1
|
Chen M, Li H, Li Y, Luo Y, He Y, Shui X, Lei W. Glycolysis modulation: New therapeutic strategies to improve pulmonary hypertension (Review). Int J Mol Med 2024; 54:115. [PMID: 39422043 PMCID: PMC11518579 DOI: 10.3892/ijmm.2024.5439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive life‑threatening cardiopulmonary vascular disease involving various pathological mechanisms, including hypoxia, cellular metabolism, inflammation, abnormal proliferation and apoptosis. Specifically, metabolism has attracted the most attention. Glucose metabolism is essential to maintain the cardiopulmonary vascular function. However, once exposed to a noxious stimulus, intracellular glucose metabolism changes or switches to an alternative pathway more suitable for adaptation, which is known as metabolic reprogramming. By promoting the switch from oxidative phosphorylation to glycolysis, cellular metabolic reprogramming plays an important role in PH development. Suppression of glucose oxidation and secondary upregulation of glycolysis are responsible for various features of PH, including the proliferation and apoptosis resistance of pulmonary artery endothelial and smooth muscle cells. In the present review, the roles and importance of the glucose metabolism shift were discussed to aid in the development of new treatment approaches for PH.
Collapse
Affiliation(s)
- Meihong Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hui Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yun Li
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yangui Luo
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xiaorong Shui
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
2
|
Hough RF, Alvira CM, Bastarache JA, Erzurum SC, Kuebler WM, Schmidt EP, Shimoda LA, Abman SH, Alvarez DF, Belvitch P, Bhattacharya J, Birukov KG, Chan SY, Cornfield DN, Dudek SM, Garcia JGN, Harrington EO, Hsia CCW, Islam MN, Jonigk DD, Kalinichenko VV, Kolb TM, Lee JY, Mammoto A, Mehta D, Rounds S, Schupp JC, Shaver CM, Suresh K, Tambe DT, Ventetuolo CE, Yoder MC, Stevens T, Damarla M. Studying the Pulmonary Endothelium in Health and Disease: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 71:388-406. [PMID: 39189891 PMCID: PMC11450313 DOI: 10.1165/rcmb.2024-0330st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Lung endothelium resides at the interface between the circulation and the underlying tissue, where it senses biochemical and mechanical properties of both the blood as it flows through the vascular circuit and the vessel wall. The endothelium performs the bidirectional signaling between the blood and tissue compartments that is necessary to maintain homeostasis while physically separating both, facilitating a tightly regulated exchange of water, solutes, cells, and signals. Disruption in endothelial function contributes to vascular disease, which can manifest in discrete vascular locations along the artery-to-capillary-to-vein axis. Although our understanding of mechanisms that contribute to endothelial cell injury and repair in acute and chronic vascular disease have advanced, pathophysiological mechanisms that underlie site-specific vascular disease remain incompletely understood. In an effort to improve the translatability of mechanistic studies of the endothelium, the American Thoracic Society convened a workshop to optimize rigor, reproducibility, and translation of discovery to advance our understanding of endothelial cell function in health and disease.
Collapse
|
3
|
Wawrzyniak R, Grešner P, Lewicka E, Macioszek S, Furga A, Zieba B, J. Markuszewski M, Da̧browska-Kugacka A. Metabolomics Meets Clinics: A Multivariate Analysis of Plasma and Urine Metabolic Signatures in Pulmonary Arterial Hypertension. J Proteome Res 2024; 23:2795-2804. [PMID: 37827514 PMCID: PMC11302416 DOI: 10.1021/acs.jproteome.3c00255] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 10/14/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe, multifactorial, and frequently misdiagnosed disorder. The aim of this observational study was to compare the plasma and urine metabolomic profiles of PAH patients and healthy control subjects. Plasma and urine metabolomic profiles were analyzed using the GC-MS technique. Correlations between metabolite levels and clinical parameters among PAH patients, as well as the between-group differences, were evaluated. The linear discriminant analysis model, which allows for subject classification in terms of PAH with the highest possible precision, was developed and proposed. Plasma pyruvic acid, cholesterol, threonine, urine 3-(3-hydroxyphenyl)-3-hydroxypropanoic acid, butyric acid, 1,2-benzenediol, glucoheptonic acid, and 2-oxo-glutaric acid were found to build a relatively accurate classification model for PAH patients. The model reached an accuracy of 91% and significantly improved subject classification (OR = 119 [95% CI: 20.3-698.3], p < 0.0001). Five metabolites were detected in urine that provide easily available and noninvasive tests as compared to right heart catheterization. The selected panel of metabolites has potential for early recognition of patients with dyspnea and faster referral to a reference center.
