1
|
Zhang J, Qin X, Qin Y, Qi F, Wang Y, Sun J, Yan L, Sun W, Guo X. Proteomic- and metabolomic-based mechanisms of androgen-mediated right ventricular maladaptive remodeling under pressure overload. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167768. [PMID: 40049298 DOI: 10.1016/j.bbadis.2025.167768] [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: 11/15/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Right ventricular (RV) maladaptive remodeling has been demonstrated to be more severe in males than in females under similar afterload, with androgen potentially involved. However, the mechanism remains unknown. METHODS We performed RV proteomics and metabolomics in male and castrated rats with pulmonary artery banding (PAB) or sham surgery. The core pathway was tested in other sets of male, castrated male, and testosterone-replaced rats with and without pathway inhibitors administration and in RV remodeling patients. Metabolite verification was carried out by matching secondary spectra. RESULTS With the same extent of increases in RV afterload, male PAB rats exhibited more pronounced RV hypertrophy and fibrosis than castrated PAB rats (p < 0.05). The omics analysis indicated that pathways and functions related to oxidative stress were exhibited in the male group, with the platelet-derived growth factor (PDGF) pathway being among them. More proteins and metabolites associated with fatty acid metabolism were downregulated in males. Correlation analysis showed that PDGF receptor beta (PDGFRB) and signal transducer and activator of transcription 3 (STAT3) were negatively correlated with carnitine and reactive oxygen species scavenging metabolites only in male rats. The activation of the PDGF pathway was verified in testosterone-replaced PAB rats and male patients with RV remodeling. Treatments with PDGFRB inhibitor and STAT3 inhibitor could reverse RV maladaptive remodeling in male and testosterone-replaced PAB rats but not in castrated ones. CONCLUSIONS Androgen might exacerbate RV maladaptive remodeling via intensified oxidative stress and insufficient energy supply, with activating the PDGFRB-STAT3 signaling being one of the possible pathways.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhan Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Qi
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yufei Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiameng Sun
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China; Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Wei Sun
- Proteomics Center, Core Facility of Instrument, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China.
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Liu B, Yi D, Li S, Ramirez K, Xia X, Cao Y, Zhao H, Tripathi A, Qiu S, Kala M, Rafikov R, Gu H, de Jesus Perez V, Lemay SE, Glembotski CC, Knox KS, Bonnet S, Kalinichenko VV, Zhao YY, Fallon MB, Boucherat O, Dai Z. Single-Cell and Spatial Transcriptomics Identified Fatty Acid-Binding Proteins Controlling Endothelial Glycolytic and Arterial Programming in Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2025. [PMID: 40401371 DOI: 10.1161/atvbaha.124.321173] [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: 05/01/2024] [Accepted: 05/09/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a devastating disease characterized by obliterative vascular remodeling and persistent increase of vascular resistance, leading to right heart failure and premature death. Understanding the cellular and molecular mechanisms will help develop novel therapeutic approaches for patients with PAH. Recent studies showed that FABP (fatty acid-binding protein) 4 and FABP5 are expressed in endothelial cells (ECs) across multiple tissues, and circulating FABP4 level is elevated in patients with PAH. However, the role of endothelial FABP4/5 in the pathogenesis of PAH remains undetermined. METHODS FABP4/5 expression was examined in pulmonary arterial ECs and lung tissues from patients with idiopathic PAH and pulmonary hypertension (PH) rat models. Plasma proteome analysis was performed in human PAH samples. Echocardiography, hemodynamics, histology, and immunostaining were performed to evaluate the lung and heart PH phenotypes in Egln1Tie2Cre (CKO) mice and Egln1Tie2Cre/Fabp4/5-/- (TKO) mice. Bulk RNA sequencing (RNA-seq), single-cell RNA sequencing analysis, and spatial transcriptomic analysis were performed to understand the cellular and molecular mechanisms of endothelial FABP4/5-mediated PAH pathogenesis. RESULTS Both FABP4 and FABP5 were highly induced in ECs of CKO mice and pulmonary arterial ECs from patients with idiopathic PAH (IPAH) and in whole lungs of PH rats. Plasma levels of FABP4/5 were upregulated in patients with IPAH and directly correlated with severity of hemodynamics and biochemical parameters. Genetic deletion of both Fabp4 and Fabp5 in CKO mice caused a reduction of right ventricular systolic pressure and right ventricular hypertrophy, attenuated pulmonary vascular remodeling, and prevented the right heart failure secondary to PH. FABP4/5 deletion also normalized EC glycolysis and distal arterial programming, reduced reactive oxygen species and HIF (hypoxia-inducible factor)-2α expression, and decreased aberrant EC proliferation in CKO lungs. CONCLUSIONS PH causes aberrant expression of FABP4/5 in pulmonary ECs, which leads to enhanced EC glycolysis and distal arterial programming, contributing to the accumulation of arterial ECs and vascular remodeling and exacerbating the disease.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., C.C.G., Z.D.)
- Division of Pulmonary Critical Care and Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, MO (B.L., A.T., Z.D.)
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., C.C.G., Z.D.)
| | - Shuai Li
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Now with GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, China (S.L.)
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Xiaomei Xia
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Yanhong Cao
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Hanqiu Zhao
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Ankit Tripathi
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Division of Pulmonary Critical Care and Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, MO (B.L., A.T., Z.D.)
| | - Shenfeng Qiu
- Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix. (S.Q.)
- Division of Pulmonary Critical Care and Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, MO (B.L., A.T., Z.D.)
| | - Mrinalini Kala
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Ruslan Rafikov
- Department of Medicine, Indiana University College of Medicine, Indianapolis (R.R.)
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix (H.G.)
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA (V.d.j.P., O.B.)
| | - Sarah-Eve Lemay
- Pulmonary Hypertension and Vascular Biology Research Group, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada (S.-E.L., S.B.)
| | - Christopher C Glembotski
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., C.C.G., Z.D.)
| | - Kenneth S Knox
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Sebastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada (S.-E.L., S.B.)
| | - Vladimir V Kalinichenko
- Phoenix Children's Health Research Institute, College of Medicine-Phoenix, University of Arizona, Phoenix. (V.V.K.)
- Division of Neonatology, Phoenix Children's Hospital, Phoenix, AZ (V.V.K.)
| | - You-Yang Zhao
- Program for Lung and Vascular Biology and Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, IL (Y.-Y.Z.)
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL (Y.-Y.Z.)
| | - Michael B Fallon
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
| | - Olivier Boucherat
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA (V.d.j.P., O.B.)
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., K.S.K., Z.D.)
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., S.L., K.R., X.X., Y.C., H.Z., A.T., M.K., C.C.G., K.S.K., M.B.F., Z.D.)
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix. (B.L., D.Y., C.C.G., Z.D.)
| |
Collapse
|
3
|
Rosen DT, Kolb TM, Mathai SC, Suresh K, Damico R, Hsu S, Tedford RJ, Hemnes AR, Leopold JA, Horn EM, Berman-Rosenzweig ES, Rischard F, Frantz RP, Erzurum SC, Beck GJ, Hill NS, Barnard J, Farha S, Grunig G, Jellis C, Kwon DH, Mehra R, Park MM, Tang WHW, Hassoun PM, Simpson CE. Dysregulated Tricarboxylic Acid Cycle Metabolism Is Associated With Right Ventricular Maladaptation in Pulmonary Vascular Disease. J Am Heart Assoc 2025:e041127. [PMID: 40401597 DOI: 10.1161/jaha.124.041127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Right ventricular (RV) maladaptation to elevated pulmonary afterload is the primary determinant of outcomes in pulmonary artery (PA) hypertension; however, the pathobiological mechanisms underlying RV decompensation remain poorly understood. METHODS We performed global untargeted metabolomics on plasma from 55 patients who underwent gold-standard RV-PA coupling measurements using multibeat pressure volume loop assessment in a single-center cohort and from 1027 patients with coupling surrogate measurements in a larger multicenter cohort, the PVDOMICS (Pulmonary Vascular Disease Phenomics) study. Age and sex-adjusted linear regression was performed to identify associations between metabolites and coupling metrics. Additionally, we performed a metabolic flux analysis using gene expression data from RV tissue in an independent cohort of 32 patients. Partial least squares-discriminant analysis was used to identify metabolites and reactions characteristic of the decompensated RV. RESULTS RV-PA coupling was negatively associated with tricarboxylic acid (TCA) cycle intermediate levels. Specifically, plasma α-ketoglutarate and fumarate were significantly associated with all coupling metrics in both cohorts. Metabolic flux analysis indicated that decompensated RVs exhibited aberrant TCA cycle activity, including reduced acetyl coenzyme A entry and increased lactate elimination, suggesting a shift from the TCA cycle toward glycolysis at the RV tissue level. CONCLUSIONS We identify an association between circulating TCA cycle intermediate levels and RV-PA uncoupling in 2 independent cohorts, and dysregulated TCA cycle metabolism in decompensated PA hypertension RVs, suggesting that aberrant TCA cycle metabolism could represent a hallmark of RV maladaptation in PA hypertension. Further study of this pathway is warranted to develop novel biomarkers of RV function or RV-targeted therapies.
Collapse
Affiliation(s)
- Darin T Rosen
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| | - Todd M Kolb
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| | - Stephen C Mathai
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| | - Rachel Damico
- Division of Pulmonary, Critical Care and Sleep Medicine University of Miami FL USA
| | - Steven Hsu
- Division of Cardiology Johns Hopkins University Baltimore MD USA
| | - Ryan J Tedford
- Division of Cardiology, Department of Medicine Medical University of South Carolina Charleston SC USA
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine Vanderbilt University Nashville TN USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine Brigham and Women's Hospital Boston MA USA
| | - Evelyn M Horn
- Division of Cardiology, Department of Medicine Cornell University Medical Center New York NY USA
| | | | - Franz Rischard
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine University of Arizona College of Medicine Tucson AZ USA
| | - Robert P Frantz
- Division of Circulatory Failure, Department of Cardiovascular Medicine Mayo Clinic Rochester MN USA
| | | | - Gerald J Beck
- Cleveland Clinic Lerner Research Institute Cleveland OH USA
| | - Nicholas S Hill
- Pulmonary, Critical Care and Sleep Division Tufts University Boston MA USA
| | - John Barnard
- Cleveland Clinic Lerner Research Institute Cleveland OH USA
| | - Samar Farha
- Cleveland Clinic Lerner Research Institute Cleveland OH USA
| | - Gabriele Grunig
- New York University Grossman School of Medicine New York NY USA
| | - Christine Jellis
- Heart, Vascular and Thoracic Institute, Cleveland Clinic Cleveland OH USA
| | - Deborah H Kwon
- Heart, Vascular and Thoracic Institute, Cleveland Clinic Cleveland OH USA
| | - Reena Mehra
- Division of Pulmonary, Critical Care and Sleep Medicine University of Washington Medical Center Seattle WA USA
| | - Margaret M Park
- Heart, Vascular and Thoracic Institute, Cleveland Clinic Cleveland OH USA
| | - W H Wilson Tang
- Cleveland Clinic Lerner Research Institute Cleveland OH USA
- Heart, Vascular and Thoracic Institute, Cleveland Clinic Cleveland OH USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| | - Catherine E Simpson
- Division of Pulmonary and Critical Care Medicine Johns Hopkins University Baltimore MD USA
| |
Collapse
|
4
|
Li S, Li C, Sun W, Cao Y, Qi X, Zhang J, Xing Y, Zhou J, Wang L. Spatially Resolved Metabolomics Reveals Metabolic Heterogeneity Among Pulmonary Fibrosis. JOURNAL OF MASS SPECTROMETRY : JMS 2025; 60:e5138. [PMID: 40264277 DOI: 10.1002/jms.5138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/18/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive lung disease with fatal consequences. The study of PF is challenging due to the complex mechanism involved, the need to understand the heterogeneity and spatial organization within lung tissues. In this study, we investigate the metabolic heterogeneity between two forms of lung fibrosis: idiopathic pulmonary fibrosis (IPF) and silicosis, using advanced spatially-resolved metabolomics techniques. Employing high-resolution mass spectrometry imaging, we spatially mapped and identified over 260 metabolites in lung tissue sections from mouse models of IPF and silicosis. Histological analysis confirmed fibrosis in both models, with distinct pathological features: alveolar destruction and collagen deposition in IPF, and nodule formation in silicosis. Metabolomic analysis revealed significant differences between IPF and silicosis in key metabolic pathways, including phospholipid metabolism, purine/pyrimidine metabolism, and the TCA cycle. Notably, phosphocholine was elevated in silicosis but reduced in IPF, while carnitine levels decreased in both conditions. Additionally, glycolytic activity was increased in both models, but TCA cycle intermediates showed opposing trends. These findings highlight the spatial metabolic heterogeneity of PF and suggest potential metabolic targets for therapeutic intervention. Further investigation into the regulatory mechanisms behind these metabolic shifts may open new avenues for fibrosis treatment.
