxidant pathways in the pulmonary artery in HPH. However, AMPK plays contradictory roles in promoting HPH development. This review describes the main findings related to AMPK participation in HPH and its potential as a therapeutic target. It also extrapolates known AMPK functions to discuss the less-studied HAPH context.
Collapse
Affiliation(s)
- Karen Flores
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
- Correspondence: ; Tel.: +56-572526392
| | - Patricia Siques
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Silvia M. Arribas
- Department of Physiology, University Autonoma of Madrid, 28049 Madrid, Spain;
| |
Collapse
|
30
|
Role of Ion Channel Remodeling in Endothelial Dysfunction Induced by Pulmonary Arterial Hypertension. Biomolecules 2022; 12:biom12040484. [PMID: 35454073 PMCID: PMC9031742 DOI: 10.3390/biom12040484] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction is a key player in advancing vascular pathology in pulmonary arterial hypertension (PAH), a disease essentially characterized by intense remodeling of the pulmonary vasculature, vasoconstriction, endothelial dysfunction, inflammation, oxidative stress, and thrombosis in situ. These vascular features culminate in an increase in pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past years, there has been a great development in our understanding of pulmonary endothelial biology related to the genetic and molecular mechanisms that modulate the endothelial response to direct or indirect injury and how their dysregulation can promote PAH pathogenesis. Ion channels are key regulators of vasoconstriction and proliferative/apoptotic phenotypes; however, they are poorly studied at the endothelial level. The current review will describe and categorize different expression, functions, regulation, and remodeling of endothelial ion channels (K+, Ca2+, Na+, and Cl− channels) in PAH. We will focus on the potential pathogenic role of ion channel deregulation in the onset and progression of endothelial dysfunction during the development of PAH and its potential therapeutic role.
Collapse
|
31
|
Tobal R, Potjewijd J, van Empel VPM, Ysermans R, Schurgers LJ, Reutelingsperger CP, Damoiseaux JGMC, van Paassen P. Vascular Remodeling in Pulmonary Arterial Hypertension: The Potential Involvement of Innate and Adaptive Immunity. Front Med (Lausanne) 2022; 8:806899. [PMID: 35004784 PMCID: PMC8727487 DOI: 10.3389/fmed.2021.806899] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe disease with high morbidity and mortality. Current therapies are mainly focused on vasodilative agents to improve prognosis. However, recent literature has shown the important interaction between immune cells and stromal vascular cells in the pathogenic modifications of the pulmonary vasculature. The immunological pathogenesis of PAH is known as a complex interplay between immune cells and vascular stromal cells, via direct contacts and/or their production of extra-cellular/diffusible factors such as cytokines, chemokines, and growth factors. These include, the B-cell—mast-cell axis, endothelium mediated fibroblast activation and subsequent M2 macrophage polarization, anti-endothelial cell antibodies and the versatile role of IL-6 on vascular cells. This review aims to outline the major pathophysiological changes in vascular cells caused by immunological mechanisms, leading to vascular remodeling, increased pulmonary vascular resistance and eventually PAH. Considering the underlying immunological mechanisms, these mechanisms may be key to halt progression of disease.
Collapse
Affiliation(s)
- Rachid Tobal
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Judith Potjewijd
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Vanessa P M van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Renee Ysermans
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht, Netherlands
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pieter van Paassen
- Division of Nephrology and Clinical and Experimental Immunology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
32
|
Cell-to-Cell Crosstalk: A New Insight into Pulmonary Hypertension. Rev Physiol Biochem Pharmacol 2022; 184:159-179. [PMID: 35380274 DOI: 10.1007/112_2022_70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Pulmonary hypertension (PH) is a disease with high pulmonary arterial pressure, pulmonary vasoconstriction, pulmonary vascular remodeling, and microthrombosis in complex plexiform lesions, but it has been unclear of the exact mechanism of PH. A new understanding of the pathogenesis of PH is occurred and focused on the role of crosstalk between the cells on pulmonary vessels and pulmonary alveoli. It was found that the crosstalks among the endothelial cells, smooth muscle cells, fibroblasts, pericytes, alveolar epithelial cells, and macrophages play important roles in cell proliferation, migration, inflammation, and so on. Therefore, the heterogeneity of multiple pulmonary blood vessels and alveolar cells and tracking the transmitters of cell communication could be conducive to the further insights into the pathogenesis of PH to discover the potential therapeutic targets for PH.