Collapse
Affiliation(s)
- Renata Wawrzyniak
- Department
of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Peter Grešner
- Laboratory
of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University
of Gdańsk, Dȩbinki
1, 80-211 Gdańsk, Poland
| | - Ewa Lewicka
- Department
of Cardiology and Electrotherapy, Medical
University of Gdansk, Debinki 7, 80-210 Gdańsk, Poland
| | - Szymon Macioszek
- Department
of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Artur Furga
- Department
of General, Endocrine and Transplant Surgery, Invasive Medicine Center, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Bożena Zieba
- First
Department of Cardiology, Medical University
of Gdansk, Smoluchowskiego
17, 80-214 Gdańsk, Poland
| | - Michał J. Markuszewski
- Department
of Biopharmaceutics and Pharmacodynamics, Medical University of Gdańsk, Hallera 107, 80-416 Gdańsk, Poland
| | - Alicja Da̧browska-Kugacka
- Department
of Cardiology and Electrotherapy, Medical
University of Gdansk, Debinki 7, 80-210 Gdańsk, Poland
| |
Collapse
|
4
|
Bordan Z, Batori R, Haigh S, Li X, Meadows ML, Brown ZL, West MA, Dong K, Han W, Su Y, Ma Q, Huo Y, Zhou J, Abdelbary M, Sullivan J, Weintraub NL, Stepp DW, Chen F, Barman SA, Fulton DJR. PDZ-Binding Kinase, a Novel Regulator of Vascular Remodeling in Pulmonary Arterial Hypertension. Circulation 2024; 150:393-410. [PMID: 38682326 PMCID: PMC11650665 DOI: 10.1161/circulationaha.123.067095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/04/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is high blood pressure in the lungs that originates from structural changes in small resistance arteries. A defining feature of PAH is the inappropriate remodeling of pulmonary arteries (PA) leading to right ventricle failure and death. Although treatment of PAH has improved, the long-term prognosis for patients remains poor, and more effective targets are needed. METHODS Gene expression was analyzed by microarray, RNA sequencing, quantitative polymerase chain reaction, Western blotting, and immunostaining of lung and isolated PA in multiple mouse and rat models of pulmonary hypertension (PH) and human PAH. PH was assessed by digital ultrasound, hemodynamic measurements, and morphometry. RESULTS Microarray analysis of the transcriptome of hypertensive rat PA identified a novel candidate, PBK (PDZ-binding kinase), that was upregulated in multiple models and species including humans. PBK is a serine/threonine kinase with important roles in cell proliferation that is minimally expressed in normal tissues but significantly increased in highly proliferative tissues. PBK was robustly upregulated in the medial layer of PA, where it overlaps with markers of smooth muscle cells. Gain-of-function approaches show that active forms of PBK increase PA smooth muscle cell proliferation, whereas silencing PBK, dominant negative PBK, and pharmacological inhibitors of PBK all reduce proliferation. Pharmacological inhibitors of PBK were effective in PH reversal strategies in both mouse and rat models, providing translational significance. In a complementary genetic approach, PBK was knocked out in rats using CRISPR/Cas9 editing, and loss of PBK prevented the development of PH. We found that PBK bound to PRC1 (protein regulator of cytokinesis 1) in PA smooth muscle cells and that multiple genes involved in cytokinesis were upregulated in experimental models of PH and human PAH. Active PBK increased PRC1 phosphorylation and supported cytokinesis in PA smooth muscle cells, whereas silencing or dominant negative PBK reduced cytokinesis and the number of cells in the G2/M phase of the cell cycle. CONCLUSIONS PBK is a newly described target for PAH that is upregulated in proliferating PA smooth muscle cells, where it contributes to proliferation through changes in cytokinesis and cell cycle dynamics to promote medial thickening, fibrosis, increased PA resistance, elevated right ventricular systolic pressure, right ventricular remodeling, and PH.