Collapse
Affiliation(s)
- Shengxi Li
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Li
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Wei Sun
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun, China
| | - Yinghao Cao
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianmei Qi
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiawei Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinyu Zhou
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Wang
- State Key Laboratory of Common Mechanism Research for Major Disease, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Zhang L, Zhang Y, Ding Y, Jin T, Song Y, Li L, Wang X, Zeng Y. Cer(d18:1/16:0) as a biomarkers for acute coronary syndrome in Chinese populations. Sci Rep 2025; 15:14014. [PMID: 40269114 PMCID: PMC12019627 DOI: 10.1038/s41598-025-98010-2] [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: 12/09/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
Ceramides play a crucial role in atherosclerosis progression and have been linked to cardiovascular events. The objective of this study was to investigate the association between serum ceramide levels and Acute coronary syndrome, as well as evaluate their potential for predicting ACS in Chinese population. Data of 1327 patients with suspected or known coronary artery disease from Beijing anzhen Hospital and Handan First hospital were collected. Plasma ceramide were measured using the LC-MS/MS system. The area under the ROC curve was used to screen the most valuable predictor. Distinctive ACS-related variables were screened out using Boruta and LASSO regression. Multivariate Logistic models and restricted cubic spline analysis were conducted to examine the associations between Ceramide and ACS. Cer (d18:1/14:0), Cer (d18:1/16:0), Cer (d18:1/18:0), Cer (d18:1/20:0), Cer (d18:1/22:0), and Cer (d18:1/24:0) were significantly elevated in the ACS group. Diagnostic performance assessments showed that Cer(d18:1/16:0) had superior accuracy in detecting ACS compared to other ceramides tested. The Boruta algorithm identified 8 significant variables related to ACS. Cer(d18:1/16:0) associated with ACS were discovered using the LASSO logistic regression technique. Multivariate logistic regression models further supported the relationship between Cer(d18:1/16:0) and ACS. Additionally, a significant nonlinear relationship was observed between Cer(d18:1/16:0) and ACS, with a threshold of 150umol/L. The study found that ceramides, particularly Cer(d18:1/16:0), were significantly associated with ACS and could be a potential biomarker for predicting and diagnosing ACS in Chinese populations experiencing chest pain.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Yang Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing, 100029, People's Republic of China
| | - YaoDong Ding
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing, 100029, People's Republic of China
| | - Tong Jin
- Department of Vascular SurgeryBeijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China
| | - Yi Song
- Beijing Dong Zhi Men Hospital, Beijing, China
| | - Lin Li
- Beijing Health Biotechnology Co., Ltd., Beijing, China
| | - XiaoFang Wang
- Department of Pediatric Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing, China.
| | - Yong Zeng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Disease, 2 Anzhen Road, Beijing, 100029, People's Republic of China.
| |
Collapse
|
6
|
Garry JD, Davogustto GE, Agrawal V, Ye F, Tomasek K, Su YR, Absi T, West JD, Hemnes A, Brittain EL. Transcriptional Signatures of the Right Ventricle in End-Stage Heart Failure. Pulm Circ 2025; 15:e70090. [PMID: 40276472 PMCID: PMC12020025 DOI: 10.1002/pul2.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
The molecular mechanisms driving right ventricular (RV) adaptation to stress and failure in end-stage heart failure (HF) are largely unknown. We aimed to characterize myocardial transcriptional changes in the RV caused by left sided HF and comparing RV compensation to failure. Additionally, we compared transcriptomic changes between right and left ventricular (LV) failure. Paired right and left ventricular myocardial tissue samples were obtained from 33 human subjects with end stage HF referred for transplantation and 8 control donors with unused transplant hearts. RV samples from end stage HF subjects were subdivided into compensated (n = 25) and failing (n = 8) categories based on pulmonary artery pulsatility index of < 1.85. All samples underwent bulk tissue RNA-sequencing. We compared gene expression between groups and performed pathway enrichment analysis. Pathways related to fatty acid metabolism and mitochondrial function were negatively enriched, while extracellular structure-related pathways were positively enriched in stressed RVs (compensated and failing) compared to controls. Compensated and failing RVs were differentiated by transcriptional changes in protein production/processing and immune system pathways. PPAR signaling and fatty acid metabolism pathways were consistently enriched in the RV compared to the LV. The RV has a distinct transcriptional signature under stress and in failure. Overlapping molecular mechanisms may underlie RV failure in pulmonary arterial hypertension and HF. Fatty Acid metabolism and associated signaling pathways appear enriched in the RV compared to the LV.
Collapse
Affiliation(s)
- Jonah D. Garry
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Giovanni E. Davogustto
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vineet Agrawal
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Veteran AffairsTennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Fei Ye
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kelsey Tomasek
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yan Ru Su
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Tarek Absi
- Department of Cardiac SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - James D. West
- Division of Allergy, Pulmonary, and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Anna Hemnes
- Division of Allergy, Pulmonary, and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Evan L. Brittain
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
7
|
Su L, Wang X, Lin Y, Zhang Y, Yao D, Pan T, Huang X. Exploring the Causal Relationship Between Gut Microbiota and Pulmonary Artery Hypertension: Insights From Mendelian Randomization. J Am Heart Assoc 2025; 14:e038150. [PMID: 40079338 DOI: 10.1161/jaha.124.038150] [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: 08/06/2024] [Accepted: 02/14/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Research into the "gut-lung" axis links gut microbiota to pulmonary artery hypertension (PAH). However, the mechanisms by which gut microbiota influence PAH remain unclear. We aimed to investigate the causal relationship between the gut microbiota and PAH using Mendelian randomization analysis, identify key microbiota and metabolites, and explore the regulatory role of associated genes in PAH pathogenesis. METHODS AND RESULTS We examined the association between gut microbiota taxa and PAH using inverse variance weighted 2-sample Mendelian randomization analysis, Mendelian randomization-Egger, weighted median, and weighted mode methods. Additionally, we identified PAH-regulating genes in the intestinal microbiome using bioinformatics tools and validated their expression levels in the lung tissue of hypoxia-induced PAH mice models by quantitative reverse transcription polymerase chain reaction. Eleven gut microbiota taxa were associated with PAH. The order Clostridiales and genera Eubacterium fissicatena group, Lachnospiraceae UCG004, and Ruminococcaceae UCG002 were positively associated with PAH, whereas the order Bifidobacteriales; family Bifidobacteriaceae; and genera Eubacterium eligens group, Sutterella, Methanobrevibacter, Sellimonas, and Tyzzerella3 were negatively associated with PAH, with some exhibiting bidirectional causality. These microbiota modulate 24 metabolites, including palmitoylcholine, oleoylcholine, and 3,7-dimethylurate, to influence PAH. Hypoxia-induced PAH mice had significantly downregulated 1,4,5-trisphosphate receptor type 2, degrading enzyme, nuclear receptor-interacting protein 1, and growth factor-binding protein 1 in lung tissues, indicating their potential role in PAH regulation. CONCLUSIONS These findings suggest that gut microbiota composition and associated metabolites contribute to PAH development by regulating lung tissue gene expression. Our findings have implications for advancing gut microbiota-based PAH diagnostic technologies and targeted therapies.
Collapse
Affiliation(s)
- Lihuang Su
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Xinghong Wang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Ya Lin
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Yiying Zhang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Dan Yao
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Tongtong Pan
- Department of Gerontology The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, Wenzhou Key Laboratory of Interdisciplinary and Translational Medicine The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang China
| |
Collapse
|
8
|
Zuo H, Liu X, Wang Y, Ding H, Wan W, Zheng S, Hou S, Hu K. SREBF1 facilitates pathological retinal neovascularization by reprogramming the fatty acid metabolism of endothelial cells. Exp Eye Res 2025; 252:110239. [PMID: 39800283 DOI: 10.1016/j.exer.2025.110239] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Retinopathy of prematurity (ROP) is a proliferative retinal vascular disorder that critically affects the visual development of premature infants, potentially leading to irreversible vision loss or even blindness. Despite its significance, the underlying mechanisms of this disease remain insufficiently understood. In this study, we utilized the oxygen-induced retinopathy (OIR) mouse model and conducted endothelial functional assays to explore the role of Sterol Regulatory Element-Binding Protein 1 (SREBF1) in ROP pathogenesis. SREBF1 expression levels, along with its downstream targets, were investigated through Western blotting, RT-qPCR, and immunofluorescence staining techniques. Furthermore, Co-Immunoprecipitation (Co-IP) was employed to examine the molecular mechanisms involved. Our results demonstrated a significant increase in SREBF1 expression in both the OIR mouse model and hypoxic primary human retinal microvascular endothelial cells (HRMECs). Interventions conducted both in vivo and in vitro showed notable efficacy in reducing pathological neovascularization. Importantly, we discovered that SREBF1 plays a key role in modulating lipid metabolism in HRMECs by regulating the expression of ACC1 and FASN, leading to cellular reprogramming. This reprogramming influences HRMEC proliferation, migration, and tube formation through the HIF-1α/TGF-β signaling pathway, ultimately contributing to pathological retinal neovascularization. These findings provide new insights into the role of SREBF1 in angiogenesis within the context of ROP, offering potential therapeutic targets for the management and treatment of this disease.
Collapse
Affiliation(s)
- Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China; Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yakun Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Huannan Ding
- Chongqing Medical University, Chongqing, PR China
| | - Wenjuan Wan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Shijie Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Shengping Hou
- Chongqing Medical University, Chongqing, PR China; Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
9
|
Kazmirczak F, Moon R, Vogel NT, Tollison W, Lahti MT, Carney JP, Mendelson JB, Markowski T, Higgins L, Murray K, Guerrero C, Prins KW. Ferroptosis Inhibition Combats Metabolic Derangements and Improves Cardiac Function in Pulmonary Artery Banded Pigs. Am J Respir Crit Care Med 2025; 211:512-515. [PMID: 39700526 PMCID: PMC11936134 DOI: 10.1164/rccm.202406-1197rl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024] Open
Affiliation(s)
| | - Ryan Moon
- Cardiovascular Division, Lillehei Heart Institute
| | | | - Walt Tollison
- Experimental Surgical Services Laboratory, Department of Surgery
| | - Matt T. Lahti
- Experimental Surgical Services Laboratory, Department of Surgery
| | - John P. Carney
- Experimental Surgical Services Laboratory, Department of Surgery
| | | | - Todd Markowski
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - LeeAnn Higgins
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Kevin Murray
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | - Candace Guerrero
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota
| | | |
Collapse
|
10
|
Ghofrani HA, Gomberg-Maitland M, Zhao L, Grimminger F. Mechanisms and treatment of pulmonary arterial hypertension. Nat Rev Cardiol 2025; 22:105-120. [PMID: 39112561 DOI: 10.1038/s41569-024-01064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2024] [Indexed: 11/28/2024]
Abstract
Substantial progress has been made in the management of pulmonary arterial hypertension (PAH) in the past 25 years, but the disease remains life-limiting. Established therapies for PAH are mostly limited to symptomatic relief by correcting the imbalance of vasoactive factors. The tyrosine kinase inhibitor imatinib, the first predominantly non-vasodilatory drug to be tested in patients with PAH, improved exercise capacity and pulmonary haemodynamics compared with placebo but at the expense of adverse events such as subdural haematoma. Given that administration by inhalation might reduce the risk of systemic adverse effects, inhaled formulations of tyrosine kinase inhibitors are currently in clinical development. Other novel therapeutic approaches for PAH include suppression of activin receptor type IIA signalling with sotatercept, which has shown substantial efficacy in clinical trials and was approved for use in the USA in 2024, but the long-term safety of the drug remains unclear. Future advances in the management of PAH will focus on right ventricular function and involve deep phenotyping and the development of a personalized medicine approach. In this Review, we summarize the mechanisms underlying PAH, provide an overview of available PAH therapies and their limitations, describe the development of newer, predominantly non-vasodilatory drugs that are currently being tested in phase II or III clinical trials, and discuss future directions for PAH research.
Collapse
Affiliation(s)
- Hossein-Ardeschir Ghofrani
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| | - Mardi Gomberg-Maitland
- George Washington University School of Medicine and Health Sciences, Department of Medicine, Washington, DC, USA
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
11
|
Mendelson JB, Sternbach JD, Moon RA, Hartweck LM, Clark SR, Tollison W, Lahti MT, Carney JP, Markowski T, Higgins L, Kazmirczak F, Prins KW. Glycoprotein 130 Antagonism Counteracts Metabolic and Inflammatory Alterations to Enhance Right Ventricle Function in Pulmonary Artery Banded Pigs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633954. [PMID: 39896622 PMCID: PMC11785131 DOI: 10.1101/2025.01.20.633954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background Right ventricular dysfunction (RVD) is a risk factor for death in multiple cardiovascular diseases, but RV-enhancing therapies are lacking. Inhibition of glycoprotein-130 (GP130) signaling with the small molecule SC144 improves RV function in rodent RVD via anti-inflammatory and metabolic mechanisms. However, SC144's efficacy and molecular effects in a translational large animal model of RVD are unknown. Methods 4-week-old castrated male pigs underwent pulmonary artery banding (PAB). After 3 weeks, PAB pigs were randomized into 2 groups (daily injections of SC144 [2.2 mg/kg, PAB-SC144, n=5] or vehicle [PAB-Veh, n=5] for 3 weeks). Five age-matched pigs served as controls. Cardiac MRI quantified RV size/function. Right heart catheterization evaluated hemodynamics. Single-nucleus RNA sequencing delineated cell-type specific changes between experimental groups. Electron microscopy evaluated RV mitochondrial morphology. Phosphoproteomics identified dysregulated RV kinases. Lipidomics and metabolomics quantified lipid species and metabolites in RV tissue. Quantitative proteomics examined RV mitochondrial protein regulation. Results SC144 significantly improved RV ejection fraction (Control: 60±4%, PAB-Veh: 22±10%, PAB-SC144: 37±6%) despite similar RV afterload. Single-nucleus RNA sequencing demonstrated PAB-Veh pigs had lower cardiomyocyte and higher macrophage/lymphocyte/pericyte/endothelial cell abundances as compared to control, and many of these changes were blunted by SC144. SC144 combatted the downregulation of cardiomyocyte metabolic genes induced by PAB. Kinome enrichment analysis suggested SC144 counteracted RV mTORC1 activation. Correspondingly, SC144 rebalanced RV autophagy pathway proteins and improved mitochondrial morphology. Integrated lipidomics, metabolomics, and proteomics analyses revealed SC144 restored fatty acid metabolism. Finally, CellChat analysis revealed SC144 restored pericyte-endothelial cell cross-talk. Conclusion GP130 antagonism blunts elevated immune cell abundance, reduces pro-inflammatory gene transcription in macrophages and lymphocytes, rebalances autophagy and preserves fatty acid metabolism in cardiomyocytes, and restores endothelial cell and pericyte communication to improve RV function.