Collapse
|
33
|
Ali Kazem T, Zeylabi F, Filayih Hassan A, Paridar P, Pezeshki SP, Pezeshki SMS. Diabetes mellitus and COVID-19: review of a lethal interaction from the cellular and molecular level to the bedside. Expert Rev Endocrinol Metab 2022; 17:1-19. [PMID: 34781797 DOI: 10.1080/17446651.2022.2002145] [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: 08/20/2021] [Accepted: 10/25/2021] [Indexed: 01/08/2023]
Abstract
INTRODUCTION While the main mode of transmission of coronavirus disease 2019 (COVID-19) is close contact with other individuals, the presence of chronic underlying diseases such as Diabetes Mellitus (DM) increases the chance of hospitalization and mortality rate due to infection. AREAS COVERED To investigate the effects of COVID-19 infection in DM patients, we reviewed literature from Google Scholar search engine and PubMed database from '2013 to 2020' using the terms "COVID-19; SARS-CoV-2; Diabetes mellitus; obesity; Angiotensin-converting enzyme 2; ACE2; Insulin and Metformin. Evidence suggests that COVID-19 exacerbates the course of diabetes. Presence of pro-inflammatory conditions, increased expression of receptors, and more difficult control of glucose levels in diabetics COVID-19 patients are some of the problems that diabetic patients may face. Also, psychological problems caused by the COVID-19 epidemic in diabetic patients is one of the most important problems in these patients, which is less covered. EXPERT OPINION DM is a strong and independent risk factor with a poor prognosis, which increases the risk of COVID-19 infection, the need for emergency services, the rate of hospitalization in the intensive care unit and also increases the mortality rate of COVID-19 patients.
Collapse
Affiliation(s)
| | - Fatemeh Zeylabi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Pouria Paridar
- Islamic Azad University, North-Tehran Branch, Tehran, Iran
| | - Seyedeh Pardis Pezeshki
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mohammad Sadegh Pezeshki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
34
|
Perros F, Humbert M, Dorfmüller P. Smouldering fire or conflagration? An illustrated update on the concept of inflammation in pulmonary arterial hypertension. Eur Respir Rev 2021; 30:30/162/210161. [PMID: 34937704 DOI: 10.1183/16000617.0161-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/20/2021] [Indexed: 11/05/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare condition that is characterised by a progressive increase of pulmonary vascular resistances that leads to right ventricular failure and death, if untreated. The underlying narrowing of the pulmonary vasculature relies on several independent and interdependent biological pathways, such as genetic predisposition and epigenetic changes, imbalance of vasodilating and vasoconstrictive mediators, as well as dysimmunity and inflammation that will trigger endothelial dysfunction, smooth muscle cell proliferation, fibroblast activation and collagen deposition. Progressive constriction of the pulmonary vasculature, in turn, initiates and sustains hypertrophic and maladaptive myocardial remodelling of the right ventricle. In this review, we focus on the role of inflammation and dysimmunity in PAH which is generally accepted today, although existing PAH-specific medical therapies still lack targeted immune-modulating approaches.
Collapse
Affiliation(s)
- Frédéric Perros
- Université Paris-Saclay, School of Medicine, Le Kremlin Bicêtre, France.,INSERM UMR S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), INSERM, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, School of Medicine, Le Kremlin Bicêtre, France.,INSERM UMR S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Dept of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Peter Dorfmüller
- Institut für Pathologie, Universitätklinikum Giessen und Marburg, Giessen, Germany .,Deutsches Zentrum für Lungenforschung (DZL), Giessen, Germany
| |
Collapse
|
35
|
Zhu Y, Sun Y, Zhang S, Li C, Zhao Y, Zhao B, Li G. Xinmai 'an extract enhances the efficacy of sildenafil in the treatment of pulmonary arterial hypertension via inhibiting MAPK signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:594-605. [PMID: 34010580 PMCID: PMC8143608 DOI: 10.1080/13880209.2021.1917629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Xinmai 'an tablet has been used to improve myocardial blood supply. Recently, some compounds from its formula have shown that they can treat pulmonary arterial hypertension (PAH). OBJECTIVE This study investigates the effects of Xinmai 'an extract (XMA) on PAH and further tests the co-therapeutic enhancement with sildenafil (SIL). MATERIALS AND METHODS Pulmonary artery smooth muscle cells were subjected to stimulation with SIL (12.5 μM) and XMA (250 μg/mL) for 48 h. Sprague-Dawley rats were randomly grouped into eight groups (n = 8 per group): (I) control group received saline; (II) MCT group received MCT (60 mg/kg); (III) SIL-Low group received MCT + SIL at 10 mg/kg/day; (IV) SIL-high group received MCT + SIL at 30 mg/kg/day; (V) XMA-High group received MCT + XMA at 251.6 mg/kg/day; (VI) SIL (Low)+XMA (Low) group received SIL (10 mg/kg) + XMA at 62.9 mg/kg/day; (VII) SIL (Low)+XMA (Medium) group received SIL (10 mg/kg) + XMA at 125.8 mg/kg/day; (VIII) SIL (Low)+XMA (High) group received SIL (10 mg/kg) + XMA at 251.6 mg/kg/day. Both XMA and SIL were given by gavage and were maintained daily for 2 weeks. RESULTS XMA could improve SIL's efficacy in the treatment of PAH by decreasing cell viability more effectively at non-cytotoxic concentrations (250 μg/mL) and reducing Right Ventricular Systolic Pressure (RVSP) in PAH rat. Potential mechanisms might at least in part be through activating the MAPK signalling pathway. DISCUSSION AND CONCLUSIONS The combination of XMA and SIL can improve the efficacy of pulmonary hypertension and reduce the dosage of SIL.