Collapse
Affiliation(s)
- Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Robert Batori
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Stephen Haigh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Xueyi Li
- Departments of Ophthalmology and Medicine, Stanford University School of Medicine, Palo Alto, California, United States
| | - Mary Louise Meadows
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zach L Brown
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Madison A. West
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kunzhe Dong
- Immunology Center of Georgia, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Weihong Han
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Qian Ma
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mahmoud Abdelbary
- School of Medicine, Oregon Health & Science University Portland, OR, United States
| | - Jennifer Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Neal. L. Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Scott A. Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
5
|
Zhang H, Chen L, Li J, Sun J, Zhao Q, Wang S, Li G. STAT3 phosphorylation at Tyr705 affects DRP1 (dynamin-related protein 1) controlled-mitochondrial fission during the development of apoptotic-resistance in pulmonary arterial endothelial cells. Genes Genomics 2024; 46:751-762. [PMID: 38733520 PMCID: PMC11208226 DOI: 10.1007/s13258-024-01522-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 05/01/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The apoptosis-resistant pulmonary arterial endothelial cells (PAECs) are known to be major players in the pulmonary remodeling of pulmonary arterial hypertension (PAH) and exhibit an abnormal metabolic profile with mitochondrial dysfunction. Mitochondrial fission has been shown to regulate the apoptosis of several cell types, but this is largely unexplored in the PAECs. OBJECTIVE The roles of mitochondrial fission control by Dynamin related protein-1 (DRP1) in the development of PAECs apoptosis suppression were investigated in present study and the potential mechanisms behind this were furtherly explored. METHODS The mitochondrial morphology was investigated in PAECs from PAH rats with the pulmonary plexiform lesions, and the relations of it with DRP1 expression and apoptosis were furtherly identified in apoptosis-resistant PAECs induced by hypoxia. PAECs were isolated from rats with severe PAH and from normal subjects, the apoptotic-resistant PAECs were induced by hypoxia. DRP1 gene knockdown was achieved via DRP1-siRNA, DRP1 and STAT3 phosphorylation were blocked using its inhibitors, respectively. Apoptosis was analyzed by flow cytometry, and mitochondrial morphology was investigated by transmission electron microscope and confocal microscopy. RESULTS The PAECs isolated from PAH rats with the pulmonary plexiform-like lesions and displayed lower apoptotic rate with increased DRP1 expression and mitochondrial fragmentation. In addition, similar observations were achieved in apoptosis-resistant PAECs induced by hypoxia. Targeting DRP1 using siRNA and pharmacologic blockade prevented the mitochondrial fission and subsequent apoptotic resistance in PAECs under hypoxia. Mechanistically, STAT3 phosphorylation at Tyr705 was shown to be activated in both PAH and hypoxia-treated PAECs, leading to the regulation of DRP1 expression. Of importance, targeting STAT3Tyr705 phosphorylation prevented DRP1 disruption on apoptosis in PAECs under hypoxia. CONCLUSIONS These data indicated that STAT3 phosphorylation at Tyr705 impacted DRP1-controlled mitochondrial fission during the development of apoptosis-resistance in PAECs, suggesting mitochondrial dynamics may represent a therapeutic target for PAH.
Collapse
Affiliation(s)
- Han Zhang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Li Chen
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiachen Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Jiashu Sun
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Qixu Zhao
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Sheng Wang
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China
| | - Gang Li
- Beijing Anzhen Hospital, Capital Medical University, 2 Anzhen Road, Beijing, 100029, China.