Collapse
Affiliation(s)
- Jenna B Mendelson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN
| | - Jacob D Sternbach
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Ryan A Moon
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Lynn M Hartweck
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Sophia R Clark
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Walt Tollison
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Matthew T Lahti
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - John P Carney
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Todd Markowski
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - LeeAnn Higgins
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - Felipe Kazmirczak
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| | - Kurt W Prins
- Lillehei Heart Institute, Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN
| |
Collapse
|
12
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
13
|
Yanagisawa A, Kim JD, Naito A, Kobayashi T, Misawa T, Sakao S, Jujo-Sanada T, Kawasaki T, Muroi SI, Sasaki SI, Suzuki T, Hayakawa Y, Nakagawa Y, Kasuya Y, Tatsumi K. Deciphering the inhibitory effects of trimetazidine on pulmonary hypertension development via decreasing fatty acid oxidation and promoting glucose oxidation. Sci Rep 2024; 14:27069. [PMID: 39511196 PMCID: PMC11544210 DOI: 10.1038/s41598-024-76100-x] [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: 05/13/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by vascular remodeling, resulting in right ventricular failure and death. Dysregulation of energy metabolism is linked to PH pathogenesis. Trimetazidine (TMZ), a selective long-chain 3-ketoacyl coenzyme A thiolase inhibitor, is critical in maintaining energy metabolism. Despite the indicated TMZ's inhibitory effect on pulmonary vascular remodeling in PH development, the integrated evaluation of the changes in biomolecules, such as metabolites and transcripts, that TMZ induces in the lung and heart tissues is largely unknown in vivo. For an improved understanding of the molecular mechanism involving the effects of TMZ on PH development, we performed a comprehensive analysis of the changes in cardiac metabolites and pulmonary transcripts of SU5416-Hypoxia (Su/Hx) rats treated with TMZ. Metabolomic analysis of the Su/Hx-induced PH hearts demonstrated that TMZ reduced the long-chain fatty acid concentration. Additionally, TMZ alleviated PH degree and excessive strain on the right heart functions in rats with Su/Hx-induced PH. We identified the candidate target genes for TMZ treatment during PH development. Interestingly, the mRNA levels of the fatty acid transporters were substantially downregulated by TMZ administration in the lungs with Su/Hx-induced PH. Notably, TMZ suppressed excessive proliferation of human pulmonary artery smooth muscle cells under hypoxic conditions. Our study suggests that TMZ ameliorates PH development by involving energy metabolism in the lungs and heart.
Collapse
Affiliation(s)
- Asako Yanagisawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jun-Dal Kim
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Takayuki Kobayashi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoko Misawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Pulmonary Medicine, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Takayuki Jujo-Sanada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, National Institutes of Biomedical Innovation, Osaka, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin-Ichi Muroi
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - So-Ichiro Sasaki
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yoshimi Nakagawa
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - Yoshitoshi Kasuya
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Molecular and Systems Pharmacology, Faculty of Pharmacy, Juntendo University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
14
|
Kazmirczak F, Moon R, Vogel NT, Tollison W, T.Lahti M, Carney JP, Mendelson JB, Markowski T, Higgins L, Murray K, Guerrero C, Prins KW. Ferroptosis Inhibition Combats Metabolic Derangements and Improves Cardiac Function in Pulmonary Artery Banded Pigs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590907. [PMID: 39484509 PMCID: PMC11526868 DOI: 10.1101/2024.04.24.590907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Right heart failure (RHF) is a leading cause of mortality in multiple cardiovascular diseases and preclinical and human data suggest impaired metabolism is a significant contributor to right-sided cardiac dysfunction. Ferroptosis is a nonapopotic form of cell death driven by impaired metabolism. Rodent data suggests ferroptosis inhibition can restore mitochondrial electron transport chain function and enhance cardiac contractility in left heart failure models, but the effects of ferroptosis inhibition in translational large animal models of RHF are unknown. Here, we showed ferrostatin-1 mediated ferroptosis antagonism improve right heart structure and function in pulmonary artery banded pigs. Molecularly, ferrostatin-1 restored mitochondrial cristae structure and combatted downregulation of electron transport chain proteins. Metabolomics and lipidomics analyses revealed ferrostatin-1 improved fatty acid metabolism. Thus, these translational data suggest ferroptosis may be a therapeutic target for RHF.
Collapse
Affiliation(s)
- Felipe Kazmirczak
- Cardiovascular Division, Lillehei Heart Institute, University of Minnesota
| | - Ryan Moon
- Cardiovascular Division, Lillehei Heart Institute, University of Minnesota
| | - Neal T. Vogel
- Cardiovascular Division, Lillehei Heart Institute, University of Minnesota
| | - Walt Tollison
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota
| | - Matt T.Lahti
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota
| | - John P. Carney
- Experimental Surgical Services Laboratory, Department of Surgery, University of Minnesota
| | - Jenna B Mendelson
- Department of Integrative Biology and Physiology, University of Minnesota
| | - Todd Markowski
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota
| | - LeeAnn Higgins
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota
| | - Kevin Murray
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota
| | - Candace Guerrero
- Center for Metabolomics and Proteomics, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota
| | - Kurt W. Prins
- Cardiovascular Division, Lillehei Heart Institute, University of Minnesota
| |
Collapse
|
15
|
Flores K, Almeida C, Arriaza K, Pena E, El Alam S. mTOR in the Development of Hypoxic Pulmonary Hypertension Associated with Cardiometabolic Risk Factors. Int J Mol Sci 2024; 25:11023. [PMID: 39456805 PMCID: PMC11508063 DOI: 10.3390/ijms252011023] [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: 09/12/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
The pathophysiology of pulmonary hypertension is complex and multifactorial. It is a disease characterized by increased pulmonary vascular resistance at the level due to sustained vasoconstriction and remodeling of the pulmonary arteries, which triggers an increase in the mean pulmonary artery pressure and subsequent right ventricular hypertrophy, which in some cases can cause right heart failure. Hypoxic pulmonary hypertension (HPH) is currently classified into Group 3 of the five different groups of pulmonary hypertensions, which are determined according to the cause of the disease. HPH mainly develops as a product of lung diseases, among the most prevalent causes of obstructive sleep apnea (OSA), chronic obstructive pulmonary disease (COPD), or hypobaric hypoxia due to exposure to high altitudes. Additionally, cardiometabolic risk factors converge on molecular mechanisms involving overactivation of the mammalian target of rapamycin (mTOR), which correspond to a central axis in the development of HPH. The aim of this review is to summarize the role of mTOR in the development of HPH associated with metabolic risk factors and its therapeutic alternatives, which will be discussed in this review.
Collapse
Affiliation(s)
| | | | - Karem Arriaza
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1110939, Chile; (K.F.); (C.A.); (E.P.); (S.E.A.)
| | | | | |
Collapse
|
16
|
Abolfazli S, Butler AE, Kesharwani P, Sahebkar A. The beneficial impact of curcumin on cardiac lipotoxicity. J Pharm Pharmacol 2024; 76:1269-1283. [PMID: 39180454 DOI: 10.1093/jpp/rgae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/02/2024] [Indexed: 08/26/2024]
Abstract
Lipotoxicity is defined as a prolonged metabolic imbalance of lipids that results in ectopic fat distribution in peripheral organs such as the liver, heart, and kidney. The harmful consequences of excessive lipid accumulation in cardiomyocytes cause cardiac lipotoxicity, which alters the structure and function of the heart. Obesity and diabetes are linked to lipotoxic cardiomyopathy. These anomalies might be caused by a harmful metabolic shift that accumulates toxic lipids and shifts glucose oxidation to less fatty acid oxidation. Research has linked fatty acids, fatty acyl coenzyme A, diacylglycerol, and ceramide to lipotoxic stress in cells. This stress can be brought on by apoptosis, impaired insulin signaling, endoplasmic reticulum stress, protein kinase C activation, p38 Ras-mitogen-activated protein kinase (MAPK) activation, or modification of peroxisome proliferator-activated receptors (PPARs) family members. Curcuma longa is used to extract curcumin, a hydrophobic polyphenol derivative with a variety of pharmacological characteristics. Throughout the years, curcumin has been utilized as an anti-inflammatory, antioxidant, anticancer, hepatoprotective, cardioprotective, anti-diabetic, and anti-obesity drug. Curcumin reduces cardiac lipotoxicity by inhibiting apoptosis and decreasing the expression of apoptosis-related proteins, reducing the expression of inflammatory cytokines, activating the autophagy signaling pathway, and inhibiting the expression of endoplasmic reticulum stress marker proteins.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University Medical Science, Sari, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Hemnes AR, Celermajer DS, D'Alto M, Haddad F, Hassoun PM, Prins KW, Naeije R, Vonk Noordegraaf A. Pathophysiology of the right ventricle and its pulmonary vascular interaction. Eur Respir J 2024; 64:2401321. [PMID: 39209482 PMCID: PMC11525331 DOI: 10.1183/13993003.01321-2024] [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: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024]
Abstract
The right ventricle and its stress response is perhaps the most important arbiter of survival in patients with pulmonary hypertension of many causes. The physiology of the cardiopulmonary unit and definition of right heart failure proposed in the 2018 World Symposium on Pulmonary Hypertension have proven useful constructs in subsequent years. Here, we review updated knowledge of basic mechanisms that drive right ventricular function in health and disease, and which may be useful for therapeutic intervention in the future. We further contextualise new knowledge on assessment of right ventricular function with a focus on metrics readily available to clinicians and updated understanding of the roles of the right atrium and tricuspid regurgitation. Typical right ventricular phenotypes in relevant forms of pulmonary vascular disease are reviewed and recent studies of pharmacological interventions on chronic right ventricular failure are discussed. Finally, unanswered questions and future directions are proposed.
Collapse
Affiliation(s)
- Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David S Celermajer
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Michele D'Alto
- Department of Cardiology, Monaldi Hospital, Naples, Italy
| | - Francois Haddad
- Division of Cardiovascular Medicine, Stanford University and Stanford Cardiovascular Institute, Palo Alto, CA, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kurt W Prins
- Lillehei Heart Institute, Cardiovascular Division, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | - Anton Vonk Noordegraaf
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Tóth EN, Celant LR, Niglas M, Jansen S, Tramper J, Baxan N, Ashek A, Wessels JN, Marcus JT, Meijboom LJ, Houweling AC, Nossent EJ, Aman J, Grynblat J, Perros F, Montani D, Vonk Noordegraaf A, Zhao L, de Man FS, Bogaard HJ. Deep phenotyping of unaffected carriers of pathogenic BMPR2 variants screened for pulmonary arterial hypertension. Eur Respir J 2024; 64:2400442. [PMID: 38991711 PMCID: PMC11447285 DOI: 10.1183/13993003.00442-2024] [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: 03/04/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION Pathogenic variants in the gene encoding for BMPR2 are a major genetic risk factor for heritable pulmonary arterial hypertension. Owing to incomplete penetrance, deep phenotyping of unaffected carriers of a pathogenic BMPR2 variant through multimodality screening may aid in early diagnosis and identify susceptibility traits for future development of pulmonary arterial hypertension. METHODS 28 unaffected carriers (44±16 years, 57% female) and 21 healthy controls (44±18 years, 48% female) underwent annual screening, including cardiac magnetic resonance imaging, transthoracic echocardiography, cardiopulmonary exercise testing and right heart catheterisation. Right ventricular pressure-volume loops were constructed to assess load-independent contractility and compared with a healthy control group. A transgenic Bmpr2Δ71Ex1/+ rat model was employed to validate findings from humans. RESULTS Unaffected carriers had lower indexed right ventricular end-diastolic (79.5±17.6 mL·m-2 versus 62.7±15.3 mL·m-2; p=0.001), end-systolic (34.2±10.5 mL·m-2 versus 27.1±8.3 mL·m-2; p=0.014) and left ventricular end-diastolic (68.9±14.1 mL·m-2 versus 58.5±10.7 mL·m-2; p=0.007) volumes than control subjects. Bmpr2Δ71Ex1/+ rats were also observed to have smaller cardiac volumes than wild-type rats. Pressure-volume loop analysis showed that unaffected carriers had significantly higher afterload (arterial elastance 0.15±0.06 versus 0.27±0.08 mmHg·mL-1; p<0.001) and end-systolic elastance (0.28±0.07 versus 0.35±0.10 mmHg·mL-1; p=0.047) in addition to lower right ventricular pulmonary artery coupling (end-systolic elastance/arterial elastance 2.24±1.03 versus 1.36±0.37; p=0.006). During the 4-year follow-up period, two unaffected carriers developed pulmonary arterial hypertension, with normal N-terminal pro-brain natriuretic peptide and transthoracic echocardiography indices at diagnosis. CONCLUSION Unaffected BMPR2 mutation carriers have an altered cardiac phenotype mimicked in Bmpr2Δ71Ex1/+ transgenic rats. Future efforts to establish an effective screening protocol for individuals at risk for developing pulmonary arterial hypertension warrant longer follow-up periods.
