1
|
Wang J, Hu S, Xu Y, Wang T. Omega-6 polyunsaturated fatty acids and their metabolites: a potential targeted therapy for pulmonary hypertension. Respir Res 2025; 26:102. [PMID: 40089708 PMCID: PMC11909876 DOI: 10.1186/s12931-025-03172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Pulmonary hypertension (PH) is a progressive and life-threatening cardiopulmonary disease that is not uncommon. The modulation of the pulmonary artery (PA) involves various fatty acids, including omega-6 polyunsaturated fatty acids (ω-6 PUFAs) and ω-6 PUFAs-derived oxylipins. These lipid mediators are produced through cyclooxygenase (COX), lipoxygenase (LOX), cytochrome P450 (CYP450), and non-enzymatic pathways. They play a crucial role in the occurrence and development of PH by regulating the function and phenotype of pulmonary artery endothelial cells (PAECs), pulmonary artery smooth muscle cells (PASMCs), pulmonary fibroblasts, alveolar macrophages, and inflammatory cells. The alterations in ω-6 PUFAs and oxylipins are pivotal in causing vasoconstriction, pulmonary remodeling, and ultimately leading to right heart failure in PH. Despite the limited understanding of the PH pathophysiology, there is potential for novel interventions through dietary and pharmacological approaches targeting ω-6 PUFAs and oxylipins. The aim of this review is to summarize the significant advances in clinical and basic research on omega-6 PUFAs and oxylipins in pulmonary vascular disease, particularly PH, and to propose a potential targeted therapeutic modality against omega-6 PUFAs.
Collapse
Affiliation(s)
- Jiayao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shunlian Hu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yahan Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
- The Center for Biomedical Research, Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, National Health Committee (NHC), Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
2
|
Farha S, Asosingh K, Hassoun PM, Barnard J, Comhair S, Reichard A, Wanner N, Radeva M, Aldred MA, Beck GJ, Berman-Rosenzweig E, Borlaug BA, Finet JE, Frantz RP, Grunig G, Hemnes AR, Hill N, Horn EM, Jellis C, Leopold JA, Mehra R, Park MM, Rischard FP, Tang WHW, Erzurum SC. Alterations in Mitochondrial Function in Pulmonary Vascular Diseases. Antioxid Redox Signal 2025; 42:361-377. [PMID: 39655485 DOI: 10.1089/ars.2024.0557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Aims: Alterations of mitochondrial bioenergetics and arginine metabolism are universally present and mechanistically linked to pulmonary arterial hypertension (PAH), but there is little knowledge of arginine metabolism and mitochondrial functions across the different pulmonary hypertension (PH) groups. We hypothesize that abnormalities in mitochondrial functions are present across all PH groups and associated with clinical phenotypes. We test the hypothesis in PH patients and healthy controls from the Pulmonary Vascular Disease Phenomics Program cohort, who had comprehensive clinical phenotyping and follow-up for at least 4 years for death or transplant status. Mitochondrial transmembrane potential, superoxide production, and mass were measured by flow cytometry in fresh platelets. Metabolomics analysis was performed on plasma samples. Global arginine bioavailability was calculated as the ratio of arginine/(ornithine+citrulline). Results: Global arginine bioavailability is consistently lower than controls in all PH groups. Although the mitochondrial mass is similar across all PH groups and controls, superoxide production and transmembrane potential vary across groups. Mitochondrial superoxide is higher in group 1 PAH and lowest in group 3 compared with other groups, while transmembrane potential is lower in group 1 PAH than controls or group 3. The alterations in mitochondrial functions of group 1 PAH are associated with changes in fatty acid metabolism. Mitochondrial transmembrane potential in group 1 PAH is associated with transplant-free survival. Conclusion: While alterations in mitochondrial function are found in all PH groups, group 1 PAH has a unique mitochondrial phenotype with greater superoxide and lower transmembrane potential linked to fatty acid metabolism, and clinically to survival. Antioxid. Redox Signal. 42, 361-377.
Collapse
Affiliation(s)
- Samar Farha
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Ohio, USA
| | - Kewal Asosingh
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - John Barnard
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Suzy Comhair
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Andrew Reichard
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nicholas Wanner
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Milena Radeva
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Micheala A Aldred
- Department of Medicine, Indiana University School of Medicine Indianapolis, Indianapolis, Indiana, USA
| | - Gerald J Beck
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - J Emanuel Finet
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert P Frantz
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Gabriele Grunig
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nicholas Hill
- Division of Pulmonary, Critical Care, and Sleep Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Evelyn M Horn
- Division of Cardiology, Weill Cornell Medical Center, New York, New York, USA
| | - Christine Jellis
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reena Mehra
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, Washington, USA
| | - Margaret M Park
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Franz P Rischard
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Arizona, Tucson, Arizona, USA
| | - W H Wilson Tang
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Heart Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil C Erzurum
- Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Lerner Research Institute, Cleveland Clinic, Ohio, USA
| |
Collapse
|
3
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Arboleda-Mojica SL, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Interplay of Transcriptomic Regulation, Microbiota, and Signaling Pathways in Lung and Gut Inflammation-Induced Tumorigenesis. Cells 2024; 14:1. [PMID: 39791702 PMCID: PMC11720097 DOI: 10.3390/cells14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn's disease, and twelve ulcerative colitis high throughput sequencing datasets using R language specialized libraries and gene enrichment analyses identified a regulatory network in each inflammatory disease. IRF9 and LINC01089 in pulmonary arterial hypertension are related to the regulation of signaling pathways like MAPK, NOTCH, human papillomavirus, and hepatitis c infection. ZNF91 and TP53TG1 in Crohn's disease are related to the regulation of PPAR, MAPK, and metabolic signaling pathways. ZNF91, VDR, DLEU1, SATB2-AS1, and TP53TG1 in ulcerative colitis are related to the regulation of PPAR, AMPK, and metabolic signaling pathways. The activation of the transcriptomic network and signaling pathways might be related to the interaction of the characteristic microbiota of the inflammatory disease, with the lung and gut cell receptors present in membrane rafts and complexes. The transcriptomic analysis highlights the impact of several coding and non-coding RNAs, suggesting their relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during lung and gut cell adaptation to inflammatory phenotypes.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Natalia Belén Pedroza-Aconcha
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
4
|
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
|
5
|
Sumer C, Okumus G, Isik EG, Turkmen C, Bilge AK, Inanc M. (18)F-fluorodeoxyglucose uptake by positron emission tomography in patients with IPAH and CTEPH. Pulm Circ 2024; 14:e12363. [PMID: 38618292 PMCID: PMC11009453 DOI: 10.1002/pul2.12363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 02/26/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is driven by pathologies associated with increased metabolism such as pulmonary revascularization, vasoconstriction and smooth muscle cell proliferation in pulmonary artery wall. 18-fluorodeoxyglucose positron emission tomography (18FDG-PET) is an imaging technique sensitive to glucose metabolism and might be considered as a non-invasive method for diagnosis due to significant role of inflammation in idiopathic pulmonary artery hypertension (IPAH) and chronic thromboembolic pulmonary hypertension (CTEPH). The present study aimed to investigate the role of PET/CT imaging of patients with IPAH and CTEPH as an alternative diagnosis method. Demographic characteristics, FDG uptake in lungs, pulmonary artery and right ventricle (RV) of 17 patients (10 IPAH, 7 CTEPH), and 30 controls were evaluated. PET scanning, 6-min walk test, pro-BNP level, right heart catheterization of patients were performed both at the onsert and after 6-month PAH specific treatment. IPAH and CTEPH patients had significantly higher left lung FDG (p = 0.006), right lung FDG (p = 0.004), right atrial (RA) FDG (p < 0.001) and RV FDG (p < 0.001) uptakes than controls. Positive correlation was detected between the RV FDG uptake and the mean pulmonary artery pressure (mPAP) (r = 0.7, p = 0.012) and between the RA FDG uptake and the right atrial pressure (RAP) (r = 0.5, p = 0.02). Increased RV FDG and RA FDG uptakes predicts the presence of pulmonary hypertension and correlates with mPAP and RAP, respectively, which are important indicators in the prognosis of PAH. Further studies are required whether FDG PET imaging can be used to diagnose or predict the prognosis of pulmonary hypertension.
Collapse
Affiliation(s)
- Celik Sumer
- Department of Pulmonary DiseasesIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| | - Gulfer Okumus
- Department of Pulmonary DiseasesIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| | - Emine Goknur Isik
- Department of Nuclear MedicineIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| | - Cuneyt Turkmen
- Department of Nuclear MedicineIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| | - Ahmet Kaya Bilge
- Department of CardiologyIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| | - Murat Inanc
- Department of RheumatologyIstanbul University Istanbul Faculty of MedicineIstanbulTurkey
| |
Collapse
|
6
|
Franzoni L, Oliveira RCD, Busin D, Turella DJP, Costa RR, Saffi MAL, Silveira ADD, Stein R. Non-Invasive Assessment of Cardiodynamics by Impedance Cardiography during the Six-Minute Walk Test in Patients with Heart Failure. Arq Bras Cardiol 2023; 120:e20230087. [PMID: 38232243 DOI: 10.36660/abc.20230087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/21/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Central Illustration: Non-Invasive Assessment of Cardiodynamics by Impedance Cardiography during the Six-Minute Walk Test in Patients with Heart Failure. The six-minute walk test (6MWT) is commonly used to evaluate heart failure (HF) patients. However, several clinical factors can influence the distance walked in the test. Signal-morphology impedance cardiography (SM-ICG) is a useful tool to noninvasively assess hemodynamics. OBJECTIVE This study aimed to compare cardiac output (CO), heart rate (HR), and stroke volume (SV) acceleration and deceleration responses to 6MWT in individuals with HF and reduced ejection fraction (HFrEF) and healthy controls. METHODS This is a cross-sectional observational study. CO, HR, SV and cardiac index (CI) were evaluated before, during, and after the 6MWT assessed by SM-ICG. The level of significance adopted in the statistical analysis was 5%. RESULTS Twenty-seven participants were included (13 HFrEF and 14 healthy controls). CO and HR acceleration significantly differed between groups (p<0.01; p=0.039, respectively). We found significant differences in SV, CO and CI between groups (p<0.01). Linear regression showed an impaired SV contribution to CO change in HFrEF group (22.9% versus 57.4%). CONCLUSION The main finding of the study was that individuals with HFrEF showed lower CO and HR acceleration values during the submaximal exercise test compared to healthy controls. This may indicate an imbalance in the autonomic response to exercise in this condition.