| |
Collapse
|
6
|
Luo Y, Qi X, Zhang Z, Zhang J, Li B, Shu T, Li X, Hu H, Li J, Tang Q, Zhou Y, Wang M, Fan T, Guo W, Liu Y, Zhang J, Pang J, Yang P, Gao R, Chen W, Yan C, Xing Y, Du W, Wang J, Wang C. Inactivation of Malic Enzyme 1 in Endothelial Cells Alleviates Pulmonary Hypertension. Circulation 2024; 149:1354-1371. [PMID: 38314588 DOI: 10.1161/circulationaha.123.067579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive cardiopulmonary disease with a high mortality rate. Although growing evidence has revealed the importance of dysregulated energetic metabolism in the pathogenesis of PH, the underlying cellular and molecular mechanisms are not fully understood. In this study, we focused on ME1 (malic enzyme 1), a key enzyme linking glycolysis to the tricarboxylic acid cycle. We aimed to determine the role and mechanistic action of ME1 in PH. METHODS Global and endothelial-specific ME1 knockout mice were used to investigate the role of ME1 in hypoxia- and SU5416/hypoxia (SuHx)-induced PH. Small hairpin RNA and ME1 enzymatic inhibitor (ME1*) were used to study the mechanism of ME1 in pulmonary artery endothelial cells. Downstream key metabolic pathways and mediators of ME1 were identified by metabolomics analysis in vivo and ME1-mediated energetic alterations were examined by Seahorse metabolic analysis in vitro. The pharmacological effect of ME1* on PH treatment was evaluated in PH animal models induced by SuHx. RESULTS We found that ME1 protein level and enzymatic activity were highly elevated in lung tissues of patients and mice with PH, primarily in vascular endothelial cells. Global knockout of ME1 protected mice from developing hypoxia- or SuHx-induced PH. Endothelial-specific ME1 deletion similarly attenuated pulmonary vascular remodeling and PH development in mice, suggesting a critical role of endothelial ME1 in PH. Mechanistic studies revealed that ME1 inhibition promoted downstream adenosine production and activated A2AR-mediated adenosine signaling, which leads to an increase in nitric oxide generation and a decrease in proinflammatory molecule expression in endothelial cells. ME1 inhibition activated adenosine production in an ATP-dependent manner through regulating malate-aspartate NADH (nicotinamide adenine dinucleotide plus hydrogen) shuttle and thereby balancing oxidative phosphorylation and glycolysis. Pharmacological inactivation of ME1 attenuated the progression of PH in both preventive and therapeutic settings by promoting adenosine production in vivo. CONCLUSIONS Our findings indicate that ME1 upregulation in endothelial cells plays a causative role in PH development by negatively regulating adenosine production and subsequently dysregulating endothelial functions. Our findings also suggest that ME1 may represent as a novel pharmacological target for upregulating protective adenosine signaling in PH therapy.
Collapse
Affiliation(s)
- Ya Luo
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.L.)
| | - Xianmei Qi
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Zhenxi Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jiawei Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Bolun Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ting Shu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Xiaona Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Huiyuan Hu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jinqiu Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Qihao Tang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Yitian Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Mingyao Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
| | - Tianfei Fan
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjun Guo
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ying Liu
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China (J.Z.)
| | - Junling Pang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ran Gao
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenhui Chen
- Department of Lung Transplantation, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China (W.C.)
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (C.Y.)
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
- Chinese Academy of Engineering, Beijing, China (C.W.)
| |
Collapse
|
7
|
Poojary G, Morris N, Joshi MB, Babu AS. Role of Exercise in Pulmonary Hypertension: Evidence from Bench to Bedside. Pulse (Basel) 2024; 12:66-75. [PMID: 39022559 PMCID: PMC11249447 DOI: 10.1159/000539537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/13/2024] [Indexed: 07/20/2024] Open
Abstract
Background Pulmonary hypertension (PH) is a debilitating condition characterized by elevated pulmonary arterial pressure and progressive vascular remodelling, leading to exercise intolerance. The progression of PAH is regulated at a cellular and molecular level which influences various physiological processes. Exercise plays an important role in improving function in PH. Although the signalling pathways that regulate cardio-protection through exercise have not been fully understood, the positive impact of exercise on the various physiological systems is well established. Summary Exercise has emerged as a potential adjunctive therapy for PH, with growing evidence supporting its beneficial effects on various aspects of the disease pathophysiology. This review highlights the contributions of cellular and molecular pathways and physiological processes to exercise intolerance. Preclinical studies have provided insight into the mechanisms underlying exercise-induced improvements in PH which are modulated through improvements in endothelial function, inflammation, oxidative stress, and mitochondrial function. Along with preclinical studies, various clinical studies have demonstrated that exercise training can lead to significant improvements in exercise capacity, haemodynamics, quality of life, and functional status. Moreover, exercise interventions have been shown to improve skeletal muscle function and enhance pulmonary vascular remodelling, contributing to overall disease management. Further research efforts aimed at better understanding the role of exercise in PH pathophysiology, and refining exercise interventions are warranted to realize its full potential in the management of this complex disease. Key Messages Despite the promising benefits of exercise in PH, several challenges remain, including the optimal intensity, duration, and type of exercise training, as well as patient selection criteria and long-term adherence. Additionally, the mechanisms underlying the observed improvements require further elucidation to optimize exercise protocols and personalize treatment strategies. Nonetheless, exercise represents a promising therapeutic approach that can complement existing pharmacological therapies and improve outcomes in PH patients.