Collapse
Affiliation(s)
- Yaolu Zhu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yabin Sun
- Modern Chinese Medicine Institute, Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Company Limited, Guangzhou, China
| | - Shichang Zhang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuyuan Li
- Office of the General Manager, Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Company Limited, Guangzhou, China
| | - Yiwei Zhao
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Boxin Zhao
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guofeng Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
37
|
Xin Z, Wang J, Li S, Sun C, Jiang W, Xin Q, Wang J, Qi T, Li K, Zhang Z, Luan Y. A review of BMP and Wnt signaling pathway in the pathogenesis of pulmonary arterial hypertension. Clin Exp Hypertens 2021; 44:175-180. [PMID: 34821188 DOI: 10.1080/10641963.2021.1996590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disease characterized by a progressive elevation in mean pulmonary arterial pressure. This occurs due to abnormal remodeling of small peripheral lung vasculature resulting in progressive occlusion of the artery lumen that eventually causes right heart failure and death. Current therapeutic options for PAH are limited and focused mainly on reversal of pulmonary vasoconstriction and proliferation of vascular cells. Although these treatments can relieve disease symptoms, PAH remains a progressive lethal disease.Bone morphogenetic proteins (BMPs) and their receptors were required for PAH-induced right ventricular hypertrophy. Emerging data suggest that restoration of BMP type II receptor (BMPR2) signaling in PAH is a promising alternative that could prevent and reverse pulmonary vascular remodeling. BMPR2 mutations have been identified in >70% of familial and roughly 15% of sporadic PAH cases. Wingless (Wnt) are a family of secreted glycoproteins with varying expression patterns and a range of functions, Wnt signaling pathway is divided into canonical signaling pathway and non-canonical signaling pathway. A recent study reports that interaction between BMP and Wnt closely associated with lung development, those cascade coordination regulation stem cell fate which determine lung branching morphogenes. The promoting effect of BMPR2 on proliferation, survival, and motility of endothelial cells was through recruiting Wnts signaling pathway, the interaction between BMP and Wnt closely associated with lung development.Therefore, in this review, we outline the latest advances of BMP and Wnt signaling pathway in the pathogenesis of PAH and disease progression.
Collapse
Affiliation(s)
- Zhihong Xin
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Junfu Wang
- Clinical laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Susu Li
- College of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Chao Sun
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Wan Jiang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Qian Xin
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Jue Wang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Tonggnag Qi
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Kailin Li
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Zhaohua Zhang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Yun Luan
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| |
Collapse
|
38
|
Han Z, Li X, Cui X, Yuan H, Wang H. The roles of immune system and autoimmunity in pulmonary arterial hypertension: A Review. Pulm Pharmacol Ther 2021; 72:102094. [PMID: 34740751 DOI: 10.1016/j.pupt.2021.102094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/08/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disease characterized by increased pulmonary artery pressure which if left untreated, can lead to poor quality of life and ultimately death. It is a group of conditions and includes idiopathic PAH, familial/hereditary PAH and associated PAH. The condition has been studied for many years and its association with the immune system and in particular autoimmunity has been investigated. The mechanisms for the pathobiology of PAH are unclear although research has highlighted the role of adaptive and innate immune systems in its development. Diagnostics and therapeutic approaches range from cytokine treatments to the use of immunomodulating drugs, although there is still scope for improvements in the field. This article discusses the mechanisms linked to PAH, its association with other conditions and recent therapeutic interventions.
Collapse
Affiliation(s)
- Zhijie Han
- Department of Rheumatology and Immunology, Laizhou People's Hospital, Laizhou 261400, Shandong Province, China
| | - Xiujuan Li
- Department of Cardiology, Laizhou People's Hospital, Laizhou 261400,Shandong Province, China
| | - Xiuli Cui
- Department of Cardiology, Laizhou People's Hospital, Laizhou 261400,Shandong Province, China
| | - Hongjuan Yuan
- Department of Cardiology, Laizhou People's Hospital, Laizhou 261400,Shandong Province, China
| | - Haiping Wang
- Department of Cardiology, Laizhou People's Hospital, Laizhou 261400,Shandong Province, China.
| |
Collapse
|
39
|
Liu G, Wan N, Liu Q, Chen Y, Cui H, Wang Y, Ren J, Shen X, Lu W, Yu Y, Shen Y, Wang J. Resolvin E1 Attenuates Pulmonary Hypertension by Suppressing Wnt7a/β-Catenin Signaling. Hypertension 2021; 78:1914-1926. [PMID: 34689593 DOI: 10.1161/hypertensionaha.121.17809] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Guizhu Liu
- From the State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (G.L., Y.C., W.L., Y.Y., J.W.)
| | - Naifu Wan
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao tong University School of Medicine, Shanghai, China (N.W.)
| | - Qian Liu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Q.L., Y.W., Y.Y., Y.S.)
| | - Yuqin Chen
- From the State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (G.L., Y.C., W.L., Y.Y., J.W.)