Collapse
Affiliation(s)
- Leandro Franzoni
- Programa de Pós-Graduação em Ciências da Saúde: Cardiologia e Ciências Cardiovasculares ( UFRGS ), Porto Alegre , RS - Brasil
| | - Rafael Cechet de Oliveira
- Programa de Pós-Graduação em Ciências da Saúde: Cardiologia e Ciências Cardiovasculares ( UFRGS ), Porto Alegre , RS - Brasil
| | - Diego Busin
- Universidade de Caxias do Sul , Caxias do Sul , RS - Brasil
| | | | - Rochelle Rocha Costa
- Universidade de Brasília - Programa de Pós-Graduação em Educação Física , Porto Alegre , RS - Brasil
| | | | | | - Ricardo Stein
- Programa de Pós-Graduação em Ciências da Saúde: Cardiologia e Ciências Cardiovasculares ( UFRGS ), Porto Alegre , RS - Brasil
| |
Collapse
|
7
|
Otálora-Otálora BA, López-Rivera JJ, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Host Transcriptional Regulatory Genes and Microbiome Networks Crosstalk through Immune Receptors Establishing Normal and Tumor Multiomics Metafirm of the Oral-Gut-Lung Axis. Int J Mol Sci 2023; 24:16638. [PMID: 38068961 PMCID: PMC10706695 DOI: 10.3390/ijms242316638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
The microbiome has shown a correlation with the diet and lifestyle of each population in health and disease, the ability to communicate at the cellular level with the host through innate and adaptative immune receptors, and therefore an important role in modulating inflammatory process related to the establishment and progression of cancer. The oral cavity is one of the most important interaction windows between the human body and the environment, allowing the entry of an important number of microorganisms and their passage across the gastrointestinal tract and lungs. In this review, the contribution of the microbiome network to the establishment of systemic diseases like cancer is analyzed through their synergistic interactions and bidirectional crosstalk in the oral-gut-lung axis as well as its communication with the host cells. Moreover, the impact of the characteristic microbiota of each population in the formation of the multiomics molecular metafirm of the oral-gut-lung axis is also analyzed through state-of-the-art sequencing techniques, which allow a global study of the molecular processes involved of the flow of the microbiota environmental signals through cancer-related cells and its relationship with the establishment of the transcription factor network responsible for the control of regulatory processes involved with tumorigenesis.
Collapse
Affiliation(s)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
8
|
Pokharel MD, Marciano DP, Fu P, Franco MC, Unwalla H, Tieu K, Fineman JR, Wang T, Black SM. Metabolic reprogramming, oxidative stress, and pulmonary hypertension. Redox Biol 2023; 64:102797. [PMID: 37392518 PMCID: PMC10363484 DOI: 10.1016/j.redox.2023.102797] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/03/2023] Open
Abstract
Mitochondria are highly dynamic organelles essential for cell metabolism, growth, and function. It is becoming increasingly clear that endothelial cell dysfunction significantly contributes to the pathogenesis and vascular remodeling of various lung diseases, including pulmonary arterial hypertension (PAH), and that mitochondria are at the center of this dysfunction. The more we uncover the role mitochondria play in pulmonary vascular disease, the more apparent it becomes that multiple pathways are involved. To achieve effective treatments, we must understand how these pathways are dysregulated to be able to intervene therapeutically. We know that nitric oxide signaling, glucose metabolism, fatty acid oxidation, and the TCA cycle are abnormal in PAH, along with alterations in the mitochondrial membrane potential, proliferation, and apoptosis. However, these pathways are incompletely characterized in PAH, especially in endothelial cells, highlighting the urgent need for further research. This review summarizes what is currently known about how mitochondrial metabolism facilitates a metabolic shift in endothelial cells that induces vascular remodeling during PAH.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - David P Marciano
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Maria Clara Franco
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-Medicine, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, The University of California San Francisco, San Francisco, CA, 94143, USA; Cardiovascular Research Institute, The University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
9
|
Cao J, Wei X, Liu MF, An GS, Li J, Du QX, Sun JH. Forensic identification of sudden cardiac death: a new approach combining metabolomics and machine learning. Anal Bioanal Chem 2023; 415:2291-2305. [PMID: 36933055 DOI: 10.1007/s00216-023-04651-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023]
Abstract
The determination of sudden cardiac death (SCD) is one of the difficult tasks in the forensic practice, especially in the absence of specific morphological changes in the autopsies and histological investigations. In this study, we combined the metabolic characteristics from corpse specimens of cardiac blood and cardiac muscle to predict SCD. Firstly, ultra-high performance liquid chromatography coupled with high-resolution mass spectrometry (UPLC-HRMS)-based untargeted metabolomics was applied to obtain the metabolomic profiles of the specimens, and 18 and 16 differential metabolites were identified in the cardiac blood and cardiac muscle from the corpses of those who died of SCD, respectively. Several possible metabolic pathways were proposed to explain these metabolic alterations, including the metabolism of energy, amino acids, and lipids. Then, we validated the capability of these combinations of differential metabolites to distinguish between SCD and non-SCD through multiple machine learning algorithms. The results showed that stacking model integrated differential metabolites featured from the specimens showed the best performance with 92.31% accuracy, 93.08% precision, 92.31% recall, 91.96% F1 score, and 0.92 AUC. Our results revealed that the SCD metabolic signature identified by metabolomics and ensemble learning in cardiac blood and cardiac muscle has potential in SCD post-mortem diagnosis and metabolic mechanism investigations.
Collapse
Affiliation(s)
- Jie Cao
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Xue Wei
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Ming-Feng Liu
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Guo-Shuai An
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Jian Li
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Qiu-Xiang Du
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China
| | - Jun-Hong Sun
- School of Forensic Medicine, Shanxi Medical University, Yuci District, Jinzhong, Shanxi, 030604, People's Republic of China.
| |
Collapse
|
10
|
Tofovic SP. Purine Nucleoside Phosphorylase: A New Pharmacological Target in Sickle Cell Disease and Hemolytic Vasculopathy. Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
11
|
Gu S, Goel K, Forbes LM, Kheyfets VO, Yu YRA, Tuder RM, Stenmark KR. Tensions in Taxonomies: Current Understanding and Future Directions in the Pathobiologic Basis and Treatment of Group 1 and Group 3 Pulmonary Hypertension. Compr Physiol 2023; 13:4295-4319. [PMID: 36715285 PMCID: PMC10392122 DOI: 10.1002/cphy.c220010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In the over 100 years since the recognition of pulmonary hypertension (PH), immense progress and significant achievements have been made with regard to understanding the pathophysiology of the disease and its treatment. These advances have been mostly in idiopathic pulmonary arterial hypertension (IPAH), which was classified as Group 1 Pulmonary Hypertension (PH) at the Second World Symposia on PH in 1998. However, the pathobiology of PH due to chronic lung disease, classified as Group 3 PH, remains poorly understood and its treatments thus remain limited. We review the history of the classification of the five groups of PH and aim to provide a state-of-the-art review of the understanding of the pathogenesis of Group 1 PH and Group 3 PH including insights gained from novel high-throughput omics technologies that have revealed heterogeneities within these categories as well as similarities between them. Leveraging the substantial gains made in understanding the genomics, epigenomics, proteomics, and metabolomics of PAH to understand the full spectrum of the complex, heterogeneous disease of PH is needed. Multimodal omics data as well as supervised and unbiased machine learning approaches after careful consideration of the powerful advantages as well as of the limitations and pitfalls of these technologies could lead to earlier diagnosis, more precise risk stratification, better predictions of disease response, new sub-phenotype groupings within types of PH, and identification of shared pathways between PAH and other types of PH that could lead to new treatment targets. © 2023 American Physiological Society. Compr Physiol 13:4295-4319, 2023.
Collapse
Affiliation(s)
- Sue Gu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Khushboo Goel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- National Jewish Health, Denver, Colorodo, USA
| | - Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Vitaly O. Kheyfets
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Yen-rei A. Yu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
| | - Rubin M. Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Lab, University of Colorado School of Medicine, Colorado, USA
- Department of Pediatrics Section of Critical Care Medicine, University of Colorado Anschutz Medical Campus, Colorado, USA
| |
Collapse
|
12
|
Abstract
Pulmonary arterial hypertension forms the first and most severe of the 5 categories of pulmonary hypertension. Disease pathogenesis is driven by progressive remodeling of peripheral pulmonary arteries, caused by the excessive proliferation of vascular wall cells, including endothelial cells, smooth muscle cells and fibroblasts, and perivascular inflammation. Compelling evidence from animal models suggests endothelial cell dysfunction is a key initial trigger of pulmonary vascular remodeling, which is characterised by hyperproliferation and early apoptosis followed by enrichment of apoptosis-resistant populations. Dysfunctional pulmonary arterial endothelial cells lose their ability to produce vasodilatory mediators, together leading to augmented pulmonary arterial smooth muscle cell responses, increased pulmonary vascular pressures and right ventricular afterload, and progressive right ventricular hypertrophy and heart failure. It is recognized that a range of abnormal cellular molecular signatures underpin the pathophysiology of pulmonary arterial hypertension and are enhanced by loss-of-function mutations in the BMPR2 gene, the most common genetic cause of pulmonary arterial hypertension and associated with worse disease prognosis. Widespread metabolic abnormalities are observed in the heart, pulmonary vasculature, and systemic tissues, and may underpin heterogeneity in responsivity to treatment. Metabolic abnormalities include hyperglycolytic reprogramming, mitochondrial dysfunction, aberrant polyamine and sphingosine metabolism, reduced insulin sensitivity, and defective iron handling. This review critically discusses published mechanisms linking metabolic abnormalities with dysfunctional BMPR2 (bone morphogenetic protein receptor 2) signaling; hypothesized mechanistic links requiring further validation; and their relevance to pulmonary arterial hypertension pathogenesis and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Iona Cuthbertson
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| | - Paola Caruso
- Department of Medicine, University of Cambridge School of Clinical Medicine, Heart and Lung Research Institute, United Kingdom
| |
Collapse
|
13
|
Swisher JW, Weaver E. The Evolving Management and Treatment Options for Patients with Pulmonary Hypertension: Current Evidence and Challenges. Vasc Health Risk Manag 2023; 19:103-126. [PMID: 36895278 PMCID: PMC9990521 DOI: 10.2147/vhrm.s321025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/01/2023] [Indexed: 03/06/2023] Open
Abstract
Pulmonary hypertension may develop as a disease process specific to pulmonary arteries with no identifiable cause or may occur in relation to other cardiopulmonary and systemic illnesses. The World Health Organization (WHO) classifies pulmonary hypertensive diseases on the basis of primary mechanisms causing increased pulmonary vascular resistance. Effective management of pulmonary hypertension begins with accurately diagnosing and classifying the disease in order to determine appropriate treatment. Pulmonary arterial hypertension (PAH) is a particularly challenging form of pulmonary hypertension as it involves a progressive, hyperproliferative arterial process that leads to right heart failure and death if untreated. Over the last two decades, our understanding of the pathobiology and genetics behind PAH has evolved and led to the development of several targeted disease modifiers that ameliorate hemodynamics and quality of life. Effective risk management strategies and more aggressive treatment protocols have also allowed better outcomes for patients with PAH. For those patients who experience progressive PAH with medical therapy, lung transplantation remains a life-saving option. More recent work has been directed at developing effective treatment strategies for other forms of pulmonary hypertension, such as chronic thromboembolic pulmonary hypertension (CTEPH) and pulmonary hypertension due to other lung or heart diseases. The discovery of new disease pathways and modifiers affecting the pulmonary circulation is an ongoing area of intense investigation.