Collapse
Affiliation(s)
- Ganesha Poojary
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Norman Morris
- School of Health Sciences and Social Work, Griffith University, Gold Coast Campus, Southport, QLD, Australia
- Allied Health Research Collaborative, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Manjunath B. Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Abraham Samuel Babu
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, India
- Department of Cardiology, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
8
|
Singh N, Eickhoff C, Garcia-Agundez A, Bertone P, Paudel SS, Tambe DT, Litzky LA, Cox-Flaherty K, Klinger JR, Monaghan SF, Mullin CJ, Pereira M, Walsh T, Whittenhall M, Stevens T, Harrington EO, Ventetuolo CE. Transcriptional profiles of pulmonary artery endothelial cells in pulmonary hypertension. Sci Rep 2023; 13:22534. [PMID: 38110438 PMCID: PMC10728171 DOI: 10.1038/s41598-023-48077-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by endothelial cell (EC) dysfunction. There are no data from living patients to inform whether differential gene expression of pulmonary artery ECs (PAECs) can discern disease subtypes, progression and pathogenesis. We aimed to further validate our previously described method to propagate ECs from right heart catheter (RHC) balloon tips and to perform additional PAEC phenotyping. We performed bulk RNA sequencing of PAECs from RHC balloons. Using unsupervised dimensionality reduction and clustering we compared transcriptional signatures from PAH to controls and other forms of pulmonary hypertension. Select PAEC samples underwent single cell and population growth characterization and anoikis quantification. Fifty-four specimens were analyzed from 49 subjects. The transcriptome appeared stable over limited passages. Six genes involved in sex steroid signaling, metabolism, and oncogenesis were significantly upregulated in PAH subjects as compared to controls. Genes regulating BMP and Wnt signaling, oxidative stress and cellular metabolism were differentially expressed in PAH subjects. Changes in gene expression tracked with clinical events in PAH subjects with serial samples over time. Functional assays demonstrated enhanced replication competency and anoikis resistance. Our findings recapitulate fundamental biological processes of PAH and provide new evidence of a cancer-like phenotype in ECs from the central vasculature of PAH patients. This "cell biopsy" method may provide insight into patient and lung EC heterogeneity to advance precision medicine approaches in PAH.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Carsten Eickhoff
- Department of Computer Science, Brown University, Providence, RI, USA
| | | | - Paul Bertone
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Sunita S Paudel
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Dhananjay T Tambe
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Department of Mechanical Aerospace and Biomedical Engineering, College of Engineering, University of South Alabama, Mobile, AL, USA
| | - Leslie A Litzky
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - James R Klinger
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Sean F Monaghan
- Department of Surgery, Alpert Medical School of Brown University, Providence, RI, USA
| | - Christopher J Mullin
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | - Mary Whittenhall
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Elizabeth O Harrington
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Corey E Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Health Services, Policy and Practice, Brown University, Providence, RI, USA.