| | - Hui Cui
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China (H.C., X.S.)
| | - Yuanyang Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Q.L., Y.W., Y.Y., Y.S.)
| | - Jiaoqi Ren
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China (J.R.)
| | - Xia Shen
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China (H.C., X.S.).,CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (X.S., Y.Y.)
| | - Wenju Lu
- From the State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (G.L., Y.C., W.L., Y.Y., J.W.)
| | - Ying Yu
- From the State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (G.L., Y.C., W.L., Y.Y., J.W.).,Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Q.L., Y.W., Y.Y., Y.S.).,CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China (X.S., Y.Y.)
| | - Yujun Shen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China (Q.L., Y.W., Y.Y., Y.S.)
| | - Jian Wang
- From the State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China (G.L., Y.C., W.L., Y.Y., J.W.).,Department of Medicine, University of California, San Diego, La Jolla (J.W.)
| |
Collapse
|
40
|
Jian M, He S, Liu Y, Liu X, Gui J, Zheng M, Feng B, Zhang X, Liu C. The high-risk factors of different severities of bronchopulmonary dysplasia (BPD) based on the national institute of child health and human development (NICHD) diagnosis criteria in 2018. J Bras Pneumol 2021; 47:e20210125. [PMID: 34614093 PMCID: PMC8642818 DOI: 10.36416/1806-3756/e20210125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/19/2021] [Indexed: 11/29/2022] Open
Abstract
Objective To investigate the clinical characteristics of preterm infants with different severities of bronchopulmonary dysplasia (BPD) and disclose the high-risk factors of exacerbating BPD. Methods Collection of clinical data of 91 preterm infants admitted to the NICU and diagnosed with BPD, categorized in groups according to the disease severity: 41 mild cases,, 24 moderate cases, and 26 severe cases. Comparison and analysis of perinatal risk factors, treatment, complications and prognosis of the infants with different severity degrees. Results The severe group had a higher proportion of infants with congenital heart disease (CHD) higher than the moderate group (P < 0.05), and a higher ratio of pneumonia and mechanical ventilation (MV) ≥ seven days than the mild group (P < 0.05). The severe group also presented higher reintubation incidence than both the mild and moderate groups (P < 0.05). The groups presented different (P < 0.05) incidence rates of hemodynamically significant patent ductus arteriosus (hsPDA) . Ridit analysis suggested that the premature infants (PIs) with hsPDA, multiple microbial pulmonary infections, or Klebsiella pneumoniae pneumonia had more severe illness. Conclusion CHD, hsPDA, MV ≥ seven days, reintubation, pneumonia, especially multiple microbial pulmonary infections, and Klebsiella pneumoniae pneumonia are correlated with the severity of BPD and can be used as BPD progression predictor.
Collapse
Affiliation(s)
- Minqiao Jian
- Department of Clinical Medicine, School of Medicine, South China University of Technology, Guangzhou, China.,Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shaoru He
- Department of Clinical Medicine, School of Medicine, South China University of Technology, Guangzhou, China.,Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yumei Liu
- Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaoqing Liu
- Department of NICU, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Juan Gui
- Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Manli Zheng
- Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bowen Feng
- Department of NICU, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohui Zhang
- Department of NICU, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Caisheng Liu
- Department of Clinical Medicine, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
41
|
Sweatt AJ, Miyagawa K, Rhodes CJ, Taylor S, Del Rosario PA, Hsi A, Haddad F, Spiekerkoetter E, Bental-Roof M, Bland RD, Swietlik EM, Gräf S, Wilkins MR, Morrell NW, Nicolls MR, Rabinovitch M, Zamanian RT. Severe Pulmonary Arterial Hypertension Is Characterized by Increased Neutrophil Elastase and Relative Elafin Deficiency. Chest 2021; 160:1442-1458. [PMID: 34181952 PMCID: PMC8546243 DOI: 10.1016/j.chest.2021.06.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Preclinical evidence implicates neutrophil elastase (NE) in pulmonary arterial hypertension (PAH) pathogenesis, and the NE inhibitor elafin is under early therapeutic investigation. RESEARCH QUESTION Are circulating NE and elafin levels abnormal in PAH and are they associated with clinical severity? STUDY DESIGN AND METHODS In an observational Stanford University PAH cohort (n = 249), plasma NE and elafin levels were measured in comparison with those of healthy control participants (n = 106). NE and elafin measurements were then related to PAH clinical features and relevant ancillary biomarkers. Cox regression models were fitted with cubic spline functions to associate NE and elafin levels with survival. To validate prognostic relationships, we analyzed two United Kingdom cohorts (n = 75 and n = 357). Mixed-effects models evaluated NE and elafin changes during disease progression. Finally, we studied effects of NE-elafin balance on pulmonary artery endothelial cells (PAECs) from patients with PAH. RESULTS Relative to control participants, patients with PAH were found to have increased NE levels (205.1 ng/mL [interquartile range (IQR), 123.6-387.3 ng/mL] vs 97.6 ng/mL [IQR, 74.4-126.6 ng/mL]; P < .0001) and decreased elafin levels (32.0 ng/mL [IQR, 15.3-59.1 ng/mL] vs 45.5 ng/mL [IQR, 28.1-92.8 ng/mL]; P < .0001) independent of PAH subtype, illness duration, and therapies. Higher NE levels were associated with worse symptom severity, shorter 6-min walk distance, higher N-terminal pro-type brain natriuretic peptide levels, greater right ventricular dysfunction, worse hemodynamics, increased circulating neutrophil levels, elevated cytokine levels, and lower blood BMPR2 expression. In Stanford patients, NE levels of > 168.5 ng/mL portended increased mortality risk after adjustment for known clinical predictors (hazard ratio [HR], 2.52; CI, 1.36-4.65, P = .003) or prognostic cytokines (HR, 2.63; CI, 1.42-4.87; P = .001), and the NE level added incremental value to established PAH risk scores. Similar prognostic thresholds were identified in validation cohorts. Longitudinal NE changes tracked with clinical trends and outcomes. PAH PAECs exhibited increased apoptosis and attenuated angiogenesis when exposed to NE at the level observed in patients' blood. Elafin rescued PAEC homeostasis, yet the required dose exceeded levels found in patients. INTERPRETATION Blood levels of NE are increased while elafin levels are deficient across PAH subtypes. Higher NE levels are associated with worse clinical disease severity and outcomes, and this target-specific biomarker could facilitate therapeutic development of elafin.