Collapse
Affiliation(s)
- John W Swisher
- East Tennessee Pulmonary Hypertension Center, StatCare Pulmonary Consultants, Knoxville, TN, USA
| | - Eric Weaver
- East Tennessee Pulmonary Hypertension Center, StatCare Pulmonary Consultants, Knoxville, TN, USA
| |
Collapse
|
14
|
Alsabeelah N, Kumar V. Protective Effect of Triclosan in Monocrotaline-Induced Pulmonary Arterial Hypertension: FASN Inhibition a Novel Approach. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2022; 14:171-177. [PMID: 37051426 PMCID: PMC10084994 DOI: 10.4103/jpbs.jpbs_307_22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/10/2022] [Accepted: 09/27/2022] [Indexed: 02/17/2023] Open
Abstract
Background Novel pharmacological approaches are needed to improve the outcomes of patients with idiopathic pulmonary hypertension. Fatty acid synthase (FASN) inhibitors have shown beneficial effects in preclinical models of pulmonary arterial hypertension (PAH), because of their role in the regulation of pulmonary artery vasoconstrictor tone and remodeling. Objective We compared a Triclosan (FASN inhibitor), for the first time with the dual endothelin receptor antagonist, macitentan, in a monocrotaline-induced rat pulmonary hypertension model. Methods Different methods (hemodynamics, histology of right ventricle and pulmonary vessels, and circulating biomarkers) showed consistently that 30 mg/kg daily of Triclosan (FASN inhibitor) and 10 mg/kg daily of macitentan slowed the progression of PAH both at the functional and structural levels. Results Treatments started on day 14 after monocrotaline injection and lasted 14 days. The findings of all experimental methods show that the FASN inhibitor has more similar effects as compared to macitentan. Conclusion Our study reveals that inhibition of FAS decreases RV hypertrophy and improves cardiac function associated with PAH with the regulation of metabolic functions and governs further studies to establish "FASN inhibitor as a potential therapeutic approach" for the management of PAH.
Collapse
Affiliation(s)
- Nimer Alsabeelah
- Pharmacy Practice Department, Pharmacy College, University of Hafr Al Batin, Saudi Arabia
| | - Vinay Kumar
- Department of Pharmacology, KIET Group of Institutions (KIET School of Pharmacy), Delhi-NCR, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
15
|
Lee MH, Menezes TCF, Reisz JA, Ferreira EVM, Graham BB, Oliveira RKF. Exercise metabolomics in pulmonary arterial hypertension: Where pulmonary vascular metabolism meets exercise physiology. Front Physiol 2022; 13:963881. [PMID: 36171971 PMCID: PMC9510894 DOI: 10.3389/fphys.2022.963881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/23/2022] [Indexed: 01/29/2023] Open
Abstract
Pulmonary arterial hypertension is an incurable disease marked by dysregulated metabolism, both at the cellular level in the pulmonary vasculature, and at the whole-body level characterized by impaired exercise oxygen consumption. Though both altered pulmonary vascular metabolism and abnormal exercise physiology are key markers of disease severity and pulmonary arterial remodeling, their precise interactions are relatively unknown. Herein we review normal pulmonary vascular physiology and the current understanding of pulmonary vascular cell metabolism and cardiopulmonary response to exercise in Pulmonary arterial hypertension. We additionally introduce a newly developed international collaborative effort aimed at quantifying exercise-induced changes in pulmonary vascular metabolism, which will inform about underlying pathophysiology and clinical management. We support our investigative approach by presenting preliminary data and discuss potential future applications of our research platform.
Collapse
Affiliation(s)
- Michael H. Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Thaís C. F. Menezes
- Division of Respiratory Diseases, Department of Medicine, Federal University of SP, São Paulo, Brazil
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eloara V. M. Ferreira
- Division of Respiratory Diseases, Department of Medicine, Federal University of SP, São Paulo, Brazil
| | - Brian B. Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Rudolf K. F. Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of SP, São Paulo, Brazil,*Correspondence: Rudolf K. F. Oliveira,
| |
Collapse
|
16
|
Lee MH, Sanders L, Kumar R, Hernandez-Saavedra D, Yun X, Ford JA, Perez MJ, Mickael C, Gandjeva A, Koyanagi DE, Harral JW, Irwin DC, Kassa B, Eckel RH, Shimoda LA, Graham BB, Tuder RM. Contribution of fatty acid oxidation to the pathogenesis of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2022; 323:L355-L371. [PMID: 35763400 PMCID: PMC9448289 DOI: 10.1152/ajplung.00039.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/05/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022] Open
Abstract
Dysregulated metabolism characterizes both animal and human forms of pulmonary hypertension (PH). Enzymes involved in fatty acid metabolism have previously not been assessed in human pulmonary arteries affected by pulmonary arterial hypertension (PAH), and how inhibition of fatty acid oxidation (FAO) may attenuate PH remains unclear. Fatty acid metabolism gene transcription was quantified in laser-dissected pulmonary arteries from 10 explanted lungs with advanced PAH (5 idiopathic, 5 associated with systemic sclerosis), and 5 donors without lung diseases. Effects of oxfenicine, a FAO inhibitor, on female Sugen 5416-chronic hypoxia (SuHx) rats were studied in vivo using right heart catheterization, and ex vivo using perfused lungs and pulmonary artery ring segments. The impact of pharmacologic (oxfenicine) and genetic (carnitine palmitoyltransferase 1a heterozygosity) FAO suppression was additionally probed in mouse models of Schistosoma and hypoxia-induced PH. Potential mechanisms underlying FAO-induced PH pathogenesis were examined by quantifying ATP and mitochondrial mass in oxfenicine-treated SuHx pulmonary arterial cells, and by assessing pulmonary arterial macrophage infiltration with immunohistochemistry. We found upregulated pulmonary arterial transcription of 26 and 13 FAO genes in idiopathic and systemic sclerosis-associated PAH, respectively. In addition to promoting de-remodeling of pulmonary arteries in SuHx rats, oxfenicine attenuated endothelin-1-induced vasoconstriction. FAO inhibition also conferred modest benefit in the two mouse models of PH. Oxfenicine increased mitochondrial mass in cultured rat pulmonary arterial cells, and decreased the density of perivascular macrophage infiltration in pulmonary arteries of treated SuHx rats. In summary, FAO inhibition attenuated experimental PH, and may be beneficial in human PAH.
Collapse
Affiliation(s)
- Michael H Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Linda Sanders
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Rahul Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Daniel Hernandez-Saavedra
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Joshay A Ford
- University of Colorado School of Medicine, Aurora, Colorado
| | - Mario J Perez
- Department of Psychiatry, University of Colorado, Aurora, Colorado
| | - Claudia Mickael
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Aneta Gandjeva
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Daniel E Koyanagi
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Julie W Harral
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - David C Irwin
- Cardiovascular Pulmonary Research Laboratory, Department of Pediatrics and Medicine, University of Colorado, Aurora, Colorado
| | - Biruk Kassa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Robert H Eckel
- Division of Endocrinology, Department of Medicine, University of Colorado, Aurora, Colorado
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brian B Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Rubin M Tuder
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
17
|
Liu X, Zhang L, Zhang W. Metabolic reprogramming: A novel metabolic model for pulmonary hypertension. Front Cardiovasc Med 2022; 9:957524. [PMID: 36093148 PMCID: PMC9458918 DOI: 10.3389/fcvm.2022.957524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension, or PAH, is a condition that is characterized by pulmonary artery pressures above 20 mmHg (at rest). In the treatment of PAH, the pulmonary vascular system is regulated to ensure a diastolic and contraction balance; nevertheless, this treatment does not prevent or reverse pulmonary vascular remodeling and still causes pulmonary hypertension to progress. According to Warburg, the link between metabolism and proliferation in PAH is similar to that of cancer, with a common aerobic glycolytic phenotype. By activating HIF, aerobic glycolysis is enhanced and cell proliferation is triggered. Aside from glutamine metabolism, the Randle cycle is also present in PAH. Enhanced glutamine metabolism replenishes carbon intermediates used by glycolysis and provides energy to over-proliferating and anti-apoptotic pulmonary vascular cells. By activating the Randle cycle, aerobic oxidation is enhanced, ATP is increased, and myocardial injury is reduced. PAH is predisposed by epigenetic dysregulation of DNA methylation, histone acetylation, and microRNA. This article discusses the abnormal metabolism of PAH and how metabolic therapy can be used to combat remodeling.
Collapse
|
18
|
Jiang L, Goncharov DA, Shen Y, Lin D, Chang B, Pena A, DeLisser H, Goncharova EA, Kudryashova TV. Akt-Dependent Glycolysis-Driven Lipogenesis Supports Proliferation and Survival of Human Pulmonary Arterial Smooth Muscle Cells in Pulmonary Hypertension. Front Med (Lausanne) 2022; 9:886868. [PMID: 35836951 PMCID: PMC9274086 DOI: 10.3389/fmed.2022.886868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Hyper-proliferation of pulmonary arterial vascular smooth muscle cells (PAVSMC) is an important pathological component of pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). Lipogenesis is linked to numerous proliferative diseases, but its role in PAVSMC proliferation in PAH remains to be elucidated. We found that early-passage human PAH PAVSMC had significant up-regulation of key fatty acids synthesis enzymes ATP-citrate lyase (ACLY), acetyl-CoA carboxylase (ACC), and fatty acid synthase (FASN), and increased unstimulated proliferation compared to control human PAVSMC. Treatment with an allosteric ACC inhibitor 5-tetradecyloxy-2-furoic acid (TOFA) significantly decreased proliferation and induced apoptosis of human PAH PAVSMC. Intracellular lipid content and proliferation of PAH PAVSMC were not reduced by incubation in lipid-depleted media but suppressed by a non-metabolizable analog of glucose 2-Deoxy-D-glucose (2-DG) and partially restored by addition of pyruvate. Protein kinase Akt was upregulated in human PAH PAVSMC in a sirtuin 7 (SIRT7)- and c-Jun N-terminal kinase (JNK)-dependent manner. Pharmacological inhibition of Akt down-regulated ACLY and ACC, significantly reduced intracellular lipid content, inhibited proliferation and induced apoptosis of human PAH PAVSMC. Taken together, these data demonstrate that human PAH PAVSMC have up-regulated lipogenesis, which is supported in an Akt- and glycolysis-dependent manner and is required for increased proliferation and survival. Our data suggest that there is a mechanistic link between glycolysis, lipogenesis, and the proliferation of human PAH PAVSMC and call for further studies to determine the potential attractiveness of a SIRT7/JNK-Akt-lipogenesis axis as a target pathway to inhibit PAVSMC hyper-proliferation in PAH.
Collapse
Affiliation(s)
- Lifeng Jiang
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Dmitry A Goncharov
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Yuanjun Shen
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Derek Lin
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Baojun Chang
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andressa Pena
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Horace DeLisser
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Elena A Goncharova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Tatiana V Kudryashova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
19
|
Abstract
Pulmonary hypertension (PH) because of chronic lung disease is categorized as Group 3 PH in the most recent classification system. Prevalence of these diseases is increasing over time, creating a growing need for effective therapeutic options. Recent approval of the first pulmonary arterial hypertension therapy for the treatment of Group 3 PH related to interstitial lung disease represents an encouraging advancement. This review focuses on molecular mechanisms contributing to pulmonary vasculopathy in chronic hypoxia, the pathology and epidemiology of Group 3 PH, the right ventricular dysfunction observed in this population and clinical trial data that inform the use of pulmonary vasodilators in Group 3 PH.
Collapse
Affiliation(s)
- Navneet Singh
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI
| | - Peter Dorfmüller
- Department of Pathology, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Germany (P.D.).,German Center for Lung Research (DZL), Giessen, Germany (P.D.)
| | - Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA (O.A.S.)