| |
Collapse
|
9
|
Zhao C, Le X, Li M, Hu Y, Li X, Chen Z, Hu G, Hu L, Li Q. Inhibition of Hsp110-STAT3 interaction in endothelial cells alleviates vascular remodeling in hypoxic pulmonary arterial Hypertension model. Respir Res 2023; 24:289. [PMID: 37978368 PMCID: PMC10655391 DOI: 10.1186/s12931-023-02600-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive and devastating disease characterized by pulmonary vascular remodeling which is associated with the malignant phenotypes of pulmonary vascular cells. Recently, the effects of heat shock protein 110 (Hsp110) in human arterial smooth muscle cells were reported. However, the underlying roles and mechanisms of Hsp110 in human pulmonary arterial endothelial cells (HPAECs) that was disordered firstly at the early stage of PAH remain unknown. METHODS In this research, the expression of Hsp110 in PAH human patients and rat models was investigated, and the Hsp110 localization was determined both in vivo and in vitro. The roles and mechanism of elevated Hsp110 in excessive cell proliferation and migration of HPAECs were assessed respectively exposed to hypoxia. Small molecule inhibitors targeting Hsp110-STAT3 interaction were screened via fluorescence polarization, anti-aggregation and western blot assays. Moreover, the effects of compound 6 on HPAECs abnormal phenotypes in vitro and pulmonary vascular remodeling of hypoxia-indued PAH rats in vivo by interrupting Hsp110-STAT3 interaction were evaluated. RESULTS Our studies demonstrated that Hsp110 expression was increased in the serum of patients with PAH, as well as in the lungs and pulmonary arteries of PAH rats, when compared to their respective healthy subjects. Moreover, Hsp110 levels were significantly elevated in HPAECs under hypoxia and mediated its aberrant phenotypes. Furthermore, boosted Hsp110-STAT3 interaction resulted in abnormal proliferation and migration via elevating p-STAT3 and c-Myc in HPAECs. Notably, we successfully identified compound 6 as potent Hsp110-STAT3 interaction inhibitor, which effectively inhibited HPAECs proliferation and migration, and significantly ameliorated right heart hypertrophy and vascular remodeling of rats with PAH. CONCLUSIONS Our studies suggest that elevated Hsp110 plays a vital role in HPAECs and inhibition of the Hsp110-STAT3 interaction is a novel strategy for improving vascular remodeling. In addition, compound 6 could serve as a promising lead compound for developing first-in-class drugs against PAH.
Collapse
Affiliation(s)
- Congke Zhao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Xiangyang Le
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Mengqi Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanbo Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China.
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
- Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China.
- Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Yi D, Liu B, Ding H, Li S, Li R, Pan J, Ramirez K, Xia X, Kala M, Ye Q, Lee WH, Frye RE, Wang T, Zhao Y, Knox KS, Glembotski CC, Fallon MB, Dai Z. E2F1 Mediates SOX17 Deficiency-Induced Pulmonary Hypertension. Hypertension 2023; 80:2357-2371. [PMID: 37737027 PMCID: PMC10591929 DOI: 10.1161/hypertensionaha.123.21241] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Rare genetic variants and genetic variation at loci in an enhancer in SOX17 (SRY-box transcription factor 17) are identified in patients with idiopathic pulmonary arterial hypertension (PAH) and PAH with congenital heart disease. However, the exact role of genetic variants or mutations in SOX17 in PAH pathogenesis has not been reported. METHODS SOX17 expression was evaluated in the lungs and pulmonary endothelial cells (ECs) of patients with idiopathic PAH. Mice with Tie2Cre-mediated Sox17 knockdown and EC-specific Sox17 deletion were generated to determine the role of SOX17 deficiency in the pathogenesis of PAH. Human pulmonary ECs were cultured to understand the role of SOX17 deficiency. Single-cell RNA sequencing, RNA-sequencing analysis, and luciferase assay were performed to understand the underlying molecular mechanisms of SOX17 deficiency-induced PAH. E2F1 (E2F transcription factor 1) inhibitor HLM006474 was used in EC-specific Sox17 mice. RESULTS SOX17 expression was downregulated in the lung and pulmonary ECs from patients with idiopathic PAH. Mice with Tie2Cre-mediated Sox17 knockdown and EC-specific Sox17 deletion induced spontaneously mild pulmonary hypertension. Loss of endothelial Sox17 in EC exacerbated hypoxia-induced pulmonary hypertension in mice. Loss of SOX17 in lung ECs induced endothelial dysfunctions including upregulation of cell cycle programming, proliferative and antiapoptotic phenotypes, augmentation of paracrine effect on pulmonary arterial smooth muscle cells, impaired cellular junction, and BMP (bone morphogenetic protein) signaling. E2F1 signaling was shown to mediate the SOX17 deficiency-induced EC dysfunction. Pharmacological inhibition of E2F1 in Sox17 EC-deficient mice attenuated pulmonary hypertension development. CONCLUSIONS Our study demonstrated that endothelial SOX17 deficiency induces pulmonary hypertension through E2F1. Thus, targeting E2F1 signaling represents a promising approach in patients with PAH.