Collapse
Affiliation(s)
- Eszter N Tóth
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
- Contributed equally
| | - Lucas R Celant
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
- Contributed equally
| | - Marili Niglas
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Samara Jansen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Jelco Tramper
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Nicoleta Baxan
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Ali Ashek
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Jeroen N Wessels
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - J Tim Marcus
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, The Netherlands
| | - Lilian J Meijboom
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Radiology and Nuclear Medicine, Amsterdam, The Netherlands
| | - Arjan C Houweling
- Amsterdam UMC location AMC, Department of Human Genetics, Amsterdam, The Netherlands
| | - Esther J Nossent
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
| | - Jurjan Aman
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
| | - Julien Grynblat
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Marie Lannelongue Hospital and Bicêtre Hospital, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, School of Medicine Gif-sur-Yvette, Gif-sur-Yvette, France
- M3C-Necker, Hôpital Necker-Enfants Malades, AP-HP, Université de Paris Cité, Cardiologie Congénitale et Pédiatrique, Paris, France
| | - Frédéric Perros
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - David Montani
- INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Marie Lannelongue Hospital and Bicêtre Hospital, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- Université Paris-Saclay, School of Medicine Gif-sur-Yvette, Gif-sur-Yvette, France
| | - Anton Vonk Noordegraaf
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Lan Zhao
- Imperial College London, National Heart and Lung Institute, London, UK
| | - Frances S de Man
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pulmonary Medicine, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Hemnes A, Fortune N, Simon K, Trenary IA, Shay S, Austin E, Young JD, Britain E, West J, Talati M. A multimodal approach identifies lactate as a central feature of right ventricular failure that is detectable in human plasma. Front Med (Lausanne) 2024; 11:1387195. [PMID: 39346939 PMCID: PMC11428650 DOI: 10.3389/fmed.2024.1387195] [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: 02/16/2024] [Accepted: 08/23/2024] [Indexed: 10/01/2024] Open
Abstract
Background In PAH metabolic abnormalities in multiple pathways are well-recognized features of right ventricular dysfunction, however, prior work has focused mainly on the use of a single "omic" modality to describe a single deranged pathway. We integrated metabolomic and epigenomic data using transcriptomics in failing and non-failing RVs from a rodent model to provide novel mechanistic insight and translated these findings to accessible human specimens by correlation with plasma from PAH patients. Methods Study was conducted in a doxycycline-inducible BMPR2 mutant mouse model of RV failure. Plasma was collected from controls and PAH patients. Transcriptomic and metabolomic analyses were done on mouse RV tissue and human plasma. For mouse RV, we layered metabolomic and transcriptomic data for multiple metabolic pathways and compared our findings with metabolomic and transcriptomic data obtained for human plasma. We confirmed our key findings in cultured cardiomyocyte cells with BMPR2 mutation. Results In failing mouse RVs, (1) in the glycolysis pathway, glucose is converted to lactate via aerobic glycolysis, but may also be utilized for glycogen, fatty acid, and nucleic acid synthesis, (2) in the fatty acid pathway, FAs are accumulated in the cytoplasm because the transfer of FAs to mitochondria is reduced, however, the ß-oxidation pathway is likely to be functional. (3) the TCA cycle is altered at multiple checkpoints and accumulates citrate, and the glutaminolysis pathway is not activated. In PAH patients, plasma metabolic and transcriptomic data indicated that unlike in the failing BMPR2 mutant RV, expression of genes and metabolites measured for the glycolysis pathway, FA pathway, TCA cycle, and glutaminolysis pathway were increased. Lactate was the only metabolite that was increased both in RV and circulation. We confirmed using a stable isotope of lactate that cultured cardiomyocytes with mutant BMPR2 show a modest increase in endogenous lactate, suggesting a possibility of an increase in lactate production by cardiomyocytes in failing BMPR2 mutant RV. Conclusion In the failing RV with mutant BMPR2, lactate is produced by RV cardiomyocytes and may be secreted out, thereby increasing lactate in circulation. Lactate can potentially serve as a marker of RV dysfunction in PAH, which warrants investigation.
Collapse
Affiliation(s)
- Anna Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Niki Fortune
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Katie Simon
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Evan Britain
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Megha Talati
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
20
|
Namazi M, Eftekhar SP, Mosaed R, Shiralizadeh Dini S, Hazrati E. Pulmonary Hypertension and Right Ventricle: A Pathophysiological Insight. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2024; 18:11795468241274744. [PMID: 39257563 PMCID: PMC11384539 DOI: 10.1177/11795468241274744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 07/21/2024] [Indexed: 09/12/2024]
Abstract
Background Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by elevated pulmonary vascular pressure. Long-term PH, irrespective of its etiology, leads to increased right ventricular (RV) pressure, RV hypertrophy, and ultimately, RV failure. Main body Research indicates that RV failure secondary to hypertrophy remains the primary cause of mortality in pulmonary arterial hypertension (PAH). However, the impact of PH on RV structure and function under increased overload remains incompletely understood. Several mechanisms have been proposed, including extracellular remodeling, RV hypertrophy, metabolic disturbances, inflammation, apoptosis, autophagy, endothelial-to-mesenchymal transition, neurohormonal dysregulation, capillary rarefaction, and ischemia. Conclusions Studies have demonstrated the significant role of oxidative stress in the development of RV failure. Understanding the interplay among these mechanisms is crucial for the prevention and management of RV failure in patients with PH.
Collapse
Affiliation(s)
- Mehrshad Namazi
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Mosaed
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Hazrati
- Trauma and Surgery Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Liu X, Wu D, Bao C, Huang Z, Wang W, Sun L, Qiu L. Identification of fatty acid metabolism signature genes in patients with pulmonary arterial hypertension using WGCNA and machine learning. J Int Med Res 2024; 52:3000605241277740. [PMID: 39324181 PMCID: PMC11437540 DOI: 10.1177/03000605241277740] [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] [Indexed: 09/27/2024] Open
Abstract
OBJECTIVE To investigate the signature genes of fatty acid metabolism and their association with immune cells in pulmonary arterial hypertension (PAH). METHODS Fatty acid metabolism-related genes were obtained from the GeneCards database. In this retrospective study, a PAH-related dataset was downloaded from the Gene Expression Omnibus database and analyzed to identify differentially expressed genes (DEGs). Weighted gene co-expression network analysis (WGCNA) and machine learning algorithms, including least absolute shrinkage and selection operator (LASSO) and random forest, were used to identify the signature genes. Diagnostic efficiency was assessed by receiver operating characteristic (ROC) curve analysis and a nomogram. Immune cell infiltration was subsequently classified using CIBERSORT. RESULTS In total, 817 DEGs were screened from the GSE33463 dataset. The data were clustered into six modules via WGCNA, and the MEdarkred module was significantly related to PAH. The LASSO and random forest algorithms identified five signature genes: ARV1, KCNJ2, PEX11B, PITPNC1, and SCO1. The areas under the ROC curves of these signature genes were 0.917, 0.934, 0.947, 0.963, and 0.940, respectively. CIBERSORT suggested these signature genes may participate in immune cell infiltration. CONCLUSIONS ARV1, KCNJ2, PEX11B, PITPNC1, and SCO1 show remarkable diagnostic performance in PAH and are involved in immune cell infiltration.
Collapse
Affiliation(s)
- Xibang Liu
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Dandan Wu
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Chunmiao Bao
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Zeen Huang
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Weiwei Wang
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Lili Sun
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| | - Lin Qiu
- Department of Respiratory and Critical Care Medicine, Yuyao People's Hospital of Zhejiang Province (Affiliated Yangming Hospital), Yuyao, China
| |
Collapse
|
22
|
Shelburne NJ, Nian H, Beck GJ, Casanova NG, Desai AA, DuBrock HM, Erzurum S, Frantz RP, Hassoun PM, Hill NS, Horn EM, Jacob MS, Jellis CL, Joseloff E, Kwon DH, Brett Larive A, Leopold JA, Park MM, Rischard FP, Rosenzweig EB, Vanderpool RR, Yu C, Hemnes AR. Association of Male Sex With Worse Right Ventricular Function and Survival in Pulmonary Hypertension in the Redefining Pulmonary Hypertension Through Pulmonary Vascular Disease Phenomics Cohort. CHEST PULMONARY 2024; 2:100046. [PMID: 39524046 PMCID: PMC11548889 DOI: 10.1016/j.chpulm.2024.100046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Sex-based differences are important in the development and progression of pulmonary arterial hypertension. However, it is not established whether these differences are generalizable to all forms of pulmonary hypertension (PH). RESEARCH QUESTION What are the sex-based differences in right ventricle (RV) function and transplant-free survival in patients with PH from the Redefining Pulmonary Hypertension Through Pulmonary Vascular Disease Phenomics (PVDOMICS) cohort? STUDY DESIGN AND METHODS Patients with PH enrolled in the PVDOMICS cohort study underwent right heart catheterization, cardiac MRI, and echocardiography. A multivariable linear regression model was used to investigate the interactive effect between sex and pulmonary vascular resistance (PVR) on RV ejection fraction (RVEF). Effects of sex, RVEF, and PVR on transplant-free survival were assessed using a Cox proportional hazards model. RESULTS Seven hundred fifty patients with PH (62.8% female) were enrolled, including 397 patients with groups 2 through 5 PH. Patients with group 1 PH were predominantly female (73.4%). Male patients showed multiple markers of worse RV function with significantly lower RVEF (adjusted difference, 5.5%; 95% CI, 3.2%-7.8%; P < .001) on cardiac MRI and lower RV fractional shortening (adjusted difference, 4.0%; 95% CI, 2.3%-5.8%; P < .001) and worse RV free-wall longitudinal strain (adjusted difference, 2.4%; 95% CI, 1.2%-3.6%; P < .001) on echocardiography. Significant interaction was noted between PVR and sex on RVEF, with the largest sex-based differences in RVEF noted at mild to moderate PVR elevation. Male sex was associated with decreased transplant-free survival (adjusted hazard ratio, 1.46; 95% CI, 1.07-1.98; P = .02), partially mediated by differences in RVEF (P = .003). INTERPRETATION In patients with PH in the PVDOMICS study, female sex was more common, whereas male sex was associated with worse RV function and decreased transplant-free survival, most notably at mild to moderate elevation of PVR.
Collapse
Affiliation(s)
- Nicholas J Shelburne
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; INTEGRIS Advanced Cardiopulmonary Care, INTEGRIS Baptist Medical Center, Oklahoma City, OK
| | - Hui Nian
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Gerald J Beck
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland
| | - Nancy G Casanova
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona, Tucson, AZ
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN
| | - Hilary M DuBrock
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Serpil Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland; Respiratory Institute, Cleveland Clinic, Cleveland
| | - Robert P Frantz
- Department of Internal Medicine, the Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore
| | - Nicholas S Hill
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Evelyn M Horn
- Division of Cardiology, Department of Pediatrics and Medicine, Columbia University Medical Center-New York Presbyterian Hospital
| | - Miriam S Jacob
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland
| | | | | | - Deborah H Kwon
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland
| | - A Brett Larive
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland
| | - Jane A Leopold
- Tufts Medical Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Margaret M Park
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland
| | - Franz P Rischard
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Arizona, Tucson, AZ
| | - Erika B Rosenzweig
- Perkin Heart Failure Center, Weill Cornell Medical Center, the Division of Pediatric Cardiology, Department of Pediatrics and Medicine, Columbia University Medical Center-New York Presbyterian Hospital
| | - Rebecca R Vanderpool
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Chang Yu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [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: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
24
|
Singh I, Waxman AB. The casting of invasive cardiopulmonary exercise testing: towards a common goal. Eur Respir J 2024; 64:2400783. [PMID: 38991723 DOI: 10.1183/13993003.00783-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 07/13/2024]
Affiliation(s)
- Inderjit Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
25
|
Brittain EL, Lindsey A, Burke K, Agrawal V, Robbins I, Pugh M, Calcutt MW, Mallugari R, West J, Nian H, Hemnes AR. Carnitine consumption and effect of oral supplementation in human pulmonary arterial hypertension: A pilot study. Pulm Circ 2024; 14:e12425. [PMID: 39157054 PMCID: PMC11327271 DOI: 10.1002/pul2.12425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Carnitine is required to transport fatty acid across the mitochondrial membrane to undergo beta oxidation. In addition to disorders of fatty acid metabolism, a relative carnitine deficiency has been reported in pulmonary arterial hypertension (PAH). Here we performed an observational study in which food and supplement consumption were collected in an observation period followed by open label administration of a carnitine supplement to determine feasibility of increasing plasma carnitine levels in humans PAH. We confirmed that relative carnitine deficiency in PAH is not due to reduced dietary consumption and that plasma levels of carnitine can be increased in PAH patients with supplementation that is well tolerated.
Collapse
Affiliation(s)
- Evan L. Brittain
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Alisha Lindsey
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kelly Burke
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vineet Agrawal
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ivan Robbins
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Meredith Pugh
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - M. Wade Calcutt
- Department of BiochemistryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ravi Mallugari
- Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - James West
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Hui Nian
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
26
|
Coursen JC, Tuhy T, Naranjo M, Woods A, Hummers LK, Shah AA, Suresh K, Visovatti SH, Mathai SC, Hassoun PM, Damico RL, Simpson CE. Aberrant long-chain fatty acid metabolism associated with evolving systemic sclerosis-associated pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L54-L64. [PMID: 38651694 PMCID: PMC11380974 DOI: 10.1152/ajplung.00057.2024] [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: 02/16/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
We sought to investigate differential metabolism in patients with systemic sclerosis (SSc) who develop pulmonary arterial hypertension (PAH) versus those who do not, as a method of identifying potential disease biomarkers. In a nested case-control design, serum metabolites were assayed in SSc subjects who developed right heart catheterization-confirmed PAH (n = 22) while under surveillance in a longitudinal cohort from Johns Hopkins, then compared with metabolites assayed in matched SSc patients who did not develop PAH (n = 22). Serum samples were collected at "proximate" (within 12 months) and "distant" (within 1-5 yr) time points relative to PAH diagnosis. Metabolites were identified using liquid chromatography-mass spectroscopy (LC-MS). An LC-MS dataset from SSc subjects with either mildly elevated pulmonary pressures or overt PAH from the University of Michigan was compared. Differentially abundant metabolites were tested as predictors of PAH in two additional validation SSc cohorts. Long-chain fatty acid metabolism (LCFA) consistently differed in SSc-PAH versus SSc without PH. LCFA metabolites discriminated SSc-PAH patients with mildly elevated pressures in the Michigan cohort and predicted SSc-PAH up to 2 yr before clinical diagnosis in the Hopkins cohort. Acylcholines containing LCFA residues and linoleic acid metabolites were most important for discriminating SSc-PAH. Combinations of acylcholines and linoleic acid metabolites provided good discrimination of SSc-PAH across cohorts. Aberrant lipid metabolism is observed throughout the evolution of PAH in SSc. Lipidomic signatures of abnormal LCFA metabolism distinguish SSc-PAH patients from those without PH, including before clinical diagnosis and in mild disease.NEW & NOTEWORTHY Abnormal lipid metabolism is evident across time in the development of SSc-PAH, and dysregulated long-chain fatty acid metabolism predicts overt PAH.