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI.,Department of Health Services, Policy and Practice (C.E.V.), Brown University, Providence, RI
| |
Collapse
|
20
|
Zhao Q, Hou C, Lu Y, Qiu Q, Xie L, Xu S, Xiao T, Zhong M. Characterization and protective effects of a novel Bosentan nano-particle. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Liu D, Qin S, Su D, Wang K, Huang Y, Huang Y, Pang Y. Metabolic Reprogramming of the Right Ventricle and Pulmonary Arteries in a Flow-Associated Pulmonary Arterial Hypertension Rat Model. ACS OMEGA 2022; 7:1273-1287. [PMID: 35036789 PMCID: PMC8757344 DOI: 10.1021/acsomega.1c05895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a complex devastating disease relevant to remarkable metabolic dysregulation. Although various research studies on PAH from a metabolic perspective have been emerging, pathogenesis of PAH varies in different categories. Research on metabolic reprogramming in flow-associated PAH remains insufficient. An untargeted metabolomic profiling platform was used to evaluate the metabolic profile of pulmonary arteries (PAs) as well as the right ventricle (RV) in a flow-associated PAH rat model in the present work. A total of 79 PAs and 128 RV metabolites were significantly altered in PAH rats, among which 39 metabolites were assessed as shared dysregulated metabolites in PAs and the RV. Pathway analysis elucidated that, in PAs of PAH rats, pathways of phenylalanine, tyrosine, and tryptophan biosynthesis and linoleic acid metabolism were significantly altered, while in the RV, arginine biosynthesis and linoleic acid metabolism were altered dramatically. Further integrated analysis of shared dysregulated PA and RV metabolites demonstrated that the linoleic acid metabolism and the arachidonic acid (AA) metabolism were the key pathways involved in the pathogenesis of flow-associated PAH. Results obtained from the present work indicate that the PAH pathogenesis could be mediated by widespread metabolic reprogramming. In particular, the dysregulation of AA metabolism may considerably contribute to the development of high blood flow-associated PAH.
Collapse
Affiliation(s)
- Dongli Liu
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| | - Suyuan Qin
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| | - Danyan Su
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| | - Kai Wang
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
- Department
of Pediatrics, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325015, China
| | - Yanyun Huang
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| | - Yuqin Huang
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| | - Yusheng Pang
- Department
of Pediatrics, The First Affiliated Hospital
of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
22
|
Fujita T, Zysman M, Elgrabli D, Murayama T, Haruta M, Lanone S, Ishida T, Boczkowski J. Anti-inflammatory effect of gold nanoparticles supported on metal oxides. Sci Rep 2021; 11:23129. [PMID: 34848769 PMCID: PMC8632916 DOI: 10.1038/s41598-021-02419-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/02/2021] [Indexed: 11/09/2022] Open
Abstract
Gold (Au) can be deposited as nanoparticles (NPs) smaller than 10 nm in diameter on a variety of metal oxide (MOx) NPs. Au/MOx have high catalytic performance and selective oxidation capacity which could have implications in terms of biological activity, and more specifically in modulation of the inflammatory reaction. Therefore, the aim of this study was to examine the effect of Au/TiO2, Au/ZrO2 and Au/CeO2 on viability, phagocytic capacity and inflammatory profile (TNF-α and IL-1β secretion) of murine macrophages. The most important result of this study is an anti-inflammatory effect of Au/MOx depending on the MOx nature with particle internalization and no alteration of cell viability and phagocytosis. The effect was dependent on the MOx NPs chemical nature (Au/TiO2 > Au/ZrO2 > Au/CeO2 if we consider the number of cytokines whose concentration was reduced by the NPs), and on the inflammatory mediator considered. The effect of Au/TiO2 NPs was not related to Au NPs size (at least in the case of Au/TiO2 NPs in the range of 3-8 nm). To the best of our knowledge, this is the first demonstration of an anti-inflammatory effect of Au/MOx.
Collapse
Affiliation(s)
- Takashi Fujita
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1Minami-osawa, Hachioji, Tokyo, 192-0397, Japan. .,Department of Applied Chemistry, School of Engineering, Tokyo University of Technology, 1401-1 Katakura, Hachioji, Tokyo, 192-0982, Japan.
| | - Maeva Zysman
- Univ Paris est Creteil, INSERM, IMRB, 94010, Creteil, France.,Univ Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC, 1401, Bordeaux, France.,Service des Maladies Respiratoires, CHU Bordeaux, Bordeaux, France
| | - Dan Elgrabli
- Univ Paris est Creteil, INSERM, IMRB, 94010, Creteil, France.,SAS NaorInnov, Courbevoie, France
| | - Toru Murayama
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Masatake Haruta
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Sophie Lanone
- Univ Paris est Creteil, INSERM, IMRB, 94010, Creteil, France
| | - Tamao Ishida
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1Minami-osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Jorge Boczkowski
- Univ Paris est Creteil, INSERM, IMRB, 94010, Creteil, France. .,AP-HP, Hopital Henri Mondor, Antenne de Pneumologie, 94010, Creteil, France.
| |
Collapse
|
23
|
Mumby S, Perros F, Hui C, Xu BL, Xu W, Elyasigomari V, Hautefort A, Manaud G, Humbert M, Chung KF, Wort SJ, Adcock IM. Extracellular matrix degradation pathways and fatty acid metabolism regulate distinct pulmonary vascular cell types in pulmonary arterial hypertension. Pulm Circ 2021; 11:2045894021996190. [PMID: 34408849 PMCID: PMC8366141 DOI: 10.1177/2045894021996190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/01/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary arterial hypertension describes a group of diseases characterised by raised pulmonary vascular resistance, resulting from vascular remodelling in the pre-capillary resistance arterioles. Left untreated, patients die from right heart failure. Pulmonary vascular remodelling involves all cell types but to date the precise roles of the different cells is unknown. This study investigated differences in basal gene expression between pulmonary arterial hypertension and controls using both human pulmonary microvascular endothelial cells and human pulmonary artery smooth muscle cells. Human pulmonary microvascular endothelial cells and human pulmonary artery smooth muscle cells from pulmonary arterial hypertension patients and controls were cultured to confluence, harvested and RNA extracted. Whole genome sequencing was performed and after transcript quantification and normalisation, we examined differentially expressed genes and applied gene set enrichment analysis to the differentially expressed genes to identify putative activated pathways. Human pulmonary microvascular endothelial cells displayed 1008 significant (p ≤ 0.0001) differentially expressed genes in pulmonary arterial hypertension samples compared to controls. In human pulmonary artery smooth muscle cells, there were 229 significant (p ≤ 0.0001) differentially expressed genes between pulmonary arterial hypertension and controls. Pathway analysis revealed distinctive differences: human pulmonary microvascular endothelial cells display down-regulation of extracellular matrix organisation, collagen formation and biosynthesis, focal- and cell-adhesion molecules suggesting severe endothelial barrier dysfunction and vascular permeability in pulmonary arterial hypertension pathogenesis. In contrast, pathways in human pulmonary artery smooth muscle cells were mainly up-regulated, including those for fatty acid metabolism, biosynthesis of unsaturated fatty acids, cell–cell and adherens junction interactions suggesting a more energy-driven proliferative phenotype. This suggests that the two cell types play different mechanistic roles in pulmonary arterial hypertension pathogenesis and further studies are required to fully elucidate the role each plays and the interactions between these cell types in vascular remodelling in disease progression.
Collapse
Affiliation(s)
- Sharon Mumby
- Respiratory Science, NHLI, Imperial College London, London, UK
| | - F Perros
- UMRS 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, INSERM and Paris-Sud, Le Plessis Robinson, France.,Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec, Canada
| | - C Hui
- Centre for Respiratory & Critical Care Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - B L Xu
- Centre for Respiratory & Critical Care Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - W Xu
- Centre for Respiratory & Critical Care Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - V Elyasigomari
- Department of Computing, Data Science Institute, Imperial College London, London, UK
| | - A Hautefort
- UMRS 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, INSERM and Paris-Sud, Le Plessis Robinson, France
| | - G Manaud
- UMRS 999, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, INSERM and Paris-Sud, Le Plessis Robinson, France
| | - M Humbert
- Département Hospitalo-Universitaire Thorax Innovation, Centre de Référence de l'Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - K F Chung
- Respiratory Science, NHLI, Imperial College London, London, UK
| | - S J Wort
- Respiratory Science, NHLI, Imperial College London, London, UK.,National Pulmonary Hypertension Service, Royal Brompton Hospital, London, UK
| | - I M Adcock
- Respiratory Science, NHLI, Imperial College London, London, UK
| |
Collapse
|
24
|
Nakahara M, Ito H, Skinner JT, Lin Q, Tamosiuniene R, Nicolls MR, Keegan AD, Johns RA, Yamaji-Kegan K. The inflammatory role of dysregulated IRS2 in pulmonary vascular remodeling under hypoxic conditions. Am J Physiol Lung Cell Mol Physiol 2021; 321:L416-L428. [PMID: 34189964 PMCID: PMC8410109 DOI: 10.1152/ajplung.00068.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.
Collapse
Affiliation(s)
- Mayumi Nakahara
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Homare Ito
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| | - John T Skinner
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Qing Lin
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Rasa Tamosiuniene
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California
| | - Achsah D Keegan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Roger A Johns
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Kazuyo Yamaji-Kegan
- Department of Anesthesiology, University of Maryland Baltimore, Baltimore, Maryland
| |
Collapse
|
25
|
Metabolomic Profile in Venous Thromboembolism (VTE). Metabolites 2021; 11:metabo11080495. [PMID: 34436436 PMCID: PMC8400436 DOI: 10.3390/metabo11080495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/19/2023] Open
Abstract
Venous thromboembolism (VTE) is a condition comprising deep venous thrombosis (DVT) and pulmonary embolism (PE). The prevalence of this disease is constantly increasing and it is also a chief reason for morbidity. Therefore, the primary prevention of VTE remains a highly important public health issue. At present, its diagnosis generally relies on subjective clinical examination and ultrasound imaging. D-dimer is also used as a biomarker, but it is considered to be poorly specific and only moderately sensitive. There are also no reliable methods that could accurately guide the type of treatment and potentially identify patients who may benefit from more aggressive therapies without the risk of bleeding. The application of metabolomics profiling in the area of vascular diseases may become a turning point in early diagnosis and patient management. Among the most described metabolites possibly related to VTE are carnitine species, glucose, phenylalanine, 3-hydroxybutarate, lactic acid, tryptophan and some monounsaturated and polyunsaturated fatty acids. The cell response to acute PE was suggested to involve the uncoupling between glycolysis and oxidative phosphorylation. Despite technological advancement in the identification of metabolites and their alteration in thrombosis, we still do not understand the mechanisms and pathways responsible for the occurrence of observed alterations.