Collapse
Affiliation(s)
- Dan Yi
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Bin Liu
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Hongxu Ding
- Department of Pharmacy Practice & Science, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Shuai Li
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Rebecca Li
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Jiakai Pan
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Karina Ramirez
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Xiaomei Xia
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Mrinalini Kala
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Qinmao Ye
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Won Hee Lee
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | | | - Ting Wang
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Environmental Health Science and Center of Translational Science, Florida International University, Port Saint Lucie, Florida, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kenneth S. Knox
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Christopher C. Glembotski
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Michael B. Fallon
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- Translational Cardiovascular Research Center, College of Medicine-Phoenix, University of Arizona, Phoenix, Arizona, USA
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
- Sarver Heart Center, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
11
|
Lu G, Du R, Liu Y, Zhang S, Li J, Pei J. RGS5 as a Biomarker of Pericytes, Involvement in Vascular Remodeling and Pulmonary Arterial Hypertension. Vasc Health Risk Manag 2023; 19:673-688. [PMID: 37881333 PMCID: PMC10596204 DOI: 10.2147/vhrm.s429535] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by a sustained rise in mean pulmonary artery pressure. Pulmonary vascular remodeling serves an important role in PAH. Identifying a key driver gene to regulate vascular remodeling of the pulmonary microvasculature is critical for PAH management. Methods Differentially expressed genes were identified using the Gene Expression Omnibus (GEO) GSE117261, GSE48149, GSE113439, GSE53408 and GSE16947 datasets. A co-expression network was constructed using weighted gene co-expression network analysis. Novel and key signatures of PAH were screened using four algorithms, including weighted gene co-expression network analysis, GEO2R analysis, support vector machines recursive feature elimination and robust rank aggregation rank analysis. Regulator of G-protein signaling 5 (RGS5), a pro-apoptotic/anti-proliferative protein, which regulate arterial tone and blood pressure in vascular smooth muscle cells. The expression of RGS5 was determined using reverse transcription-quantitative PCR (RT-qPCR) in PAH and normal mice. The location of RGS5 and pericytes was detected using immunofluorescence. Results Compared with that in the normal group, RGS5 expression was upregulated in the PAH group based on GEO and RT-qPCR analyses. RGS5 expression in single cells was enriched in pericytes in single-cell RNA sequencing analysis. RGS5 co-localization with pericytes was detected in the pulmonary microvasculature of PAH. Conclusion RGS5 regulates vascular remodeling of the pulmonary microvasculature and the occurrence of PAH through pericytes, which has provided novel ideas and strategies regarding the occurrence and innovative treatment of PAH.