Collapse
Affiliation(s)
- Julie C Coursen
- Division of Hospital Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Tijana Tuhy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Mario Naranjo
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, Pennsylvania, United States
| | - Adrianne Woods
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Laura K Hummers
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Ami A Shah
- Division of Rheumatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Scott H Visovatti
- Division of Cardiology, Department of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Stephen C Mathai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Catherine E Simpson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
27
|
García-Lunar I, Jorge I, Sáiz J, Solanes N, Dantas AP, Rodríguez-Arias JJ, Ascaso M, Galán-Arriola C, Jiménez FR, Sandoval E, Nuche J, Moran-Garrido M, Camafeita E, Rigol M, Sánchez-Gonzalez J, Fuster V, Vázquez J, Barbas C, Ibáñez B, Pereda D, García-Álvarez A. Metabolic changes contribute to maladaptive right ventricular hypertrophy in pulmonary hypertension beyond pressure overload: an integrative imaging and omics investigation. Basic Res Cardiol 2024; 119:419-433. [PMID: 38536505 PMCID: PMC11143050 DOI: 10.1007/s00395-024-01041-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/10/2024] [Accepted: 02/10/2024] [Indexed: 06/01/2024]
Abstract
Right ventricular (RV) failure remains the strongest determinant of survival in pulmonary hypertension (PH). We aimed to identify relevant mechanisms, beyond pressure overload, associated with maladaptive RV hypertrophy in PH. To separate the effect of pressure overload from other potential mechanisms, we developed in pigs two experimental models of PH (M1, by pulmonary vein banding and M2, by aorto-pulmonary shunting) and compared them with a model of pure pressure overload (M3, pulmonary artery banding) and a sham-operated group. Animals were assessed at 1 and 8 months by right heart catheterization, cardiac magnetic resonance and blood sampling, and myocardial tissue was analyzed. Plasma unbiased proteomic and metabolomic data were compared among groups and integrated by an interaction network analysis. A total of 33 pigs completed follow-up (M1, n = 8; M2, n = 6; M3, n = 10; and M0, n = 9). M1 and M2 animals developed PH and reduced RV systolic function, whereas animals in M3 showed increased RV systolic pressure but maintained normal function. Significant plasma arginine and histidine deficiency and complement system activation were observed in both PH models (M1&M2), with additional alterations to taurine and purine pathways in M2. Changes in lipid metabolism were very remarkable, particularly the elevation of free fatty acids in M2. In the integrative analysis, arginine-histidine-purines deficiency, complement activation, and fatty acid accumulation were significantly associated with maladaptive RV hypertrophy. Our study integrating imaging and omics in large-animal experimental models demonstrates that, beyond pressure overload, metabolic alterations play a relevant role in RV dysfunction in PH.
Collapse
Affiliation(s)
- Inés García-Lunar
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, University Hospital La Moraleja, Madrid, Spain
| | - Inmaculada Jorge
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Jorge Sáiz
- Centre of Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Núria Solanes
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Ana Paula Dantas
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Juan José Rodríguez-Arias
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - María Ascaso
- Department of Cardiovascular Surgery, Hospital Clínic Barcelona, Barcelona, Spain
| | - Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Rafael Jiménez
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | - Elena Sandoval
- Department of Cardiovascular Surgery, Hospital Clínic Barcelona, Barcelona, Spain
| | - Jorge Nuche
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiology, Hospital 12 de Octubre, Madrid, Spain
| | - Maria Moran-Garrido
- Centre of Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Emilio Camafeita
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Montserrat Rigol
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain
| | | | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Mount Sinai Fuster Heart Hospital, Mount Sinai Hospital, New York, NY, USA
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Coral Barbas
- Centre of Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- IIS-Fundación Jiménez Diaz University Hospital, Madrid, Spain
| | - Daniel Pereda
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Department of Cardiovascular Surgery, Hospital Clínic Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Ana García-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Department of Cardiology, Hospital Clínic Barcelona-IDIBAPS, Universitat de Barcelona, Villarroel 170, 08036, Barcelona, Spain.
- Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
28
|
Luo T, Wu H, Zhu W, Zhang L, Huang Y, Yang X. Emerging therapies: Potential roles of SGLT2 inhibitors in the management of pulmonary hypertension. Respir Med 2024; 227:107631. [PMID: 38631526 DOI: 10.1016/j.rmed.2024.107631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
Pulmonary hypertension (PH) is a pathophysiological disorder that may involve multiple clinical conditions and may be associated with a variety of cardiovascular and respiratory diseases. Pulmonary hypertension due to left heart disease (PH-LHD) currently lacks targeted therapies, while Pulmonary arterial hypertension (PAH), despite approved treatments, carries considerable residual risk. Metabolic dysfunction has been linked to the pathogenesis and prognosis of PH through various studies, with emerging metabolic agents offering a potential avenue for improving patient outcomes. Sodium-glucose cotransporter 2 inhibitor (SGLT-2i), a novel hypoglycemic agent, could ameliorate metabolic dysfunction and exert cardioprotective effects. Recent small-scale studies suggest SGLT-2i treatment may improve pulmonary artery pressure in patients with PH-LHD, and the PAH animal model shows that SGLT-2i can reduce pulmonary vascular remodeling and prevent progression in PAH, suggesting potential benefits for patients with PH-LHD and perhaps PAH. This review aims to succinctly review PH's pathophysiology, and the connection between metabolic dysfunction and PH, and investigate the prospective mechanisms of action of SGLT-2i in PH-LHD and PAH management.
Collapse
Affiliation(s)
- Taimin Luo
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, 610000, China
| | - Hui Wu
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Wanlong Zhu
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; Department of Pharmacy, Panzhihua Second People's Hospital, Panzhihua, 617000, China
| | - Liaoyun Zhang
- Department of Pharmacy, Sichuan Provincial Maternity and Child Health Care Hospital & Women's and Children's Hospital, Chengdu, 610000, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
29
|
Ba H, Guo Y, Jiang Y, Li Y, Dai X, Liu Y, Li X. Unveiling the metabolic landscape of pulmonary hypertension: insights from metabolomics. Respir Res 2024; 25:221. [PMID: 38807129 PMCID: PMC11131231 DOI: 10.1186/s12931-024-02775-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/14/2024] [Indexed: 05/30/2024] Open
Abstract
Pulmonary hypertension (PH) is regarded as cardiovascular disease with an extremely poor prognosis, primarily due to irreversible vascular remodeling. Despite decades of research progress, the absence of definitive curative therapies remains a critical challenge, leading to high mortality rates. Recent studies have shown that serious metabolic disorders generally exist in PH animal models and patients of PH, which may be the cause or results of the disease. It is imperative for future research to identify critical biomarkers of metabolic dysfunction in PH pathophysiology and to uncover metabolic targets that could enhance diagnostic and therapeutic strategies. Metabolomics offers a powerful tool for the comprehensive qualitative and quantitative analysis of metabolites within specific organisms or cells. On the basis of the findings of the metabolomics research on PH, this review summarizes the latest research progress on metabolic pathways involved in processes such as amino acid metabolism, carbohydrate metabolism, lipid metabolism, and nucleotide metabolism in the context of PH.
Collapse
Affiliation(s)
- Huixue Ba
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yingfan Guo
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yujie Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xuejing Dai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China
| | - Yuan Liu
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, China.
| |
Collapse
|
30
|
Hansmann G, Chouvarine P. Fatty acid oxidation is decreased in human right heart failure: Comment on Mendelson JB et al. Multi-omic and multispecies analysis of right ventricular dysfunction. J Heart Lung Transplant 2024; 43:861-862. [PMID: 38284965 DOI: 10.1016/j.healun.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024] Open
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany.
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany; European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany
| |
Collapse
|
31
|
Liu B, Yi D, Li S, Ramirez K, Xia X, Cao Y, Zhao H, Tripathi A, Qiu S, Kala M, Rafikov R, Gu H, de jesus Perez V, Lemay SE, Glembotski CC, Knox KS, Bonnet S, Kalinichenko VV, Zhao YY, Fallon MB, Boucherat O, Dai Z. Single-cell and Spatial Transcriptomics Identified Fatty Acid-binding Proteins Controlling Endothelial Glycolytic and Arterial Programming in Pulmonary Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.11.579846. [PMID: 38370670 PMCID: PMC10871348 DOI: 10.1101/2024.02.11.579846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease characterized by obliterative vascular remodeling and persistent increase of vascular resistance, leading to right heart failure and premature death. Understanding the cellular and molecular mechanisms will help develop novel therapeutic approaches for PAH patients. Single-cell RNA sequencing (scRNAseq) analysis found that both FABP4 and FABP5 were highly induced in endothelial cells (ECs) of Egln1 Tie2Cre (CKO) mice, which was also observed in pulmonary arterial ECs (PAECs) from idiopathic PAH (IPAH) patients, and in whole lungs of pulmonary hypertension (PH) rats. Plasma levels of FABP4/5 were upregulated in IPAH patients and directly correlated with severity of hemodynamics and biochemical parameters using plasma proteome analysis. Genetic deletion of both Fabp4 and 5 in CKO mice (Egln1 Tie2Cre /Fabp4-5 -/- ,TKO) caused a reduction of right ventricular systolic pressure (RVSP) and RV hypertrophy, attenuated pulmonary vascular remodeling and prevented the right heart failure assessed by echocardiography, hemodynamic and histological analysis. Employing bulk RNA-seq and scRNA-seq, and spatial transcriptomic analysis, we showed that Fabp4/5 deletion also inhibited EC glycolysis and distal arterial programming, reduced ROS and HIF-2α expression in PH lungs. Thus, PH causes aberrant expression of FABP4/5 in pulmonary ECs which leads to enhanced ECs glycolysis and distal arterial programming, contributing to the accumulation of arterial ECs and vascular remodeling and exacerbating the disease.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Shuai Li
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Xiaomei Xia
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Yanhong Cao
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Hanqiu Zhao
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Ankit Tripathi
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Shenfeng Qiu
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Mrinalini Kala
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Ruslan Rafikov
- Department of Medicine, Indiana University College of Medicine, Indianapolis, IN, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | | | - Sarah-Eve Lemay
- Pulmonary Hypertension and Vascular Biology Research Group, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Christopher C. Glembotski
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Kenneth S Knox
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Sebastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Vladimir V. Kalinichenko
- Division of Neonatology, Phoenix Children’s Hospital, Phoenix, AZ, USA
- Phoenix Children’s Health Research Institute, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - You-Yang Zhao
- Program for Lung and Vascular Biology and Section for Injury Repair and Regeneration Research, Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael B. Fallon
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, University of Arizona, Phoenix, AZ, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Sarver Heart Center, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
32
|
Liu Q, Yang Y, Wu M, Wang M, Yang P, Zheng J, Du Z, Pang Y, Bao L, Niu Y, Zhang R. Hub gene ELK3-mediated reprogramming lipid metabolism regulates phenotypic switching of pulmonary artery smooth muscle cells to develop pulmonary arterial hypertension induced by PM 2.5. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133190. [PMID: 38071773 DOI: 10.1016/j.jhazmat.2023.133190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 02/08/2024]
Abstract
Fine particulate matter (PM2.5) as an environmental pollutant is related with respiratory and cardiovascular diseases. Pulmonary arterial hypertension (PAH) was characterized by incremental pulmonary artery pressure and pulmonary arterial remodeling, leading to right ventricular hypertrophy, and finally cardiac failure and death. The adverse effects on pulmonary artery and the molecular biological mechanism underlying PM2.5-caused PAH has not been elaborated clearly. In the current study, the ambient PM2.5 exposure mice model along with HPASMCs models were established. Based on bioinformatic methods and machine learning algorithms, the hub genes in PAH were screened and then adverse effects on pulmonary artery and potential mechanism was studied. Our results showed that chronic PM2.5 exposure contributed to increased pulmonary artery pressure, pulmonary arterial remodeling and right ventricular hypertrophy in mice. In vitro, PM2.5 induced phenotypic switching in HPASMCs, which served as the early stage of PAH. In mechanism, we investigated that PM2.5-mediated mitochondrial dysfunction could induce phenotypic switching in HPASMCs, which was possibly through reprogramming lipid metabolism. Next, we used machine learning algorithm to identify ELK3 as potential hub gene for mitochondrial fission. Besides, the effect of DNA methylation on ELK3 was further detected in HPASMCs after PM2.5 exposure. The results provided novel directions for protection of pulmonary vasculature injury, against adverse environmental stimuli. This work also provided a new idea for the prevention of PAH, as well as provided experimental evidence for the targeted therapy of PAH.