Collapse
|
26
|
Li D, Yang S, Xing Y, Pan L, Zhao R, Zhao Y, Liu L, Wu M. Novel Insights and Current Evidence for Mechanisms of Atherosclerosis: Mitochondrial Dynamics as a Potential Therapeutic Target. Front Cell Dev Biol 2021; 9:673839. [PMID: 34307357 PMCID: PMC8293691 DOI: 10.3389/fcell.2021.673839] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death worldwide. Atherosclerosis is the underlying pathological basis of CVD. Mitochondrial homeostasis is maintained through the dynamic processes of fusion and fission. Mitochondria are involved in many cellular processes, such as steroid biosynthesis, calcium homeostasis, immune cell activation, redox signaling, apoptosis, and inflammation, among others. Under stress conditions, mitochondrial dynamics, mitochondrial cristae remodeling, and mitochondrial ROS (mitoROS) production increase, mitochondrial membrane potential (MMP) decreases, calcium homeostasis is imbalanced, and mitochondrial permeability transition pore open (mPTP) and release of mitochondrial DNA (mtDNA) are activated. mtDNA recognized by TLR9 can lead to NF-κB pathway activation and pro-inflammatory factor expression. At the same time, TLR9 can also activate NLRP3 inflammasomes and release interleukin, an event that eventually leads to tissue damage and inflammatory responses. In addition, mitochondrial dysfunction may amplify the activation of NLRP3 through the production of mitochondrial ROS, which together aggravate accumulating mitochondrial damage. In addition, mtDNA defects or gene mutation can lead to mitochondrial oxidative stress. Finally, obesity, diabetes, hypertension and aging are risk factors for the progression of CVD, which are closely related to mitochondrial dynamics. Mitochondrial dynamics may represent a new target in the treatment of atherosclerosis. Antioxidants, mitochondrial inhibitors, and various new therapies to correct mitochondrial dysfunction represent a few directions for future research on therapeutic intervention and amelioration of atherosclerosis.
Collapse
Affiliation(s)
- Dan Li
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Limin Pan
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yixi Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, Fukai T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid Redox Signal 2021; 34:1319-1354. [PMID: 33899493 PMCID: PMC8418449 DOI: 10.1089/ars.2020.8161] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS; e.g., superoxide [O2•-] and hydrogen peroxide [H2O2]) and reactive nitrogen species (RNS; e.g., nitric oxide [NO•]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal. 34, 1319-1354.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
28
|
Mprah R, Adzika GK, Gyasi YI, Ndzie Noah ML, Adu-Amankwaah J, Adekunle AO, Duah M, Wowui PI, Weili Q. Glutaminolysis: A Driver of Vascular and Cardiac Remodeling in Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:667446. [PMID: 33996951 PMCID: PMC8113389 DOI: 10.3389/fcvm.2021.667446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a decimating ailment described by chronic precapillary pulmonary hypertension, an elevated mean pulmonary arterial pressure with a normal pulmonary capillary wedge pressure, and a raised pulmonary vascular resistance resulting in increased right ventricular afterload culminating in heart failure and death. Current PAH treatments regulate the vasodilatory/vasoconstrictory balance of pulmonary vessels. However, these treatment options are unable to stop the progression of, or reverse, an already established disease. Recent studies have advanced a metabolic dysregulation, featuring increased glutamine metabolism, as a mechanism driving PAH progression. Metabolic dysregulation in PAH leads to increased glutaminolysis to produce substrate to meet the high-energy requirement by hyperproliferative and apoptosis-resistant pulmonary vascular cells. This article explores the role of glutamate metabolism in PAH and how it could be targeted as an anti-remodeling therapeutic strategy.
Collapse
Affiliation(s)
- Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Yusif I. Gyasi
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, TX, United States
| | | | | | | | - Maxwell Duah
- Haematology Department, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Qiao Weili
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
29
|
Coleman RD, Chartan CA, Mourani PM. Intensive care management of right ventricular failure and pulmonary hypertension crises. Pediatr Pulmonol 2021; 56:636-648. [PMID: 33561307 DOI: 10.1002/ppul.24776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/03/2020] [Indexed: 01/22/2023]
Abstract
Pulmonary hypertension (PH), an often unrelenting disease that carries with it significant morbidity and mortality, affects not only the pulmonary vasculature but, in turn, the right ventricle as well. The survival of patients with PH is closely related to the right ventricular function. Therefore, having an understanding of how to manage right ventricular failure (RVF) and acute pulmonary hypertensive crises is imperative for clinicians who encounter these patients. This review addresses the management of these patients in detail, addressing: (a) the pathophysiology of RVF, (b) intensive care monitoring of these patients in the intensive care unit, (c) imaging of the right ventricle, (d) intubation and mechanical ventilation, (e) inotrope and vasopressor selection, (f) pulmonary vasodilator use, (g) interventional and surgical procedures for the acutely failing right ventricle, and (h) mechanical support for RVF.
Collapse
Affiliation(s)
- Ryan D Coleman
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Section of Pulmonary Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Corey A Chartan
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Section of Pulmonary Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Peter M Mourani
- Section of Critical Care Medicine and Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
30
|
Peng H, Wang X, Du J, Cui Q, Huang Y, Jin H. Metabolic Reprogramming of Vascular Endothelial Cells: Basic Research and Clinical Applications. Front Cell Dev Biol 2021; 9:626047. [PMID: 33681205 PMCID: PMC7930387 DOI: 10.3389/fcell.2021.626047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/26/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular endothelial cells (VECs) build a barrier separating the blood from the vascular wall. The vascular endothelium is the largest endocrine organ, and is well-known for its crucial role in the regulation of vascular function. The initial response to endothelial cell injury can lead to the activation of VECs. However, excessive activation leads to metabolic pathway disruption, VEC dysfunction, and angiogenesis. The pathways related to VEC metabolic reprogramming recently have been considered as key modulators of VEC function in processes such as angiogenesis, inflammation, and barrier maintenance. In this review, we focus on the changes of VEC metabolism under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Hanlin Peng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiuli Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Qinghua Cui
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.,Department of Biomedical Informatics, Centre for Non-coding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
31
|
Gropler RJ. Imaging Myocardial Metabolism. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00083-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
32
|
Swietlik EM, Ghataorhe P, Zalewska KI, Wharton J, Howard LS, Taboada D, Cannon JE, Morrell NW, Wilkins MR, Toshner M, Pepke-Zaba J, Rhodes CJ. Plasma metabolomics exhibit response to therapy in chronic thromboembolic pulmonary hypertension. Eur Respir J 2020; 57:13993003.03201-2020. [PMID: 33060150 PMCID: PMC8012591 DOI: 10.1183/13993003.03201-2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 09/20/2020] [Indexed: 12/31/2022]
Abstract
Pulmonary hypertension is a condition with limited effective treatment options. Chronic thromboembolic pulmonary hypertension (CTEPH) is a notable exception, with pulmonary endarterectomy (PEA) often proving curative. This study investigated the plasma metabolome of CTEPH patients, estimated reversibility to an effective treatment and explored the source of metabolic perturbations.We performed untargeted analysis of plasma metabolites in CTEPH patients compared to healthy controls and disease comparators. Changes in metabolic profile were evaluated in response to PEA. A subset of patients were sampled at three anatomical locations and plasma metabolite gradients calculated.We defined and validated altered plasma metabolite profiles in patients with CTEPH. 12 metabolites were confirmed by receiver operating characteristic analysis to distinguish CTEPH and both healthy (area under the curve (AUC) 0.64-0.94, all p<2×10-5) and disease controls (AUC 0.58-0.77, all p<0.05). Many of the metabolic changes were notably similar to those observed in idiopathic pulmonary arterial hypertension (IPAH). Only five metabolites (5-methylthioadenosine, N1-methyladenosine, N1-methylinosine, 7-methylguanine, N-formylmethionine) distinguished CTEPH from chronic thromboembolic disease or IPAH. Significant corrections (15-100% of perturbation) in response to PEA were observed in some, but not all metabolites. Anatomical sampling identified 188 plasma metabolites, with significant gradients in tryptophan, sphingomyelin, methionine and Krebs cycle metabolites. In addition, metabolites associated with CTEPH and gradients showed significant associations with clinical measures of disease severity.We identified a specific metabolic profile that distinguishes CTEPH from controls and disease comparators, despite the observation that most metabolic changes were common to both CTEPH and IPAH patients. Plasma metabolite gradients implicate cardiopulmonary tissue metabolism of metabolites associated with pulmonary hypertension and metabolites that respond to PEA surgery could be a suitable noninvasive marker for evaluating future targeted therapeutic interventions.
Collapse
Affiliation(s)
- Emilia M Swietlik
- Dept of Medicine, University of Cambridge, Cambridge, UK.,National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK
| | - Pavandeep Ghataorhe
- National Heart and Lung Institute, Medicine, Imperial College London, London, UK
| | - Kasia I Zalewska
- National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK.,Respiratory Unit, University Hospital Llandough, Cardiff, UK
| | - John Wharton
- National Heart and Lung Institute, Medicine, Imperial College London, London, UK
| | - Luke S Howard
- National Pulmonary Hypertension Service, Imperial College Healthcare NHS Trust and NHLI, Imperial College, Hammersmith Hospital, London, UK
| | - Dolores Taboada
- National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK
| | - John E Cannon
- National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK
| | | | | | - Martin R Wilkins
- National Heart and Lung Institute, Medicine, Imperial College London, London, UK
| | - Mark Toshner
- Dept of Medicine, University of Cambridge, Cambridge, UK.,National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK
| | - Joanna Pepke-Zaba
- Dept of Medicine, University of Cambridge, Cambridge, UK.,National Pulmonary Hypertension Service, Royal Papworth Hospital, Cambridge, UK
| | - Christopher J Rhodes
- National Heart and Lung Institute, Medicine, Imperial College London, London, UK
| |
Collapse
|
33
|
3-Bromopyruvate alleviates the development of monocrotaline-induced rat pulmonary arterial hypertension by decreasing aerobic glycolysis, inducing apoptosis, and suppressing inflammation. Chin Med J (Engl) 2020; 133:49-60. [PMID: 31923104 PMCID: PMC7028200 DOI: 10.1097/cm9.0000000000000577] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PH) is a progressive disease with limited therapeutic options, ultimately leading to right heart failure and death. Recent findings indicate the role of the Warburg effect (aerobic glycolysis) in the development of PH. However, the effect of the glycolysis inhibitor 3-bromopyruvate (3-BrPA) on the pathogenesis of PH has not been well investigated. This study aimed to determine whether 3-BrPA inhibits PH and its possible mechanism. METHODS PH was induced in adult Sprague-Dawley rats by a single intraperitoneal injection of monocrotaline (MCT). 3-BrPA, or phosphate-buffered saline (PBS) was administered via intraperitoneal injection every other day from the first day of MCT-injection to 4 weeks of follow-up, and indices such as right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), pulmonary arteriolar remodeling indicated by percent media thickness (% MT), lactate levels and glucose consumption, were evaluated. Pulmonary arteriolar remodeling and right ventricular hypertrophy were observed in hematoxylin-eosin-stained lung sections. Western blotting, immunohistochemistry, and/or immunofluorescence analyses were used to measure the expression of relevant proteins. A cytochrome C release apoptosis assay and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling staining were used to measure cell apoptosis. RESULTS MCT-induced PH showed a significant increase in glucose consumption (0 vs. 4 weeks: 0.87 ± 0.23 vs. 2.94 ± 0.47, P = 0.0042) and lactate production (0 vs. 4 weeks: 4.19 ± 0.34 vs. 8.06 ± 0.67, P = 0.0004). Treatment with 3-BrPA resulted in a concomitant reduction in glucose consumption (1.10 ± 0.35 vs. 3.25 ± 0.47, P = 0.0063), lactate production (5.09 ± 0.55 vs. 8.06 ± 0.67, P = 0.0065), MCT-induced increase in RVSP (39.70 ± 2.94 vs. 58.85 ± 2.32, P = 0.0004), pulmonary vascular remodeling (% MT, 43.45% ± 1.41% vs. 63.66% ± 1.78%, P < 0.0001), and right ventricular hypertrophy (RVHI, 38.57% ± 2.69% vs. 62.61% ± 1.57%, P < 0.0001) when compared with those of the PBS-treated group. 3-BrPA, a hexokinase 2 inhibitor, exerted its beneficial effect on PH by decreasing aerobic glycolysis and was also associated with inhibiting the expression of glucose transporter protein-1, inducing apoptosis, and suppressing inflammation. CONCLUSIONS 3-BrPA might have a potential beneficial effect on the PH treatment.