Collapse
Affiliation(s)
- Andrew J Sweatt
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA.
| | - Kazuya Miyagawa
- Department of Pediatrics-Cardiology, Stanford University, Stanford, CA; Betty Irene Moore Children's Heart Center, Stanford University, Stanford, CA
| | - Christopher J Rhodes
- National Heart and Lung Institute, Hammersmith Hospital, Imperial College, London
| | - Shalina Taylor
- Department of Pediatrics-Cardiology, Stanford University, Stanford, CA; Betty Irene Moore Children's Heart Center, Stanford University, Stanford, CA
| | - Patricia A Del Rosario
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA
| | - Andrew Hsi
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA
| | - Francois Haddad
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA
| | - Edda Spiekerkoetter
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA
| | - Michal Bental-Roof
- Department of Pediatrics-Cardiology, Stanford University, Stanford, CA; Betty Irene Moore Children's Heart Center, Stanford University, Stanford, CA
| | - Richard D Bland
- Department of Pediatrics-Neonatology, Stanford University, Stanford, CA
| | | | - Stefan Gräf
- Department of Medicine, University of Cambridge, Cambridge, England; NIHR BioResource for Translational Research, University of Cambridge, Cambridge, England; Department of Haematology, University of Cambridge, Cambridge, England; on behalf of the British Heart Foundation/Medical Research Council UK PAH Consortium (C. J. Rhodes, E. M. Swietlik, S. Gräf, M. R. Wilkins, and N. W. Morrell)
| | - Martin R Wilkins
- National Heart and Lung Institute, Hammersmith Hospital, Imperial College, London
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, England; NIHR BioResource for Translational Research, University of Cambridge, Cambridge, England
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA
| | - Marlene Rabinovitch
- Department of Pediatrics-Cardiology, Stanford University, Stanford, CA; Betty Irene Moore Children's Heart Center, Stanford University, Stanford, CA
| | - Roham T Zamanian
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University, Stanford, CA
| |
Collapse
|
42
|
Evans CE, Cober ND, Dai Z, Stewart DJ, Zhao YY. Endothelial cells in the pathogenesis of pulmonary arterial hypertension. Eur Respir J 2021; 58:13993003.03957-2020. [PMID: 33509961 DOI: 10.1183/13993003.03957-2020] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease that involves pulmonary vasoconstriction, small vessel obliteration, large vessel thickening and obstruction, and development of plexiform lesions. PAH vasculopathy leads to progressive increases in pulmonary vascular resistance, right heart failure and, ultimately, premature death. Besides other cell types that are known to be involved in PAH pathogenesis (e.g. smooth muscle cells, fibroblasts and leukocytes), recent studies have demonstrated that endothelial cells (ECs) have a crucial role in the initiation and progression of PAH. The EC-specific role in PAH is multi-faceted and affects numerous pathophysiological processes, including vasoconstriction, inflammation, coagulation, metabolism and oxidative/nitrative stress, as well as cell viability, growth and differentiation. In this review, we describe how EC dysfunction and cell signalling regulate the pathogenesis of PAH. We also highlight areas of research that warrant attention in future studies, and discuss potential molecular signalling pathways in ECs that could be targeted therapeutically in the prevention and treatment of PAH.