Collapse
Affiliation(s)
- Qingping Liu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yizhe Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengqi Wu
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Mengruo Wang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Peihao Yang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Jie Zheng
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Zhe Du
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yaxian Pang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Lei Bao
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Yujie Niu
- Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China
| | - Rong Zhang
- Department of Toxicology, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China; Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, Hebei, PR China.
| |
Collapse
|
33
|
Singh N, Al-Naamani N, Brown MB, Long GM, Thenappan T, Umar S, Ventetuolo CE, Lahm T. Extrapulmonary manifestations of pulmonary arterial hypertension. Expert Rev Respir Med 2024; 18:189-205. [PMID: 38801029 PMCID: PMC11713041 DOI: 10.1080/17476348.2024.2361037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
| | - Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, WA
| | - Gary Marshall Long
- Department of Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, IN
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota, Minneapolis, MN
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Corey E. Ventetuolo
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
- Department of Health Services, Policy and Practice, Brown University, Providence, RI
| | - Tim Lahm
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| |
Collapse
|
34
|
Oneglia AP, Szczepaniak LS, Zaha VG, Nelson MD. Myocardial steatosis across the spectrum of human health and disease. Exp Physiol 2024; 109:202-213. [PMID: 38063136 PMCID: PMC10841709 DOI: 10.1113/ep091566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/15/2023] [Indexed: 02/02/2024]
Abstract
Preclinical data strongly suggest that myocardial steatosis leads to adverse cardiac remodelling and left ventricular dysfunction. Using 1 H cardiac magnetic resonance spectroscopy, similar observations have been made across the spectrum of health and disease. The purpose of this brief review is to summarize these recent observations. We provide a brief overview of the determinants of myocardial triglyceride accumulation, summarize the current evidence that myocardial steatosis contributes to cardiac dysfunction, and identify opportunities for further research.
Collapse
Affiliation(s)
- Andrew P. Oneglia
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
| | | | - Vlad G. Zaha
- Division of Cardiology, Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Advanced Imaging Research CenterUniversity of Texas Southwestern Medical CenterArlingtonTexasUSA
| | - Michael D. Nelson
- Applied Physiology and Advanced Imaging Laboratory, Department of Kinesiology, College of Nursing and Health InnovationUniversity of Texas at ArlingtonArlingtonTexasUSA
- Clinical Imaging Research CenterUniversity of Texas at ArlingtonArlingtonTexasUSA
- Center for Healthy Living and LongevityUniversity of Texas at ArlingtonArlingtonTexasUSA
| |
Collapse
|
35
|
Mendelson JB, Sternbach JD, Doyle MJ, Mills L, Hartweck LM, Tollison W, Carney JP, Lahti MT, Bianco RW, Kalra R, Kazmirczak F, Hindmarch C, Archer SL, Prins KW, Martin CM. Multi-omic and multispecies analysis of right ventricular dysfunction. J Heart Lung Transplant 2024; 43:303-313. [PMID: 37783299 PMCID: PMC10841898 DOI: 10.1016/j.healun.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Right ventricular failure (RVF) is a leading cause of morbidity and mortality in multiple cardiovascular diseases, but there are no treatments for RVF as therapeutic targets are not clearly defined. Contemporary transcriptomic/proteomic evaluations of RVF are predominately conducted in small animal studies, and data from large animal models are sparse. Moreover, a comparison of the molecular mediators of RVF across species is lacking. METHODS Transcriptomics and proteomics analyses defined the pathways associated with cardiac magnetic resonance imaging (MRI)-derived values of RV hypertrophy, dilation, and dysfunction in control and pulmonary artery banded (PAB) pigs. Publicly available data from rat monocrotaline-induced RVF and pulmonary arterial hypertension patients with preserved or impaired RV function were used to compare molecular responses across species. RESULTS PAB pigs displayed significant right ventricle/ventricular (RV) hypertrophy, dilation, and dysfunction as quantified by cardiac magnetic resonance imaging. Transcriptomic and proteomic analyses identified pathways associated with RV dysfunction and remodeling in PAB pigs. Surprisingly, disruptions in fatty acid oxidation (FAO) and electron transport chain (ETC) proteins were different across the 3 species. FAO and ETC proteins and transcripts were mostly downregulated in rats but were predominately upregulated in PAB pigs, which more closely matched the human response. All species exhibited similar dysregulation of the dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy pathways. CONCLUSIONS The porcine metabolic molecular signature was more similar to human RVF than rodents. These data suggest there may be divergent molecular responses of RVF across species, and pigs may more accurately recapitulate metabolic aspects of human RVF.
Collapse
Affiliation(s)
- Jenna B Mendelson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
| | - Jacob D Sternbach
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Michelle J Doyle
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Lauren Mills
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Lynn M Hartweck
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Walt Tollison
- Department of Surgery, Experimental Surgical Services Laboratory, University of Minnesota, Minneapolis, Minnesota
| | - John P Carney
- Department of Surgery, Experimental Surgical Services Laboratory, University of Minnesota, Minneapolis, Minnesota
| | - Matthew T Lahti
- Department of Surgery, Experimental Surgical Services Laboratory, University of Minnesota, Minneapolis, Minnesota
| | - Richard W Bianco
- Department of Surgery, Experimental Surgical Services Laboratory, University of Minnesota, Minneapolis, Minnesota
| | - Rajat Kalra
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Felipe Kazmirczak
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Charles Hindmarch
- Queen's Cardiopulmonary Unit, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Queen's Cardiopulmonary Unit, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Kurt W Prins
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota; Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | - Cindy M Martin
- DeBakey Heart and Vascular Center, Houston Methodist, Houston, Texas
| |
Collapse
|
36
|
Pawar SG, Khan N, Salam A, Joshi M, Saravanan PB, Pandey S. The association of Pulmonary Hypertension and right ventricular systolic function - updates in diagnosis and treatment. Dis Mon 2024; 70:101635. [PMID: 37734967 DOI: 10.1016/j.disamonth.2023.101635] [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] [Indexed: 09/23/2023]
Abstract
Right ventricular (RV) systolic function is an essential but neglected component in cardiac evaluation, and its importance to the contribution to overall cardiac function is undermined. It is not only sensitive to the effect of left heart valve disease but is also more sensitive to changes in pressure overload than the left ventricle. Pulmonary Hypertension is the common and well-recognized complication of RV systolic dysfunction. It is also the leading cause of pulmonary valve disease and right ventricular dysfunction. Patients with a high pulmonary artery pressure (PAP) and a low RV ejection fraction have a seven-fold higher risk of death than heart failure patients with a normal PAP and RV ejection fraction. Furthermore, it is an independent predictor of survival in these patients. In this review, we examine the association of right ventricular systolic function with Pulmonary Hypertension by focusing on various pathological and clinical manifestations while assessing their impact. We also explore new 2022 ESC/ERS guidelines for diagnosing and treating right ventricular dysfunction in Pulmonary Hypertension.
Collapse
Affiliation(s)
| | - Nida Khan
- Jinnah Sindh Medical University, Pakistan
| | - Ajal Salam
- Government Medical College Kottayam, Kottayam, Kerala, India
| | - Muskan Joshi
- Tbilisi State Medical University, Tbilisi, Georgia
| | | | | |
Collapse
|
37
|
Fakhry B, Peterson L, Comhair SA, Sharp J, Park MM, Tang WW, Neumann DR, DiFilippo FP, Farha S, Erzurum SC, Mulya A. Blood Cholesterol and Triglycerides Associate with Right Ventricular Function in Pulmonary Hypertension. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.20.24301498. [PMID: 38343848 PMCID: PMC10854346 DOI: 10.1101/2024.01.20.24301498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Blood lipids are dysregulated in pulmonary hypertension (PH). Lower high-density lipoproteins cholesterol (HDL-C) and low-density lipoproteins cholesterol (LDL-C) are associated with disease severity and death in PH. Right ventricle (RV) dysfunction and failure are the major determinants of morbidity and mortality in PH. This study aims to test the hypothesis that dyslipidemia is associated with RV dysfunction in PH. Methods We enrolled healthy control subjects (n=12) and individuals with PH (n=30) (age: 18-65 years old). Clinical characteristics, echocardiogram, 2-[18F] fluoro-2-deoxy-D-glucose positron emission tomography (PET) scan, blood lipids, including total cholesterol (TC), triglycerides (TG), lipoproteins (LDL-C and HDL-C), and N-terminal pro-B type Natriuretic Peptide (NT-proBNP) were determined. Results Individuals with PH had lower HDL-C [PH, 41±12; control, 56±16 mg/dL, p<0.01] and higher TG to HDL-C ratio [PH, 3.6±3.1; control, 2.2±2.2, p<0.01] as compared to controls. TC, TG, and LDL-C were similar between PH and controls. Lower TC and TG were associated with worse RV function measured by RV strain (R=-0.43, p=0.02 and R=-0.37, p=0.05 respectively), RV fractional area change (R=0.51, p<0.01 and R=0.48, p<0.01 respectively), RV end-systolic area (R=-0.63, p<0.001 and R=-0.48, p<0.01 respectively), RV end-diastolic area: R=-0.58, p<0.001 and R=-0.41, p=0.03 respectively), and RV glucose uptake by PET (R=-0.46, p=0.01 and R=-0.30, p=0.10 respectively). NT-proBNP was negatively correlated with TC (R=-0.61, p=0.01) and TG (R=-0.62, p<0.02) in PH. Conclusion These findings confirm dyslipidemia is associated with worse right ventricular function in PH.
Collapse
Affiliation(s)
- Battoul Fakhry
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Laura Peterson
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Suzy A.A. Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jacqueline Sharp
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Margaret M. Park
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States
| | - W.H. Wilson Tang
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, United States
| | | | | | - Samar Farha
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Serpil C. Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anny Mulya
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
38
|
Mocumbi A, Humbert M, Saxena A, Jing ZC, Sliwa K, Thienemann F, Archer SL, Stewart S. Pulmonary hypertension. Nat Rev Dis Primers 2024; 10:1. [PMID: 38177157 DOI: 10.1038/s41572-023-00486-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 01/06/2024]
Abstract
Pulmonary hypertension encompasses a range of conditions directly or indirectly leading to elevated pressures within the pulmonary arteries. Five main groups of pulmonary hypertension are recognized, all defined by a mean pulmonary artery pressure of >20 mmHg: pulmonary arterial hypertension (rare), pulmonary hypertension associated with left-sided heart disease (very common), pulmonary hypertension associated with lung disease (common), pulmonary hypertension associated with pulmonary artery obstructions, usually related to thromboembolic disease (rare), and pulmonary hypertension with unclear and/or multifactorial mechanisms (rare). At least 1% of the world's population is affected, with a greater burden more likely in low-income and middle-income countries. Across all its forms, pulmonary hypertension is associated with adverse vascular remodelling with obstruction, stiffening and vasoconstriction of the pulmonary vasculature. Without proactive management this leads to hypertrophy and ultimately failure of the right ventricle, the main cause of death. In older individuals, dyspnoea is the most common symptom. Stepwise investigation precedes definitive diagnosis with right heart catheterization. Medical and surgical treatments are approved for pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. There are emerging treatments for other forms of pulmonary hypertension; but current therapy primarily targets the underlying cause. There are still major gaps in basic, clinical and translational knowledge; thus, further research, with a focus on vulnerable populations, is needed to better characterize, detect and effectively treat all forms of pulmonary hypertension.
Collapse
Affiliation(s)
- Ana Mocumbi
- Faculdade de Medicina, Universidade Eduardo Mondlane, Maputo, Moçambique.
- Instituto Nacional de Saúde, EN 1, Marracuene, Moçambique.