Collapse
|
34
|
Shi J, Yang Y, Cheng A, Xu G, He F. Metabolism of vascular smooth muscle cells in vascular diseases. Am J Physiol Heart Circ Physiol 2020; 319:H613-H631. [PMID: 32762559 DOI: 10.1152/ajpheart.00220.2020] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the fundamental component of the medial layer of arteries and are essential for arterial physiology and pathology. It is becoming increasingly clear that VSMCs can alter their metabolism to fulfill the bioenergetic and biosynthetic requirements. During vascular injury, VSMCs switch from a quiescent "contractile" phenotype to a highly migratory and proliferative "synthetic" phenotype. Recent studies have found that the phenotype switching of VSMCs is driven by a metabolic switch. Metabolic pathways, including aerobic glycolysis, fatty acid oxidation, and amino acid metabolism, have distinct, indispensable roles in normal and dysfunctional vasculature. VSMCs metabolism is also related to the metabolism of endothelial cells. In the present review, we present a brief overview of VSMCs metabolism and how it regulates the progression of several vascular diseases, including atherosclerosis, systemic hypertension, diabetes, pulmonary hypertension, vascular calcification, and aneurysms, and the effect of the risk factors for vascular disease (aging, cigarette smoking, and excessive alcohol drinking) on VSMC metabolism to clarify the role of VSMCs metabolism in the key pathological process.
Collapse
Affiliation(s)
- Jia Shi
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anying Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Hsu JY, Major JL, Riching AS, Sen R, Pires da Silva J, Bagchi RA. Beyond the genome: challenges and potential for epigenetics-driven therapeutic approaches in pulmonary arterial hypertension. Biochem Cell Biol 2020; 98:631-646. [PMID: 32706995 DOI: 10.1139/bcb-2020-0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease of the cardiopulmonary system caused by the narrowing of the pulmonary arteries, leading to increased vascular resistance and pressure. This leads to right ventricle remodeling, dysfunction, and eventually, death. While conventional therapies have largely focused on targeting vasodilation, other pathological features of PAH including aberrant inflammation, mitochondrial dynamics, cell proliferation, and migration have not been well explored. Thus, despite some recent improvements in PAH treatment, the life expectancy and quality of life for patients with PAH remains poor. Showing many similarities to cancers, PAH is characterized by increased pulmonary arterial smooth muscle cell proliferation, decreased apoptotic signaling pathways, and changes in metabolism. The recent successes of therapies targeting epigenetic modifiers for the treatment of cancer has prompted epigenetic research in PAH, revealing many new potential therapeutic targets. In this minireview we discuss the emergence of epigenetic dysregulation in PAH and highlight epigenetic-targeting compounds that may be effective for the treatment of PAH.
Collapse
Affiliation(s)
- Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jennifer L Major
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew S Riching
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rwik Sen
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Julie Pires da Silva
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
36
|
Tofovic SP, Jackson EK. Estradiol Metabolism: Crossroads in Pulmonary Arterial Hypertension. Int J Mol Sci 2019; 21:ijms21010116. [PMID: 31877978 PMCID: PMC6982327 DOI: 10.3390/ijms21010116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a debilitating and progressive disease that predominantly develops in women. Over the past 15 years, cumulating evidence has pointed toward dysregulated metabolism of sex hormones in animal models and patients with PAH. 17β-estradiol (E2) is metabolized at positions C2, C4, and C16, which leads to the formation of metabolites with different biological/estrogenic activity. Since the first report that 2-methoxyestradiol, a major non-estrogenic metabolite of E2, attenuates the development and progression of experimental pulmonary hypertension (PH), it has become increasingly clear that E2, E2 precursors, and E2 metabolites exhibit both protective and detrimental effects in PH. Furthermore, both experimental and clinical data suggest that E2 has divergent effects in the pulmonary vasculature versus right ventricle (estrogen paradox in PAH). The estrogen paradox is of significant clinical relevance for understanding the development, progression, and prognosis of PAH. This review updates experimental and clinical findings and provides insights into: (1) the potential impacts that pathways of estradiol metabolism (EMet) may have in PAH; (2) the beneficial and adverse effects of estrogens and their precursors/metabolites in experimental PH and human PAH; (3) the co-morbidities and pathological conditions that may alter EMet and influence the development/progression of PAH; (4) the relevance of the intracrinology of sex hormones to vascular remodeling in PAH; and (5) the advantages/disadvantages of different approaches to modulate EMet in PAH. Finally, we propose the three-tier-estrogen effects in PAH concept, which may offer reconciliation of the opposing effects of E2 in PAH and may provide a better understanding of the complex mechanisms by which EMet affects the pulmonary circulation–right ventricular interaction in PAH.
Collapse
Affiliation(s)
- Stevan P. Tofovic
- Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, BST E1240, 200 Lothrop Street, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine, 100 Technology Drive, PA 15219, USA;
- Correspondence: ; Tel.: +1-412-648-3363
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology University of Pittsburgh School of Medicine, 100 Technology Drive, PA 15219, USA;
| |
Collapse
|
37
|
Barnes JW, Tian L, Krick S, Helton ES, Denson RS, Comhair SAA, Dweik RA. O-GlcNAc Transferase Regulates Angiogenesis in Idiopathic Pulmonary Arterial Hypertension. Int J Mol Sci 2019; 20:E6299. [PMID: 31847126 PMCID: PMC6941156 DOI: 10.3390/ijms20246299] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is considered a vasculopathy characterized by elevated pulmonary vascular resistance due to vasoconstriction and/or lung remodeling such as plexiform lesions, the hallmark of the PAH, as well as cell proliferation and vascular and angiogenic dysfunction. The serine/threonine hydroxyl-linked N-Acetylglucosamine (O-GlcNAc) transferase (OGT) has been shown to drive pulmonary arterial smooth muscle cell (PASMC) proliferation in IPAH. OGT is a cellular nutrient sensor that is essential in maintaining proper cell function through the regulation of cell signaling, proliferation, and metabolism. The aim of this study was to determine the role of OGT and O-GlcNAc in vascular and angiogenic dysfunction in IPAH. Primary isolated human control and IPAH patient PASMCs and pulmonary arterial endothelial cells (PAECs) were grown in the presence or absence of OGT inhibitors and subjected to biochemical assessments in monolayer cultures and tube formation assays, in vitro vascular sprouting 3D spheroid co-culture models, and de novo vascularization models in NODSCID mice. We showed that knockdown of OGT resulted in reduced vascular endothelial growth factor (VEGF) expression in IPAH primary isolated vascular cells. In addition, specificity protein 1 (SP1), a known stimulator of VEGF expression, was shown to have higher O-GlcNAc levels in IPAH compared to control at physiological (5 mM) and high (25 mM) glucose concentrations, and knockdown resulted in decreased VEGF protein levels. Furthermore, human IPAH PAECs demonstrated a significantly higher degree of capillary tube-like structures and increased length compared to control PAECs. Addition of an OGT inhibitor, OSMI-1, significantly reduced the number of tube-like structures and tube length similar to control levels. Assessment of vascular sprouting from an in vitro 3D spheroid co-culture model using IPAH and control PAEC/PASMCs and an in vivo vascularization model using control and PAEC-embedded collagen implants demonstrated higher vascularization in IPAH compared to control. Blocking OGT activity in these experiments, however, altered the vascular sprouting and de novo vascularization in IPAH similar to control levels when compared to controls. Our findings in this report are the first to describe a role for the OGT/O-GlcNAc axis in modulating VEGF expression and vascularization in IPAH. These findings provide greater insight into the potential role that altered glucose uptake and metabolism may have on the angiogenic process and the development of plexiform lesions. Therefore, we believe that the OGT/O-GlcNAc axis may be a potential therapeutic target for treating the angiogenic dysregulation that is present in IPAH.
Collapse
Affiliation(s)
- Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, THT 422, 1720 2nd Ave S, Birmingham, AL 35294-0006, USA; (S.K.); (E.S.H.)
| | - Liping Tian
- Department of Inflammation & Immunity, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA; (L.T.); (S.A.A.C.); (R.A.D.)
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, THT 422, 1720 2nd Ave S, Birmingham, AL 35294-0006, USA; (S.K.); (E.S.H.)
| | - E. Scott Helton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, THT 422, 1720 2nd Ave S, Birmingham, AL 35294-0006, USA; (S.K.); (E.S.H.)
| | - Rebecca S. Denson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, THT 422, 1720 2nd Ave S, Birmingham, AL 35294-0006, USA; (S.K.); (E.S.H.)
| | - Suzy A. A. Comhair
- Department of Inflammation & Immunity, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA; (L.T.); (S.A.A.C.); (R.A.D.)
| | - Raed A. Dweik
- Department of Inflammation & Immunity, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA; (L.T.); (S.A.A.C.); (R.A.D.)
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
38
|
Otálora-Otálora BA, Florez M, López-Kleine L, Canas Arboleda A, Grajales Urrego DM, Rojas A. Joint Transcriptomic Analysis of Lung Cancer and Other Lung Diseases. Front Genet 2019; 10:1260. [PMID: 31867044 PMCID: PMC6908522 DOI: 10.3389/fgene.2019.01260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/14/2019] [Indexed: 12/09/2022] Open
Abstract
Background: Epidemiological and clinical evidence points cancer comorbidity with pulmonary chronic disease. The acquisition of some hallmarks of cancer by cells affected with lung pathologies as a cell adaptive mechanism to a shear stress, suggests that could be associated with the establishment of tumoral processes. Objective: To propose a bioinformatic pipeline for the identification of all deregulated genes and the transcriptional regulators (TFs) that are coexpressed during lung cancer establishment, and therefore could be important for the acquisition of the hallmarks of cancer. Methods: Ten microarray datasets (six of lung cancer, four of lung diseases) comparing normal and diseases-related lung tissue were selected to identify hub differentiated expressed genes (DEGs) in common between lung pathologies and lung cancer, along with transcriptional regulators through the utilization of specialized libraries from R language. DAVID bioinformatics tool for gene enrichment analyses was used to identify genes with experimental evidence associated to tumoral processes and signaling pathways. Coexpression networks of DEGs and TFs in lung cancer establishment were created with Coexnet library, and a survival analysis of the main hub genes was made. Results: Two hundred ten DEGs were identified in common between lung cancer and other lung diseases related to the acquisition of tumoral characteristics, which are coexpressed in a lung cancer network with TFs, suggesting that could be related to the establishment of the tumoral pathology in lung. The comparison of the coexpression networks of lung cancer and other lung diseases allowed the identification of common connectivity patterns (CCPs) with DEGs and TFs correlated to important tumoral processes and signaling pathways, that haven´t been studied to experimentally validate their role in the early stages of lung cancer. Some of the TFs identified showed a correlation between its expression levels and the survival of lung cancer patients. Conclusion: Our findings indicate that lung diseases share genes with lung cancer which are coexpressed in lung cancer, and might be able to explain the epidemiological observations that point to direct and inverse comorbid associations between some chronic lung diseases and lung cancer and represent a complex transcriptomic scenario.