Collapse
Affiliation(s)
- Colin E Evans
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicholas D Cober
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Dept of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Zhiyu Dai
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Dept of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Section of Injury Repair and Regeneration, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA .,Dept of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Dept of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
43
|
Lechartier B, Berrebeh N, Huertas A, Humbert M, Guignabert C, Tu L. Phenotypic Diversity of Vascular Smooth Muscle Cells in Pulmonary Arterial Hypertension: Implications for Therapy. Chest 2021; 161:219-231. [PMID: 34391758 DOI: 10.1016/j.chest.2021.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive incurable condition that is characterized by extensive remodelling of the pulmonary circulation, leading to severe right heart failure and death. Similar to other vascular contractile cells, pulmonary arterial smooth muscle cells (PA-SMCs) play central roles in physiological and pathological vascular remodelling due to their remarkable ability to dynamically modulate their phenotype to ensure contractile and synthetic functions. The dysfunction and molecular mechanisms underlying their contribution to the various pulmonary vascular lesions associated with PAH have been a major focus of research. The aim of this review is to describe the medial and non-medial origins of contractile cells in the pulmonary vascular wall and present evidence of how they contribute to the onset and progression of PAH. We also highlight specific potential target molecules and discuss future directions that are being explored to widen the therapeutic options for the treatment of PAH.
Collapse
Affiliation(s)
- Benoit Lechartier
- Pulmonary Division, Lausanne University Hospital, Lausanne, Switzerland; Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Nihel Berrebeh
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Alice Huertas
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Ly Tu
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
44
|
Ramirez RL, Pienkos SM, de Jesus Perez V, Zamanian RT. Pulmonary Arterial Hypertension Secondary to Drugs and Toxins. Clin Chest Med 2021; 42:19-38. [PMID: 33541612 DOI: 10.1016/j.ccm.2020.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Pulmonary arterial hypertension secondary to drugs and toxins is an important subgroup of group 1 pulmonary hypertension associated with significant morbidity and mortality. Many drugs and toxins have emerged as risk factors for pulmonary arterial hypertension, which include anorexigens, illicit agents, and several US Food and Drug Administration-approved therapeutic medications. Drugs and toxins are classified as possible or definite risk factors for pulmonary arterial hypertension. This article reviews agents that have been implicated in the development of pulmonary arterial hypertension, their pathologic mechanisms, and methods to prevent the next deadly outbreak of drug- and toxin-induced pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Ramon L Ramirez
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA
| | - Shaun M Pienkos
- Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA
| | - Roham T Zamanian
- Division of Pulmonary, Allergy and Critical Care, Stanford University School of Medicine, 300 Pasteur Drive, Room S102, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford, CA, USA.
| |
Collapse
|
45
|
Gelzinis TA. Pulmonary Hypertension in 2021: Part I-Definition, Classification, Pathophysiology, and Presentation. J Cardiothorac Vasc Anesth 2021; 36:1552-1564. [PMID: 34344595 DOI: 10.1053/j.jvca.2021.06.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 11/11/2022]
Abstract
The World Symposium on Pulmonary Hypertension (WSPH) was organized by the World Health Organization in 1973 in response to an increase in pulmonary arterial hypertension in Europe caused by aminorex, an appetite suppressant. The mandate of this meeting was to review the latest clinical and scientific research and to formulate recommendations to improve the diagnosis and management of pulmonary hypertension (PH).1 Since 1998, the WSPH has met every five years and in 2018, the sixth annual WSPH revised the hemodynamic definition of PH. This two-part series will review the updated definition, classification, pathophysiology, presentation, diagnosis, management, and perioperative management of patients with PH. In the first part of this series, the definition, classification, pathophysiology, and presentation will be reviewed.
Collapse
|
46
|
Restoration of Vitamin D Levels Improves Endothelial Function and Increases TASK-Like K + Currents in Pulmonary Arterial Hypertension Associated with Vitamin D Deficiency. Biomolecules 2021; 11:biom11060795. [PMID: 34073580 PMCID: PMC8227733 DOI: 10.3390/biom11060795] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Vitamin D (vitD) deficiency is highly prevalent in patients with pulmonary arterial hypertension (PAH). Moreover, PAH-patients with lower levels of vitD have worse prognosis. We hypothesize that recovering optimal levels of vitD in an animal model of PAH previously depleted of vitD improves the hemodynamics, the endothelial dysfunction and the ionic remodeling. Methods: Male Wistar rats were fed a vitD-free diet for five weeks and then received a single dose of Su5416 (20 mg/Kg) and were exposed to vitD-free diet and chronic hypoxia (10% O2) for three weeks to induce PAH. Following this, vitD deficient rats with PAH were housed in room air and randomly divided into two groups: (a) continued on vitD-free diet or (b) received an oral dose of 100,000 IU/Kg of vitD plus standard diet for three weeks. Hemodynamics, pulmonary vascular remodeling, pulmonary arterial contractility, and K+ currents were analyzed. Results: Recovering optimal levels of vitD improved endothelial function, measured by an increase in the endothelium-dependent vasodilator response to acetylcholine. It also increased the activity of TASK-1 potassium channels. However, vitD supplementation did not reduce pulmonary pressure and did not ameliorate pulmonary vascular remodeling and right ventricle hypertrophy. Conclusions: Altogether, these data suggest that in animals with PAH and severe deficit of vitD, restoring vitD levels to an optimal range partially improves some pathophysiological features of PAH.