| | - Marc Humbert
- Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre (Assistance Publique Hôpitaux de Paris), Université Paris-Saclay, INSERM UMR_S 999, Paris, France
- ERN-LUNG, Le Kremlin Bicêtre, Paris, France
| | - Anita Saxena
- Sharma University of Health Sciences, Haryana, New Delhi, India
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Karen Sliwa
- Cape Heart Institute, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
| | - Friedrich Thienemann
- Department of Medicine, Groote Schuur Hospital, Faculty of Health Science, University of Cape Town, Cape Town, South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Simon Stewart
- Institute of Health Research, University of Notre Dame, Fremantle, Western Australia, Australia
| |
Collapse
|
39
|
Engin A. Lipid Storage, Lipolysis, and Lipotoxicity in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:97-129. [PMID: 39287850 DOI: 10.1007/978-3-031-63657-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The ratio of free fatty acid (FFA) turnover decreases significantly with the expansion of white adipose tissue. Adipose tissue and dietary saturated fatty acid levels significantly correlate with an increase in fat cell size and number. The G0/G1 switch gene 2 increases lipid content in adipocytes and promotes adipocyte hypertrophy through the restriction of triglyceride (triacylglycerol: TAG) turnover. Hypoxia in obese adipose tissue due to hypertrophic adipocytes results in excess deposition of extracellular matrix (ECM) components. Cluster of differentiation (CD) 44, as the main receptor of the extracellular matrix component regulates cell-cell and cell-matrix interactions including diet-induced insulin resistance. Excess TAGs, sterols, and sterol esters are surrounded by the phospholipid monolayer surface and form lipid droplets (LDs). Once LDs are formed, they grow up because of the excessive amount of intracellular FFA stored and reach a final size. The ratio of FFA turnover/lipolysis decreases significantly with increases in the degree of obesity. Dysfunctional adipose tissue is unable to expand further to store excess dietary lipids, increased fluxes of plasma FFAs lead to ectopic fatty acid deposition and lipotoxicity. Reduced neo-adipogenesis and dysfunctional lipid-overloaded adipocytes are hallmarks of hypertrophic obesity linked to insulin resistance. Obesity-associated adipocyte death exhibits feature of necrosis-like programmed cell death. Adipocyte death is a prerequisite for the transition from hypertrophic to hyperplastic obesity. Increased adipocyte number in obesity has life-long effects on white adipose tissue mass. The positive correlation between the adipose tissue volume and magnetic resonance imaging proton density fat fraction estimation is used for characterization of the obesity phenotype, as well as the risk stratification and selection of appropriate treatment strategies. In obese patients with type 2 diabetes, visceral adipocytes exposed to chronic/intermittent hyperglycemia develop a new microRNAs' (miRNAs') expression pattern. Visceral preadipocytes memorize the effect of hyperglycemia via changes in miRNAs' expression profile and contribute to the progression of diabetic phenotype. Nonsteroidal anti-inflammatory drugs, metformin, and statins can be beneficial in treating the local or systemic consequences of white adipose tissue inflammation. Rapamycin inhibits leptin-induced LD formation. Collectively, in this chapter, the concept of adipose tissue remodeling in response to adipocyte death or adipogenesis, and the complexity of LD interactions with the other cellular organelles are reviewed. Furthermore, clinical perspective of fat cell turnover in obesity is also debated.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
40
|
Poojary G, Morris N, Joshi MB, Babu AS. Role of Exercise in Pulmonary Hypertension: Evidence from Bench to Bedside. Pulse (Basel) 2024; 12:66-75. [PMID: 39022559 PMCID: PMC11249447 DOI: 10.1159/000539537] [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: 04/03/2024] [Accepted: 05/13/2024] [Indexed: 07/20/2024] Open
Abstract
Background Pulmonary hypertension (PH) is a debilitating condition characterized by elevated pulmonary arterial pressure and progressive vascular remodelling, leading to exercise intolerance. The progression of PAH is regulated at a cellular and molecular level which influences various physiological processes. Exercise plays an important role in improving function in PH. Although the signalling pathways that regulate cardio-protection through exercise have not been fully understood, the positive impact of exercise on the various physiological systems is well established. Summary Exercise has emerged as a potential adjunctive therapy for PH, with growing evidence supporting its beneficial effects on various aspects of the disease pathophysiology. This review highlights the contributions of cellular and molecular pathways and physiological processes to exercise intolerance. Preclinical studies have provided insight into the mechanisms underlying exercise-induced improvements in PH which are modulated through improvements in endothelial function, inflammation, oxidative stress, and mitochondrial function. Along with preclinical studies, various clinical studies have demonstrated that exercise training can lead to significant improvements in exercise capacity, haemodynamics, quality of life, and functional status. Moreover, exercise interventions have been shown to improve skeletal muscle function and enhance pulmonary vascular remodelling, contributing to overall disease management. Further research efforts aimed at better understanding the role of exercise in PH pathophysiology, and refining exercise interventions are warranted to realize its full potential in the management of this complex disease. Key Messages Despite the promising benefits of exercise in PH, several challenges remain, including the optimal intensity, duration, and type of exercise training, as well as patient selection criteria and long-term adherence. Additionally, the mechanisms underlying the observed improvements require further elucidation to optimize exercise protocols and personalize treatment strategies. Nonetheless, exercise represents a promising therapeutic approach that can complement existing pharmacological therapies and improve outcomes in PH patients.
Collapse
Affiliation(s)
- Ganesha Poojary
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Norman Morris
- School of Health Sciences and Social Work, Griffith University, Gold Coast Campus, Southport, QLD, Australia
- Allied Health Research Collaborative, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Abraham Samuel Babu
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
- Department of Cardiology, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Simpson CE, Hemnes AR, Griffiths M, Grunig G, Wilson Tang W, Garcia JGN, Barnard J, Comhair SA, Damico RL, Mathai SC, Hassoun PM, PVDOMICS Study Group. Metabolomic Differences in Connective Tissue Disease-Associated Versus Idiopathic Pulmonary Arterial Hypertension in the PVDOMICS Cohort. Arthritis Rheumatol 2023; 75:2240-2251. [PMID: 37335853 PMCID: PMC10728345 DOI: 10.1002/art.42632] [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: 03/07/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/21/2023]
Abstract
OBJECTIVE Patients with connective tissue disease-associated pulmonary arterial hypertension (CTD-PAH) experience worse survival and derive less benefit from pulmonary vasodilator therapies than patients with idiopathic PAH (IPAH). We sought to identify differential metabolism in patients with CTD-PAH versus patients with IPAH that might underlie these observed clinical differences. METHODS Adult participants with CTD-PAH (n = 141) and IPAH (n = 165) from the Pulmonary Vascular Disease Phenomics (PVDOMICS) study were included. Detailed clinical phenotyping was performed at cohort enrollment, including broad-based global metabolomic profiling of plasma samples. Participants were followed prospectively for ascertainment of outcomes. Supervised and unsupervised machine learning algorithms and regression models were used to compare CTD-PAH versus IPAH metabolomic profiles and to measure metabolite-phenotype associations and interactions. Gradients across the pulmonary circulation were assessed using paired mixed venous and wedged samples in a subset of 115 participants. RESULTS Metabolomic profiles distinguished CTD-PAH from IPAH, with patients with CTD-PAH demonstrating aberrant lipid metabolism with lower circulating levels of sex steroid hormones and higher free fatty acids (FAs) and FA intermediates. Acylcholines were taken up by the right ventricular-pulmonary vascular (RV-PV) circulation, particularly in CTD-PAH, while free FAs and acylcarnitines were released. In both PAH subtypes, dysregulated lipid metabolites, among others, were associated with hemodynamic and RV measurements and with transplant-free survival. CONCLUSIONS CTD-PAH is characterized by aberrant lipid metabolism that may signal shifted metabolic substrate utilization. Abnormalities in RV-PV FA metabolism may imply a reduced capacity for mitochondrial beta oxidation within the diseased pulmonary circulation.
Collapse
Affiliation(s)
| | - Anna R. Hemnes
- Vanderbilt University Division of Allergy, Pulmonary, and Critical Care Medicine
| | - Megan Griffiths
- University of Texas Southwestern Medical Center, Division of Pediatric Cardiology
| | - Gabriele Grunig
- Divisions of Environmental and Pulmonary Medicine, Department of Medicine, NYU Grossman School of Medicine
| | - W.H. Wilson Tang
- Cleveland Clinic Department of Cardiovascular Medicine, Section of Heart Failure and Transplant Medicine
| | - Joe G. N. Garcia
- University of Arizona College of Medicine – Tucson, Department of Medicine
| | | | | | - Rachel L. Damico
- Johns Hopkins University Division of Pulmonary and Critical Care Medicine
| | - Stephen C. Mathai
- Johns Hopkins University Division of Pulmonary and Critical Care Medicine
| | - Paul M. Hassoun
- Johns Hopkins University Division of Pulmonary and Critical Care Medicine
| | | |
Collapse
|
42
|
Bordag N, Nagy BM, Zügner E, Ludwig H, Foris V, Nagaraj C, Biasin V, Bodenhofer U, Magnes C, Maron BA, Ulrich S, Lange TJ, Hötzenecker K, Pieber T, Olschewski H, Olschewski A. Lipidomics for diagnosis and prognosis of pulmonary hypertension. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.17.23289772. [PMID: 37292870 PMCID: PMC10246148 DOI: 10.1101/2023.05.17.23289772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Pulmonary hypertension (PH) poses a significant health threat with high morbidity and mortality, necessitating improved diagnostic tools for enhanced management. Current biomarkers for PH lack functionality and comprehensive diagnostic and prognostic capabilities. Therefore, there is a critical need to develop biomarkers that address these gaps in PH diagnostics and prognosis. Methods To address this need, we employed a comprehensive metabolomics analysis in 233 blood based samples coupled with machine learning analysis. For functional insights, human pulmonary arteries (PA) of idiopathic pulmonary arterial hypertension (PAH) lungs were investigated and the effect of extrinsic FFAs on human PA endothelial and smooth muscle cells was tested in vitro. Results PA of idiopathic PAH lungs showed lipid accumulation and altered expression of lipid homeostasis-related genes. In PA smooth muscle cells, extrinsic FFAs caused excessive proliferation and endothelial barrier dysfunction in PA endothelial cells, both hallmarks of PAH.In the training cohort of 74 PH patients, 30 disease controls without PH, and 65 healthy controls, diagnostic and prognostic markers were identified and subsequently validated in an independent cohort. Exploratory analysis showed a highly impacted metabolome in PH patients and machine learning confirmed a high diagnostic potential. Fully explainable specific free fatty acid (FFA)/lipid-ratios were derived, providing exceptional diagnostic accuracy with an area under the curve (AUC) of 0.89 in the training and 0.90 in the validation cohort, outperforming machine learning results. These ratios were also prognostic and complemented established clinical prognostic PAH scores (FPHR4p and COMPERA2.0), significantly increasing their hazard ratios (HR) from 2.5 and 3.4 to 4.2 and 6.1, respectively. Conclusion In conclusion, our research confirms the significance of lipidomic alterations in PH, introducing innovative diagnostic and prognostic biomarkers. These findings may have the potential to reshape PH management strategies.
Collapse
Affiliation(s)
- Natalie Bordag
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- CBmed GmbH, Center for Biomarker Research in Medicine, Graz, Austria
- BioMedTech, Graz, Austria
| | - Bence Miklos Nagy
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Elmar Zügner
- Institute for Biomedical Research and Technologies (HEALTH), Joanneum Research Forschungsgesellschaft m.b.H, Graz, Austria
| | - Helga Ludwig
- School of Informatics, Communications, and Media, University of Applied Sciences Upper Austria, Hagenberg, Austria
| | - Vasile Foris
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- BioMedTech, Graz, Austria
| | - Valentina Biasin
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Physiology, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Ulrich Bodenhofer
- School of Informatics, Communications, and Media, University of Applied Sciences Upper Austria, Hagenberg, Austria
| | - Christoph Magnes
- Institute for Biomedical Research and Technologies (HEALTH), Joanneum Research Forschungsgesellschaft m.b.H, Graz, Austria
| | - Bradley A. Maron
- University of Maryland School of Medicine, Baltimore, MD and The University of Maryland-Institute for Health Computing, Bethesda, MD, USA
| | - Silvia Ulrich
- Clinic of Pulmonology, University and University Hospital of Zurich, Zürich, Switzerland
| | - Tobias J. Lange
- Department of Internal Medicine II, Pulmonology and Critical Care, Kreisklinik Bad Reichenhall, Bad Reichenhall, Germany
- Faculty of Medicine, University of Regensburg, Regensburg, Germany
| | - Konrad Hötzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Pieber
- CBmed GmbH, Center for Biomarker Research in Medicine, Graz, Austria
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz Austria
- BioMedTech, Graz, Austria
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioMedTech, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
- BioMedTech, Graz, Austria
| |
Collapse
|
43
|
Simpson CE, Ambade AS, Harlan R, Roux A, Aja S, Graham D, Shah AA, Hummers LK, Hemnes AR, Leopold JA, Horn EM, Berman-Rosenzweig ES, Grunig G, Aldred MA, Barnard J, Comhair SAA, Tang WHW, Griffiths M, Rischard F, Frantz RP, Erzurum SC, Beck GJ, Hill NS, Mathai SC, Hassoun PM, Damico RL. Kynurenine pathway metabolism evolves with development of preclinical and scleroderma-associated pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2023; 325:L617-L627. [PMID: 37786941 PMCID: PMC11068393 DOI: 10.1152/ajplung.00177.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 10/04/2023] Open
Abstract
Understanding metabolic evolution underlying pulmonary arterial hypertension (PAH) development may clarify pathobiology and reveal disease-specific biomarkers. Patients with systemic sclerosis (SSc) are regularly surveilled for PAH, presenting an opportunity to examine metabolic change as disease develops in an at-risk cohort. We performed mass spectrometry-based metabolomics on longitudinal serum samples collected before and near SSc-PAH diagnosis, compared with time-matched SSc subjects without PAH, in a SSc surveillance cohort. We validated metabolic differences in a second cohort and determined metabolite-phenotype relationships. In parallel, we performed serial metabolomic and hemodynamic assessments as the disease developed in a preclinical model. For differentially expressed metabolites, we investigated corresponding gene expression in human and rodent PAH lungs. Kynurenine and its ratio to tryptophan (kyn/trp) increased over the surveillance period in patients with SSc who developed PAH. Higher kyn/trp measured two years before diagnostic right heart catheterization increased the odds of SSc-PAH diagnosis (OR 1.57, 95% CI 1.05-2.36, P = 0.028). The slope of kyn/trp rise during SSc surveillance predicted PAH development and mortality. In both clinical and experimental PAH, higher kynurenine pathway metabolites correlated with adverse pulmonary vascular and RV measurements. In human and rodent PAH lungs, expression of TDO2, which encodes tryptophan 2,3 dioxygenase (TDO), a protein that catalyzes tryptophan conversion to kynurenine, was significantly upregulated and tightly correlated with pulmonary hypertensive features. Upregulated kynurenine pathway metabolism occurs early in PAH, localizes to the lung, and may be modulated by TDO2. Kynurenine pathway metabolites may be candidate PAH biomarkers and TDO warrants exploration as a potential novel therapeutic target.NEW & NOTEWORTHY Our study shows an early increase in kynurenine pathway metabolism in at-risk subjects with systemic sclerosis who develop pulmonary arterial hypertension (PAH). We show that kynurenine pathway upregulation precedes clinical diagnosis and that this metabolic shift is associated with increased disease severity and shorter survival times. We also show that gene expression of TDO2, an enzyme that generates kynurenine from tryptophan, rises with PAH development.