Collapse
Affiliation(s)
| | - Mauro Florez
- Departamento de Estadística, Grupo de Investigación en Bioinformática y Biología de sistemas – GiBBS, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Liliana López-Kleine
- Departamento de Estadística, Grupo de Investigación en Bioinformática y Biología de sistemas – GiBBS, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | | | | | - Adriana Rojas
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
39
|
Fernández AI, Yotti R, González-Mansilla A, Mombiela T, Gutiérrez-Ibanes E, Pérez del Villar C, Navas-Tejedor P, Chazo C, Martínez-Legazpi P, Fernández-Avilés F, Bermejo J. The Biological Bases of Group 2 Pulmonary Hypertension. Int J Mol Sci 2019; 20:ijms20235884. [PMID: 31771195 PMCID: PMC6928720 DOI: 10.3390/ijms20235884] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension (PH) is a potentially fatal condition with a prevalence of around 1% in the world population and most commonly caused by left heart disease (PH-LHD). Usually, in PH-LHD, the increase of pulmonary pressure is only conditioned by the retrograde transmission of the left atrial pressure. However, in some cases, the long-term retrograde pressure overload may trigger complex and irreversible biomechanical and biological changes in the pulmonary vasculature. This latter clinical entity, designated as combined pre- and post-capillary PH, is associated with very poor outcomes. The underlying mechanisms of this progression are poorly understood, and most of the current knowledge comes from the field of Group 1-PAH. Treatment is also an unsolved issue in patients with PH-LHD. Targeting the molecular pathways that regulate pulmonary hemodynamics and vascular remodeling has provided excellent results in other forms of PH but has a neutral or detrimental result in patients with PH-LHD. Therefore, a deep and comprehensive biological characterization of PH-LHD is essential to improve the diagnostic and prognostic evaluation of patients and, eventually, identify new therapeutic targets. Ongoing research is aimed at identify candidate genes, variants, non-coding RNAs, and other biomarkers with potential diagnostic and therapeutic implications. In this review, we discuss the state-of-the-art cellular, molecular, genetic, and epigenetic mechanisms potentially involved in PH-LHD. Signaling and effective pathways are particularly emphasized, as well as the current knowledge on -omic biomarkers. Our final aim is to provide readers with the biological foundations on which to ground both clinical and pre-clinical research in the field of PH-LHD.
Collapse
Affiliation(s)
- Ana I. Fernández
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Raquel Yotti
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Ana González-Mansilla
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Teresa Mombiela
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Enrique Gutiérrez-Ibanes
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Candelas Pérez del Villar
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Paula Navas-Tejedor
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Christian Chazo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Pablo Martínez-Legazpi
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
| | - Javier Bermejo
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; (A.I.F.); (R.Y.); (A.G.-M.); (T.M.); (E.G.-I.); (C.P.d.V.); (P.N.-T.); (C.C.); (P.M.-L.); (F.F.-A.)
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación Biomédica en Red, CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain
- Facultad de Medicine, Universidad Complutense de Madrid, 28007 Madrid, Spain
- Correspondence: ; Tel.: +34-91-586-8279
| |
Collapse
|
40
|
Alruwaili N, Kandhi S, Sun D, Wolin MS. Metabolism and Redox in Pulmonary Vascular Physiology and Pathophysiology. Antioxid Redox Signal 2019; 31:752-769. [PMID: 30403147 PMCID: PMC6708269 DOI: 10.1089/ars.2018.7657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: This review considers how some systems controlling pulmonary vascular function are potentially regulated by redox processes to examine how and why conditions such as prolonged hypoxia, pathological mediators, and other factors promoting vascular remodeling contribute to the development of pulmonary hypertension (PH). Recent Advances and Critical Issues: Aspects of vascular remodeling induction mechanisms described are associated with shifts in glucose metabolism through the pentose phosphate pathway and increased cytosolic NADPH generation by glucose-6-phosphate dehydrogenase, increased glycolysis generation of cytosolic NADH and lactate, mitochondrial dysfunction associated with superoxide dismutase-2 depletion, changes in reactive oxygen species and iron metabolism, and redox signaling. Future Directions: The regulation and impact of hypoxia-inducible factor and the function of cGMP-dependent and redox regulation of protein kinase G are considered for their potential roles as key sensors and coordinators of redox and metabolic processes controlling the progression of vascular pathophysiology in PH, and how modulating aspects of metabolic and redox regulatory systems potentially function in beneficial therapeutic approaches.
Collapse
Affiliation(s)
- Norah Alruwaili
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Sharath Kandhi
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Dong Sun
- Department of Physiology, New York Medical College, Valhalla, New York
| | - Michael S Wolin
- Department of Physiology, New York Medical College, Valhalla, New York
| |
Collapse
|
41
|
Kovacs L, Cao Y, Han W, Meadows L, Kovacs-Kasa A, Kondrikov D, Verin AD, Barman SA, Dong Z, Huo Y, Su Y. PFKFB3 in Smooth Muscle Promotes Vascular Remodeling in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2019; 200:617-627. [PMID: 30817168 PMCID: PMC6727156 DOI: 10.1164/rccm.201812-2290oc] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/27/2019] [Indexed: 12/29/2022] Open
Abstract
Rationale: Glycolytic shift is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). It remains unknown how glycolysis is increased and how increased glycolysis contributes to pulmonary vascular remodeling in PAH.Objectives: To determine whether increased glycolysis is caused by 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and how PFKFB3-driven glycolysis induces vascular remodeling in PAH.Methods: PFKFB3 levels were measured in pulmonary arteries of patients and animals with PAH. Lactate levels were assessed in lungs of animals with PAH and in pulmonary artery smooth muscle cells (PASMCs). Genetic and pharmacologic approaches were used to investigate the role of PFKFB3 in PAH.Measurements and Main Results: Lactate production was elevated in lungs of PAH rodents and in platelet-derived growth factor-treated PASMCs. PFKFB3 protein was higher in pulmonary arteries of patients and rodents with PAH, in PASMCs of patients with PAH, and in platelet-derived growth factor-treated PASMCs. PFKFB3 inhibition by genetic disruption and chemical inhibitor attenuated phosphorylation/activation of extracellular signal-regulated kinase (ERK1/2) and calpain-2, and vascular remodeling in PAH rodent models, and reduced platelet-derived growth factor-induced phosphorylation/activation of ERK1/2 and calpain-2, collagen synthesis and proliferation of PASMCs. ERK1/2 inhibition attenuated phosphorylation/activation of calpain-2, and vascular remodeling in Sugen/hypoxia PAH rats, and reduced lactate-induced phosphorylation/activation of calpain-2, collagen synthesis, and proliferation of PASMCs. Calpain-2 inhibition reduced lactate-induced collagen synthesis and proliferation of PASMCs.Conclusions: Upregulated PFKFB3 mediates collagen synthesis and proliferation of PASMCs, contributing to vascular remodeling in PAH. The mechanism is through the elevation of glycolysis and lactate that results in the activation of calpain by ERK1/2-dependent phosphorylation of calpain-2.
Collapse
Affiliation(s)
| | - Yapeng Cao
- Vascular Biology Center
- Drug Discovery Center, State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China; and
| | | | | | | | | | | | | | - Zheng Dong
- Department of Cellular Biology and Anatomy, and
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Yuqing Huo
- Vascular Biology Center
- Department of Cellular Biology and Anatomy, and
| | - Yunchao Su
- Department of Pharmacology and Toxicology
- Vascular Biology Center
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
42
|
Stenmark KR, Frid MG, Graham BB, Tuder RM. Dynamic and diverse changes in the functional properties of vascular smooth muscle cells in pulmonary hypertension. Cardiovasc Res 2019; 114:551-564. [PMID: 29385432 DOI: 10.1093/cvr/cvy004] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/26/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension (PH) is the end result of interaction between pulmonary vascular tone and a complex series of cellular and molecular events termed 'vascular remodelling'. The remodelling process, which can involve the entirety of pulmonary arterial vasculature, almost universally involves medial thickening, driven by increased numbers and hypertrophy of its principal cellular constituent, smooth muscle cells (SMCs). It is noted, however that SMCs comprise heterogeneous populations of cells, which can exhibit markedly different proliferative, inflammatory, and extracellular matrix production changes during remodelling. We further consider that these functional changes in SMCs of different phenotype and their role in PH are dynamic and may undergo significant changes over time (which we will refer to as cellular plasticity); no single property can account for the complexity of the contribution of SMC to pulmonary vascular remodelling. Thus, the approaches used to pharmacologically manipulate PH by targeting the SMC phenotype(s) must take into account processes that underlie dominant phenotypes that drive the disease. We present evidence for time- and location-specific changes in SMC proliferation in various animal models of PH; we highlight the transient nature (rather than continuous) of SMC proliferation, emphasizing that the heterogenic SMC populations that reside in different locations along the pulmonary vascular tree exhibit distinct responses to the stresses associated with the development of PH. We also consider that cells that have often been termed 'SMCs' may arise from many origins, including endothelial cells, fibroblasts and resident or circulating progenitors, and thus may contribute via distinct signalling pathways to the remodelling process. Ultimately, PH is characterized by long-lived, apoptosis-resistant SMC. In line with this key pathogenic characteristic, we address the acquisition of a pro-inflammatory phenotype by SMC that is essential to the development of PH. We present evidence that metabolic alterations akin to those observed in cancer cells (cytoplasmic and mitochondrial) directly contribute to the phenotype of the SM and SM-like cells involved in PH. Finally, we raise the possibility that SMCs transition from a proliferative to a senescent, pro-inflammatory and metabolically active phenotype over time.
Collapse
Affiliation(s)
- Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Brian B Graham
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| | - Rubin M Tuder
- Pulmonary and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Avenue, RC2, B131, Aurora, CO 80045, USA
| |
Collapse
|
43
|
Dabral S, Muecke C, Valasarajan C, Schmoranzer M, Wietelmann A, Semenza GL, Meister M, Muley T, Seeger-Nukpezah T, Samakovlis C, Weissmann N, Grimminger F, Seeger W, Savai R, Pullamsetti SS. A RASSF1A-HIF1α loop drives Warburg effect in cancer and pulmonary hypertension. Nat Commun 2019; 10:2130. [PMID: 31086178 PMCID: PMC6513860 DOI: 10.1038/s41467-019-10044-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 04/05/2019] [Indexed: 12/13/2022] Open
Abstract
Hypoxia signaling plays a major role in non-malignant and malignant hyperproliferative diseases. Pulmonary hypertension (PH), a hypoxia-driven vascular disease, is characterized by a glycolytic switch similar to the Warburg effect in cancer. Ras association domain family 1A (RASSF1A) is a scaffold protein that acts as a tumour suppressor. Here we show that hypoxia promotes stabilization of RASSF1A through NOX-1- and protein kinase C- dependent phosphorylation. In parallel, hypoxia inducible factor-1 α (HIF-1α) activates RASSF1A transcription via HIF-binding sites in the RASSF1A promoter region. Vice versa, RASSF1A binds to HIF-1α, blocks its prolyl-hydroxylation and proteasomal degradation, and thus enhances the activation of the glycolytic switch. We find that this mechanism operates in experimental hypoxia-induced PH, which is blocked in RASSF1A knockout mice, in human primary PH vascular cells, and in a subset of human lung cancer cells. We conclude that RASSF1A-HIF-1α forms a feedforward loop driving hypoxia signaling in PH and cancer.