Collapse
|
47
|
Guihaire J, Deuse T, Wang D, Spin JM, Blankenberg FG, Fadel E, Reichenspurner H, Schrepfer S. Immunomodulation Therapy Using Tolerogenic Macrophages in a Rodent Model of Pulmonary Hypertension. Stem Cells Dev 2021; 30:515-525. [PMID: 33726521 DOI: 10.1089/scd.2021.0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation plays a major role in the pathogenesis of pulmonary hypertension (PH). We sought to investigate the effects of a cell-based immunomodulation in a dysimmune model of PH. PH was induced in athymic nude rats using semaxinib (Su group, n = 6). Tolerogenic macrophages (toM) were generated from monocyte isolation and then injected either the day before semaxinib injection (Prevention group, n = 6) or 3 weeks after (Reversion group, n = 6). Six athymic nude rats were used as controls. In vivo trafficking of toM was investigated with bioluminescence imaging showing that toM were mainly located into the lungs until 48 h after injection. Right ventricular (RV) end-systolic pressure and RV systolic function were assessed at 4 weeks using echocardiography. Morphometric analysis and RNA sequencing of the lungs were realized at 4 weeks. Rats treated with toM (Prevention and Reversion groups) had a significantly lower RV end-systolic pressure at 4 weeks (respectively, 25 ± 8 and 30 ± 6 mmHg vs. 67 ± 9 mmHg, P < 0.001), while RV systolic dysfunction was observed in Su and Reversion groups. Mean medial wall thickness of small arterioles was lower in Prevention and Reversion groups compared with the Su group (respectively, 10.9% ± 0.8% and 16.4% ± 1.3% vs. 28.2% ± 2.1%, P < 0.001). Similarly, cardiomyocyte area was decreased in rats treated with toM (150 ± 18 and 160 ± 86 μm2 vs. 279 ± 50 μm2, P < 0.001). A trend toward upregulation of genes involved in pulmonary arterial hypertension pathobiology was found in Su rats, while KCNK3 was significantly downregulated (fold-change = 9.8, P < 0.001). Injection of toM was associated with a less severe phenotype of PH in rats exposed to angioproliferative stress. Preserved expression of KCNK3 may explain the protective effect of toM.
Collapse
Affiliation(s)
- Julien Guihaire
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Department of Cardiac Surgery, Inserm UMR_S 999, Pulmonary Hypertension: Pathophysiology and Novel Therapies, Marie Lannelongue Hospital, Groupe Hospitalier Paris Saint Joseph, University of Paris-Saclay School of Medicine, Le Plessis Robinson, France
| | - Tobias Deuse
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Dong Wang
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| | - Joshua M Spin
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Francis G Blankenberg
- Division of Pediatric Radiology, Department of Radiology/MIPS, Lucile Salter Packard Children's Hospital, Stanford University, Palo Alto, California, USA
| | - Elie Fadel
- Thoracic and Vascular Surgery, Heart and Lung Transplantation, Marie Lannelongue Hospital, Groupe Hospitalier Paris Saint Joseph, University of Paris-Saclay School of Medicine, Le Plessis Robinson, France
| | - Hermann Reichenspurner
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- Department of Cardiovascular Surgery, University Heart Center of Hamburg, Hamburg, Germany
| | - Sonja Schrepfer
- Transplant and Stem Cells Immunobiology (TSI) Lab, University Heart Center of Hamburg, Hamburg, Germany
- Transplant and Stem Cells Immunobiology (TSI) Lab, Department of Surgery, University of California San Francisco, San Francisco, California, USA
- Cardiovascular Research Center Hamburg (CVRC) and DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
| |
Collapse
|
48
|
Koudstaal T, van Uden D, van Hulst JAC, Heukels P, Bergen IM, Geenen LW, Baggen VJM, van den Bosch AE, van den Toorn LM, Chandoesing PP, Kool M, Boersma E, Hendriks RW, Boomars KA. Plasma markers in pulmonary hypertension subgroups correlate with patient survival. Respir Res 2021; 22:137. [PMID: 33947407 PMCID: PMC8097895 DOI: 10.1186/s12931-021-01716-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/13/2021] [Indexed: 11/21/2022] Open
Abstract
Background Recent studies have provided evidence for an important contribution of the immune system in the pathophysiology of pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH). In this report, we investigated whether the inflammatory profile of pulmonary hypertension patients changes over time and correlates with patient WHO subgroups or survival. Methods 50 PAH patients (16 idiopathic (I)PAH, 24 Connective Tissue Disease (CTD)-PAH and 10 Congenital Heart Disease (CHD)-PAH), 37 CTEPH patients and 18 healthy controls (HCs) were included in the study. Plasma inflammatory markers at baseline and after 1-year follow-up were measured using ELISAs. Subsequently, correlations with hemodynamic parameters and survival were explored and data sets were subjected to unbiased multivariate analyses. Results At diagnosis, we found that plasma levels of interleukin-6 (IL-6) and the chemokines (C-X3-C) motif legend CXCL9 and CXCL13 in CTD-PAH patients were significantly increased, compared with HCs. In idiopathic PAH patients the levels of tumor growth factor-β (TGFβ), IL-10 and CXCL9 were elevated, compared with HCs. The increased CXCL9 and IL-8 concentrations in CETPH patients correlated significantly with decreased survival, suggesting that CXCL9 and IL-8 may be prognostic markers. After one year of treatment, IL-10, CXCL13 and TGFβ levels changed significantly in the PAH subgroups and CTEPH patients. Unbiased multivariate analysis revealed clustering of PH patients based on inflammatory mediators and clinical parameters, but did not separate the WHO subgroups. Importantly, these multivariate analyses separated patients with < 3 years and > 3 years survival, in particular when inflammatory mediators were combined with clinical parameters. Discussion Our study revealed elevated plasma levels of inflammatory mediators in different PAH subgroups and CTEPH at baseline and at 1-year follow-up, whereby CXCL9 and IL-8 may prove to be prognostic markers for CTEPH patients. While this study is exploratory and hypothesis generating, our data indicate an important role for IL-8 and CXCL9 in CHD and CTEPH patients considering the increased plasma levels and the observed correlation with survival. Conclusion In conclusion, our studies identified an inflammatory signature that clustered PH patients into WHO classification-independent subgroups that correlated with patient survival. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01716-w.