Collapse
Affiliation(s)
- Catherine E Simpson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Anjira S Ambade
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Robert Harlan
- Johns Hopkins All Children's Molecular Determinants Core, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, United States
| | - Aurelie Roux
- Johns Hopkins All Children's Molecular Determinants Core, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, United States
| | - Susan Aja
- Johns Hopkins All Children's Molecular Determinants Core, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, United States
| | - David Graham
- Johns Hopkins All Children's Molecular Determinants Core, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, United States
| | - Ami A Shah
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Laura K Hummers
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, United States
| | - Evelyn M Horn
- Division of Cardiology, Department of Medicine, Cornell University Medical Center, New York, New York, United States
| | - Erika S Berman-Rosenzweig
- Division of Pediatric Cardiology, Columbia University Medical Center/NewYork-Presbyterian Hospital, New York, New York, United States
| | - Gabriele Grunig
- Divisions of Environmental and Pulmonary Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, New York, United States
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - John Barnard
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Suzy A A Comhair
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - W H Wilson Tang
- Division of Heart Failure and Transplant Medicine, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, United States
| | - Megan Griffiths
- Division of Pediatric Cardiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States
| | - Franz Rischard
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona College of Medicine, Tucson, Arizona, United States
| | - Robert P Frantz
- Division of Circulatory Failure, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Serpil C Erzurum
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Gerald J Beck
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States
| | - Nicholas S Hill
- Pulmonary, Critical Care and Sleep Division, Tufts University, Boston, Massachusetts, United States
| | - Stephen C Mathai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| |
Collapse
|
44
|
Wei Y, Zhao H, Kalionis B, Huai X, Hu X, Wu W, Jiang R, Gong S, Wang L, Liu J, Xia S, Yuan P, Zhao Q. The Impact of Abnormal Lipid Metabolism on the Occurrence Risk of Idiopathic Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:14280. [PMID: 37762581 PMCID: PMC10532109 DOI: 10.3390/ijms241814280] [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: 07/23/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The aim was to determine whether lipid molecules can be used as potential biomarkers for idiopathic pulmonary arterial hypertension (IPAH), providing important reference value for early diagnosis and treatment. Liquid chromatography-mass spectrometry-based lipidomic assays allow for the simultaneous detection of a large number of lipids. In this study, lipid profiling was performed on plasma samples from 69 IPAH patients and 30 healthy controls to compare the levels of lipid molecules in the 2 groups of patients, and Cox regression analysis was used to identify meaningful metrics, along with receiver operator characteristic curves to assess the ability of the lipid molecules to predict the risk of disease in patients. Among the 14 lipid subclasses tested, 12 lipid levels were significantly higher in IPAH patients than in healthy controls. Free fatty acids (FFA) and monoacylglycerol (MAG) were significantly different between IPAH patients and healthy controls. Logistic regression analysis showed that FFA (OR: 1.239, 95%CI: 1.101, 1.394, p < 0.0001) and MAG (OR: 3.711, 95%CI: 2.214, 6.221, p < 0.001) were independent predictors of IPAH development. Among the lipid subclasses, FFA and MAG have potential as biomarkers for predicting the pathogenesis of IPAH, which may improve the early diagnosis of IPAH.
Collapse
Affiliation(s)
- Yaqin Wei
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China;
| | - Hui Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women’s Hospital, Parkville 3052, Australia;
| | - Xu Huai
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xiaoyi Hu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
- Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Wenhui Wu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Sugang Gong
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Jinming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Shijin Xia
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, China;
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| | - Qinhua Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (Y.W.); (H.Z.); (X.H.); (X.H.); (W.W.); (R.J.); (S.G.); (L.W.); (J.L.)
| |
Collapse
|
45
|
Tello K, Naeije R, de Man F, Guazzi M. Pathophysiology of the right ventricle in health and disease: an update. Cardiovasc Res 2023; 119:1891-1904. [PMID: 37463510 DOI: 10.1093/cvr/cvad108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/14/2023] [Accepted: 05/02/2023] [Indexed: 07/20/2023] Open
Abstract
The contribution of the right ventricle (RV) to cardiac output is negligible in normal resting conditions when pressures in the pulmonary circulation are low. However, the RV becomes relevant in healthy subjects during exercise and definitely so in patients with increased pulmonary artery pressures both at rest and during exercise. The adaptation of RV function to loading rests basically on an increased contractility. This is assessed by RV end-systolic elastance (Ees) to match afterload assessed by arterial elastance (Ea). The system has reserve as the Ees/Ea ratio or its imaging surrogate ejection fraction has to decrease by more than half, before the RV undergoes an increase in dimensions with eventual increase in filling pressures and systemic congestion. RV-arterial uncoupling is accompanied by an increase in diastolic elastance. Measurements of RV systolic function but also of diastolic function predict outcome in any cause pulmonary hypertension and heart failure with or without preserved left ventricular ejection fraction. Pathobiological changes in the overloaded RV include a combination of myocardial fibre hypertrophy, fibrosis and capillary rarefaction, a titin phosphorylation-related displacement of myofibril tension-length relationships to higher pressures, a metabolic shift from mitochondrial free fatty acid oxidation to cytoplasmic glycolysis, toxic lipid accumulation, and activation of apoptotic and inflammatory signalling pathways. Treatment of RV failure rests on the relief of excessive loading.
Collapse
Affiliation(s)
- Khodr Tello
- Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Klinikstrasse 36, 35392 Giessen, Germany
| | - Robert Naeije
- Pathophysiology, Faculty of Medicine, Free University of Brussels, Brussels, Belgium
| | - Frances de Man
- Pulmonary Medicine, Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Marco Guazzi
- Cardiology Division, San Paolo University Hospital, University of Milano, Milano, Italy
| |
Collapse
|
46
|
Renaud D, Scholl-Bürgi S, Karall D, Michel M. Comparative Metabolomics in Single Ventricle Patients after Fontan Palliation: A Strong Case for a Targeted Metabolic Therapy. Metabolites 2023; 13:932. [PMID: 37623876 PMCID: PMC10456471 DOI: 10.3390/metabo13080932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023] Open
Abstract
Most studies on single ventricle (SV) circulation take a physiological or anatomical approach. Although there is a tight coupling between cardiac contractility and metabolism, the metabolic perspective on this patient population is very recent. Early findings point to major metabolic disturbances, with both impaired glucose and fatty acid oxidation in the cardiomyocytes. Additionally, Fontan patients have systemic metabolic derangements such as abnormal glucose metabolism and hypocholesterolemia. Our literature review compares the metabolism of patients with a SV circulation after Fontan palliation with that of patients with a healthy biventricular (BV) heart, or different subtypes of a failing BV heart, by Pubmed review of the literature on cardiac metabolism, Fontan failure, heart failure (HF), ketosis, metabolism published in English from 1939 to 2023. Early evidence demonstrates that SV circulation is not only a hemodynamic burden requiring staged palliation, but also a metabolic issue with alterations similar to what is known for HF in a BV circulation. Alterations of fatty acid and glucose oxidation were found, resulting in metabolic instability and impaired energy production. As reported for patients with BV HF, stimulating ketone oxidation may be an effective treatment strategy for HF in these patients. Few but promising clinical trials have been conducted thus far to evaluate therapeutic ketosis with HF using a variety of instruments, including ketogenic diet, ketone esters, and sodium-glucose co-transporter-2 (SGLT2) inhibitors. An initial trial on a small cohort demonstrated favorable outcomes for Fontan patients treated with SGLT2 inhibitors. Therapeutic ketosis is worth considering in the treatment of Fontan patients, as ketones positively affect not only the myocardial energy metabolism, but also the global Fontan physiopathology. Induced ketosis seems promising as a concerted therapeutic strategy.
Collapse
Affiliation(s)
- David Renaud
- Fundamental and Biomedical Sciences, Paris-Cité University, 75006 Paris, France
- Health Sciences Faculty, Universidad Europea Miguel de Cervantes, 47012 Valladolid, Spain
- Fundacja Recover, 05-124 Skrzeszew, Poland
| | - Sabine Scholl-Bürgi
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Daniela Karall
- Department of Child and Adolescent Health, Division of Pediatrics I—Inherited Metabolic Disorders, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Miriam Michel
- Department of Child and Adolescent Health, Division of Pediatrics III—Cardiology, Pulmonology, Allergology and Cystic Fibrosis, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
47
|
Kazmirczak F, Prins KW. Diabetes Enters Stage Right: Genetic Association Studies Suggest Diabetes Promotes Pulmonary Hypertension and Right Ventricular Dysfunction. J Am Heart Assoc 2023; 12:e030954. [PMID: 37522171 PMCID: PMC10492981 DOI: 10.1161/jaha.123.030954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Affiliation(s)
- Felipe Kazmirczak
- Lillehei Heart Institute, Cardiovascular Division University of Minnesota Minneapolis MN USA
| | - Kurt W Prins
- Lillehei Heart Institute, Cardiovascular Division University of Minnesota Minneapolis MN USA
| |
Collapse
|
48
|
Bagheri M, Agrawal V, Annis J, Shi M, Ferguson JF, Freiberg MS, Mosley JD, Brittain EL. Genetics of Pulmonary Pressure and Right Ventricle Stress Identify Diabetes as a Causal Risk Factor. J Am Heart Assoc 2023; 12:e029190. [PMID: 37522172 PMCID: PMC10492967 DOI: 10.1161/jaha.122.029190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/05/2023] [Indexed: 08/01/2023]
Abstract
Background Epidemiologic studies have identified risk factors associated with pulmonary hypertension and right heart failure, but causative drivers of pulmonary hypertension and right heart adaptation are not well known. We sought to leverage unbiased genetic approaches to determine clinical conditions that share genetic architecture with pulmonary pressure and right ventricular dysfunction. Methods and Results We leveraged Vanderbilt University's deidentified electronic health records and DNA biobank to identify 14 861 subjects of European ancestry who underwent at least 1 echocardiogram with available estimates of pulmonary pressure and right ventricular function. Analyses of the study were performed between 2020 and 2022. The final analytical sample included 14 861 participants (mean [SD] age, 63 [15] years and mean [SD] body mass index, 29 [7] kg/m2). An unbiased phenome-wide association study identified diabetes as the most statistically significant clinical International Classifications of Diseases, Ninth Revision (ICD-9) code associated with polygenic risk for increased pulmonary pressure. We validated this finding further by finding significant associations between genetic risk for diabetes and a related condition, obesity, with pulmonary pressure estimate. We then used 2-sample univariable Mendelian randomization and multivariable Mendelian randomization to show that diabetes, but not obesity, was independently associated with genetic risk for increased pulmonary pressure and decreased right ventricle load stress. Conclusions Our findings show that genetic risk for diabetes is the only significant independent causative driver of genetic risk for increased pulmonary pressure and decreased right ventricle load stress. These findings suggest that therapies targeting genetic risk for diabetes may also potentially be beneficial in treating pulmonary hypertension and right heart dysfunction.
Collapse
Affiliation(s)
- Minoo Bagheri
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTNUSA
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Jeffrey Annis
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Mingjian Shi
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTNUSA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTNUSA
| | - Matthew S. Freiberg
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Jonathan D. Mosley
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTNUSA
| | - Evan L. Brittain
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTNUSA
| |
Collapse
|
49
|
Feng L, Chen X, Huang Y, Zhang X, Zheng S, Xie N. Immunometabolism changes in fibrosis: from mechanisms to therapeutic strategies. Front Pharmacol 2023; 14:1243675. [PMID: 37576819 PMCID: PMC10412938 DOI: 10.3389/fphar.2023.1243675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Immune cells are essential for initiating and developing the fibrotic process by releasing cytokines and growth factors that activate fibroblasts and promote extracellular matrix deposition. Immunometabolism describes how metabolic alterations affect the function of immune cells and how inflammation and immune responses regulate systemic metabolism. The disturbed immune cell function and their interactions with other cells in the tissue microenvironment lead to the origin and advancement of fibrosis. Understanding the dysregulated metabolic alterations and interactions between fibroblasts and the immune cells is critical for providing new therapeutic targets for fibrosis. This review provides an overview of recent advances in the pathophysiology of fibrosis from the immunometabolism aspect, highlighting the altered metabolic pathways in critical immune cell populations and the impact of inflammation on fibroblast metabolism during the development of fibrosis. We also discuss how this knowledge could be leveraged to develop novel therapeutic strategies for treating fibrotic diseases.
Collapse
Affiliation(s)
- Lixiang Feng
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xingyu Chen
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yujing Huang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaodian Zhang
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Shaojiang Zheng
- Hainan Cancer Clinical Medical Center of the First Affiliated Hospital, Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province and Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
- Department of Pathology, Hainan Women and Children Medical Center, Hainan Medical University, Haikou, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
50
|
Müller M, Donhauser E, Maske T, Bischof C, Dumitrescu D, Rudolph V, Klinke A. Mitochondrial Integrity Is Critical in Right Heart Failure Development. Int J Mol Sci 2023; 24:11108. [PMID: 37446287 PMCID: PMC10342493 DOI: 10.3390/ijms241311108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/27/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Molecular processes underlying right ventricular (RV) dysfunction (RVD) and right heart failure (RHF) need to be understood to develop tailored therapies for the abatement of mortality of a growing patient population. Today, the armament to combat RHF is poor, despite the advancing identification of pathomechanistic processes. Mitochondrial dysfunction implying diminished energy yield, the enhanced release of reactive oxygen species, and inefficient substrate metabolism emerges as a potentially significant cardiomyocyte subcellular protagonist in RHF development. Dependent on the course of the disease, mitochondrial biogenesis, substrate utilization, redox balance, and oxidative phosphorylation are affected. The objective of this review is to comprehensively analyze the current knowledge on mitochondrial dysregulation in preclinical and clinical RVD and RHF and to decipher the relationship between mitochondrial processes and the functional aspects of the right ventricle (RV).
Collapse
Affiliation(s)
- Marion Müller
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Elfi Donhauser
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Tibor Maske
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Cornelius Bischof
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Daniel Dumitrescu
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Volker Rudolph
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Anna Klinke
- Agnes Wittenborg Institute for Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany; (M.M.)
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| |
Collapse
|