Collapse
Affiliation(s)
- Swati Dabral
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Christian Muecke
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Chanil Valasarajan
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Mario Schmoranzer
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Astrid Wietelmann
- MRI and µCT Service Group, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany
| | - Gregg L Semenza
- Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, Biological Chemistry, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD21205, MD, USA
| | - Michael Meister
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, 69126, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, 69120, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, 69126, Germany.,Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, 69120, Germany
| | - Tamina Seeger-Nukpezah
- Department I of Internal Medicine and Center for Integrated Oncology, University of Cologne, Cologne, 50937, Germany
| | - Christos Samakovlis
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691, Stockholm, Sweden.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany
| | - Norbert Weissmann
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany
| | - Werner Seeger
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany
| | - Rajkumar Savai
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany.,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany
| | - Soni S Pullamsetti
- Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, 61231, Germany. .,Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), ECCPS, Member of the DZL, Justus-Liebig University, Giessen, 35392, Germany.
| |
Collapse
|
44
|
Latham GJ, Yung D. Current understanding and perioperative management of pediatric pulmonary hypertension. Paediatr Anaesth 2019; 29:441-456. [PMID: 30414333 DOI: 10.1111/pan.13542] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/20/2018] [Accepted: 11/02/2018] [Indexed: 11/27/2022]
Abstract
Pediatric pulmonary hypertension is a complex disease with multiple, diverse etiologies affecting the premature neonate to the young adult. Pediatric pulmonary arterial hypertension, whether idiopathic or associated with congenital heart disease, is the most commonly discussed form of pediatric pulmonary hypertension, as it is progressive and lethal. However, neonatal forms of pulmonary hypertension are vastly more frequent, and while most cases are transient, the risk of morbidity and mortality in this group deserves recognition. Pulmonary hypertension due to left heart disease is another subset increasingly recognized as an important cause of pediatric pulmonary hypertension. One aspect of pediatric pulmonary hypertension is very clear: anesthetizing the child with pulmonary hypertension is associated with a significantly heightened risk of morbidity and mortality. It is therefore imperative that anesthesiologists who care for children with pulmonary hypertension have a firm understanding of the pathophysiology of the various forms of pediatric pulmonary hypertension, the impact of anesthesia and sedation in the setting of pulmonary hypertension, and anesthesiologists' role as perioperative experts from preoperative planning to postoperative disposition. This review summarizes the current understanding of pediatric pulmonary hypertension physiology, preoperative risk stratification, anesthetic risk, and intraoperative considerations relevant to the underlying pathophysiology of various forms of pediatric pulmonary hypertension.
Collapse
Affiliation(s)
- Gregory J Latham
- Department of Anesthesiology and Pain Medicine, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, Washington
| | - Delphine Yung
- Department of Pediatric Cardiology, Seattle Children's Hospital, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
45
|
Zhang YL, Zhang R, Shen YF, Huang KY, He YY, Zhao JH, Jing ZC. 3-Bromopyruvate Attenuates Experimental Pulmonary Hypertension via Inhibition of Glycolysis. Am J Hypertens 2019; 32:426-432. [PMID: 30561502 DOI: 10.1093/ajh/hpy191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/10/2018] [Accepted: 12/12/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The shift of metabolism from mitochondrial oxidative phosphorylation to glycolysis and mitochondria binding partner of hexokinase are features common to cancer. These have been seen in pulmonary hypertension (PH) as well. An inhibitor of hexokinase 2 (HK 2), the small molecule 3-bromopyruvate (3-BrPA) is an incredibly powerful and swift-acting anticancer agent. However, whether it could be of potential benefit to PH has still been unknown. METHODS Sprague-Dawley rats with monocrotaline (MCT)-induced PH were administered 2 oral doses of 3-BrPA (15 and 30 mg/kg/day, respectively) for 14 days. Hemodynamic parameters were obtained by right heart catheterization. Histopathology, immunohistochemistry, transmission electron microscopy, flow cytometry, and assessments of relative protein expressions were conducted. RESULTS Compared with MCT treatment, 3-BrPA decreased mean pulmonary arterial pressure and pulmonary vascular resistance, and increased cardiac output. 3-BrPA significantly suppressed proliferation in addition to enhancing apoptosis of pulmonary artery smooth muscle cells, attenuating small pulmonary artery remodeling and right ventricular hypertrophy. Treatment with 3-BrPA markedly reduced the mitochondrial membrane potential and restored mitochondrial structure. Furthermore, 3-BrPA significantly inhibited HK 2 expression but not HK 1. The expression of both pyruvate dehydrogenase kinase and lactate dehydrogenase was decreased whereas that of pyruvate dehydrogenase and cytosolic cytochrome c was upregulated with 3-BrPA administration. CONCLUSION This study demonstrates the reversal of PH by 3-BrPA is related to alteration in glycolysis and improved mitochondria function, indicating the "metabolic targeting" as a rational therapeutic strategy for PH.
Collapse
Affiliation(s)
- Yun-Long Zhang
- Department of Bioengineering, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Rui Zhang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi-Fan Shen
- Department of Bioengineering, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Kai-Yue Huang
- Department of Bioengineering, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Yang-Yang He
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Han Zhao
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Cheng Jing
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Pulmonary Vascular Medicine and FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
46
|
Jandl K, Thekkekara Puthenparampil H, Marsh LM, Hoffmann J, Wilhelm J, Veith C, Sinn K, Klepetko W, Olschewski H, Olschewski A, Brock M, Kwapiszewska G. Long non-coding RNAs influence the transcriptome in pulmonary arterial hypertension: the role of PAXIP1-AS1. J Pathol 2019; 247:357-370. [PMID: 30450722 PMCID: PMC6900182 DOI: 10.1002/path.5195] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/18/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022]
Abstract
In idiopathic pulmonary arterial hypertension (IPAH), global transcriptional changes induce a smooth muscle cell phenotype characterised by excessive proliferation, migration, and apoptosis resistance. Long non‐coding RNAs (lncRNAs) are key regulators of cellular function. Using a compartment‐specific transcriptional profiling approach, we sought to investigate the link between transcriptional reprogramming by lncRNAs and the maladaptive smooth muscle cell phenotype in IPAH. Transcriptional profiling of small remodelled arteries from 18 IPAH patients and 17 controls revealed global perturbations in metabolic, neuronal, proliferative, and immunological processes. We demonstrated an IPAH‐specific lncRNA expression profile and identified the lncRNA PAXIP1‐AS1 as highly abundant. Comparative transcriptomic analysis and functional assays revealed an intrinsic role for PAXIP1‐AS1 in orchestrating the hyperproliferative and migratory actions of IPAH smooth muscle cells. Further, we showed that PAXIP1‐AS1 mechanistically interferes with the focal adhesion axis via regulation of expression and phosphorylation of its downstream target paxillin. Overall, we show that changes in the lncRNA transcriptome contribute to the disease‐specific transcriptional landscape in IPAH. Our results suggest that lncRNAs, such as PAXIP1‐AS1, can modulate smooth muscle cell function by affecting multiple IPAH‐specific transcriptional programmes. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | | | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Christine Veith
- Excellence Cluster Cardio-Pulmonary System, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center, German Center for Lung Lung Research, Giessen, Germany
| | - Katharina Sinn
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, 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
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Otto Loewi Research Center, Chair of Physiology, Medical University of Graz, Graz, Austria
| | - Matthias Brock
- Division of Pulmonology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Otto Loewi Research Center, Chair of Physiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
47
|
Rafikov R, McBride ML, Zemskova M, Kurdyukov S, McClain N, Niihori M, Langlais PR, Rafikova O. Inositol monophosphatase 1 as a novel interacting partner of RAGE in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 316:L428-L444. [PMID: 30604625 DOI: 10.1152/ajplung.00393.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a lethal disease characterized by progressive pulmonary vascular remodeling. The receptor for advanced glycation end products (RAGE) plays an important role in PAH by promoting proliferation of pulmonary vascular cells. RAGE is also known to mediate activation of Akt signaling, although the particular molecular mechanism remains unknown. This study aimed to identify the interacting partner of RAGE that could facilitate RAGE-mediated Akt activation and vascular remodeling in PAH. The progressive angioproliferative PAH was induced in 24 female Sprague-Dawley rats ( n = 8/group) that were randomly assigned to develop PAH for 1, 2, or 5 wk [right ventricle systolic pressure (RVSP) 56.5 ± 3.2, 63.6 ± 1.6, and 111.1 ± 4.5 mmHg, respectively, vs. 22.9 ± 1.1 mmHg in controls]. PAH triggered early and late episodes of apoptosis in rat lungs accompanied by RAGE activation. Mass spectrometry analysis has identified IMPA1 as a novel PAH-specific interacting partner of RAGE. The proximity ligation assay (PLA) confirmed the formation of RAGE/IMPA1 complex in the pulmonary artery wall. Activation of IMPA1 in response to increased glucose 6-phosphate (G6P) is known to play a critical role in inositol synthesis and recycling. Indeed, we confirmed a threefold increase in G6P ( P = 0.0005) levels in lungs of PAH rats starting from week 1 that correlated with accumulation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3), membrane translocation of PI3K, and a threefold increase in membrane Akt levels ( P = 0.02) and Akt phosphorylation. We conclude that the formation of the newly discovered RAGE-IMPA1 complex could be responsible for the stimulation of inositol pathways and activation of Akt signaling in PAH.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Matthew L McBride
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Marina Zemskova
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Sergey Kurdyukov
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Nolan McClain
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Maki Niihori
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Paul R Langlais
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| | - Olga Rafikova
- Division of Endocrinology, Department of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
48
|
Abstract
The lung is often overlooked as a metabolically active organ, yet biochemical studies have long demonstrated that glucose utilization surpasses that of many other organs, including the heart, kidney, and brain. For most cells in the lung, energy consumption is relegated to performing common cellular tasks, like mRNA transcription and protein translation. However, certain lung cell populations engage in more specialized types of energy-consuming behaviors, such as the beating of cilia or the production of surfactant. While many extrapulmonary diseases are now linked to abnormalities in cellular metabolism, the pulmonary community has only recently embraced the concept of metabolic dysfunction as a driver of respiratory pathology. Herein, we provide an overview of the major metabolic pathways in the lung and discuss how cells sense and adapt to low-energy states. Moreover, we review some of the emerging evidence that links alterations in cellular metabolism to the pathobiology of several common respiratory diseases.
Collapse
Affiliation(s)
- Gang Liu
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA;
| |
Collapse
|
49
|
Hua C, Zhao J, Wang H, Chen F, Meng H, Chen L, Zhang Q, Yan J, Yuan L. Apple polyphenol relieves hypoxia-induced pulmonary arterial hypertension via pulmonary endothelium protection and smooth muscle relaxation: In vivo and in vitro studies. Biomed Pharmacother 2018; 107:937-944. [DOI: 10.1016/j.biopha.2018.08.080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/15/2018] [Accepted: 08/15/2018] [Indexed: 12/25/2022] Open
|
50
|
3-Bromopyruvate reverses hypoxia-induced pulmonary arterial hypertension through inhibiting glycolysis: In vitro and in vivo studies. Int J Cardiol 2018; 266:236-241. [DOI: 10.1016/j.ijcard.2018.03.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 02/12/2018] [Accepted: 03/21/2018] [Indexed: 01/14/2023]
|