Collapse
Affiliation(s)
- T Koudstaal
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - D van Uden
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - J A C van Hulst
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - P Heukels
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - I M Bergen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - L W Geenen
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - V J M Baggen
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - A E van den Bosch
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - L M van den Toorn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - P P Chandoesing
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - M Kool
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - E Boersma
- Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - R W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - K A Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Bai Z, Xu L, Dai Y, Yuan Q, Zhou Z. ECM2 and GLT8D2 in human pulmonary artery hypertension: fruits from weighted gene co-expression network analysis. J Thorac Dis 2021; 13:2242-2254. [PMID: 34012575 PMCID: PMC8107565 DOI: 10.21037/jtd-20-3069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Pulmonary artery hypertension (PAH) is an incurable disease with a high mortality rate. Current medications ameliorate symptoms but cannot target adverse vascular remodeling. New therapeutic strategies for PAH need to be established. Methods Using the weighted gene coexpression network analysis (WGCNA) algorithm, we constructed a coexpression network of dataset GSE117261 from the Gene Expression Omnibus (GEO) database. Key modules were identified by the relationship between module eigengenes and clinical traits. Hub genes were screened out based on gene significance (GS), module membership (MM), and mean pulmonary artery pressure (mPAP). External validations were conducted in GSE48149 and GSE113439. Functional enrichment and immune cell infiltration were analyzed using Metascape and CIBERSORTx. Results The WGCNA analysis revealed 13 coexpression modules. The pink module had the highest correlation with PAH in terms of module eigengene (r=0.79; P=2e-18) and module significance (MS =0.43). Functional enrichment indicated genes in the pink module contributed to the immune system process and extracellular matrix (ECM). In the pink module, ECM2 (GS =0.65, MM =0.86, ρ=0.407, P=0.0019) and GLT8D2 (GS =0.63, MM =0.85, ρ=0.443, P=0.006) were identified as hub genes. For immune cells infiltration in PAH lung tissue, hub genes were positively correlated with M2 macrophages and resting mast cells, and were negatively correlated with monocytes, neutrophils, and CD4-naïve T cells. Conclusions Our research identified 2 hub genes ECM2 and GLT8D2 related to PAH. The functions of these hub genes were involved in the immune process and ECM, indicating that they might serve as candidate therapeutic targets for PAH.
Collapse
Affiliation(s)
- Zeyang Bai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianyan Xu
- Department of Radiology, Peking Union Medical College Hospital, PUMC & CAMS, Beijing, China
| | - Yong Dai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingchen Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Mercurio V, Cuomo A, Naranjo M, Hassoun PM. Inflammatory Mechanisms in the Pathogenesis of Pulmonary Arterial Hypertension: Recent Advances. Compr Physiol 2021; 11:1805-1829. [PMID: 33792903 DOI: 10.1002/cphy.c200025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammatory processes are increasingly recognized in the pathogenesis of the vascular remodeling that characterizes pulmonary arterial hypertension (PAH). Chronic inflammation may contribute to disease progression or serve as a biomarker of PAH severity. Furthermore, inflammatory pathways may represent possible therapeutic targets for novel PAH-specific drugs beyond the currently approved therapies targeting the endothelin, nitric oxide/cyclic GMP, and prostacyclin biological pathways. The main focus of this article is to provide recent advances in the understanding of the role of inflammatory pathways in the pathogenesis of PAH from preclinical studies and current clinical data supporting chronic inflammation in PAH patients and to discuss emerging therapeutic implications. © 2021 American Physiological Society. Compr Physiol 11:1805-1829, 2021.
Collapse
Affiliation(s)
- Valentina Mercurio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.,Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Mario Naranjo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|