Collapse
Affiliation(s)
- Guofang Lu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710038, People’s Republic of China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| |
Collapse
|
12
|
Almazroue H, Jin Y, Nelin LD, Barba JC, Milton AD, Trittmann JK. Human pulmonary microvascular endothelial cell DDAH1-mediated nitric oxide production promotes pulmonary smooth muscle cell apoptosis in co-culture. Am J Physiol Lung Cell Mol Physiol 2023; 325:L360-L367. [PMID: 37431589 PMCID: PMC10639007 DOI: 10.1152/ajplung.00433.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/24/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease in preterm infants, and pulmonary hypertension (PH) develops in 25%-40% of patients with BPD, increasing morbidity and mortality. BPD-PH is characterized by vasoconstriction and vascular remodeling. Nitric oxide (NO) is a pulmonary vasodilator and apoptotic mediator made in the pulmonary endothelium by NO synthase (eNOS). Asymmetric dimethylarginine (ADMA) is an endogenous eNOS inhibitor, primarily metabolized by dimethylarginine dimethylaminohydrolase-1 (DDAH1). Our hypothesis is that DDAH1 knockdown in human pulmonary microvascular endothelial cells (hPMVEC) will result in lower NO production, decreased apoptosis, and greater proliferation of human pulmonary arterial smooth muscle cells (hPASMC), whereas DDAH1 overexpression will have the opposite effect. hPMVECs were transfected with small interfering RNA targeting DDAH1 (siDDAH1)/scramble or adenoviral vector containing DDAH1 (AdDDAH1)/AdGFP for 24 h and co-cultured for 24 h with hPASMC. Analyses included Western blot for cleaved and total caspase-3, caspase-8, caspase-9, β-actin; trypan blue exclusion for viable cell numbers; terminal deoxynucleotide transferase dUTP nick end labeling (TUNEL); and BrdU incorporation. Small interfering RNA targeting DDAH1 (siDDAH1) transfected into hPMVEC resulted in lower media nitrites, cleaved caspase-3 and caspase-8 protein expression, and TUNEL staining; and greater viable cell numbers and BrdU incorporation in co-cultured hPASMC. Adenoviral-mediated transfection of the DDAH1 gene (AdDDAH1) into hPMVEC resulted in greater cleaved caspase-3 and caspase-8 protein expression and lower viable cell numbers in co-cultured hPASMC. Partial recovery of hPASMC viable cell numbers after AdDDAH1-hPMVEC transfection was observed when media were treated with hemoglobin to sequester NO. In conclusion, hPMVEC-DDAH1-mediated NO production positively regulates hPASMC apoptosis, which may prevent/attenuate aberrant pulmonary vascular proliferation/remodeling in BPD-PH.NEW & NOTEWORTHY BPD-PH is characterized by vascular remodeling. NO is an apoptotic mediator made in the pulmonary endothelium by eNOS. ADMA is an endogenous eNOS inhibitor metabolized by DDAH1. EC-DDAH1 overexpression resulted in greater cleaved caspase-3 and caspase-8 protein expression and lower viable cell numbers in co-cultured SMC. After NO sequestration, SMC viable cell numbers partially recovered despite EC-DDAH1 overexpression. EC-DDAH1-mediated NO production positively regulates SMC apoptosis, which may prevent/attenuate aberrant pulmonary vascular proliferation/remodeling in BPD-PH.
Collapse
Affiliation(s)
- Hanadi Almazroue
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - John C Barba
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - Avante D Milton
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Neonatology, Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, Ohio, United States
| |
Collapse
|
13
|
Zhou X, Jiang Y, Wang Y, Fan L, Zhu Y, Chen Y, Wang Y, Zhu Y, Wang H, Pan Z, Li Z, Zhu X, Ren R, Ge Z, Lai D, Lai EY, Chen T, Wang K, Liang P, Qin L, Liu C, Qiu C, Simons M, Yu L. Endothelial FIS1 DeSUMOylation Protects Against Hypoxic Pulmonary Hypertension. Circ Res 2023; 133:508-531. [PMID: 37589160 DOI: 10.1161/circresaha.122.321200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Hypoxia is a major cause and promoter of pulmonary hypertension (PH), a representative vascular remodeling disease with poor prognosis and high mortality. However, the mechanism underlying how pulmonary arterial system responds to hypoxic stress during PH remains unclear. Endothelial mitochondria are considered signaling organelles on oxygen tension. Results from previous clinical research and our studies suggested a potential role of posttranslational SUMOylation (small ubiquitin-like modifier modification) in endothelial mitochondria in hypoxia-related vasculopathy. METHODS Chronic hypoxia mouse model and Sugen/hypoxia rat model were employed as PH animal models. Mitochondrial morphology and subcellular structure were determined by transmission electron and immunofluorescent microscopies. Mitochondrial metabolism was determined by mitochondrial oxygen consumption rate and extracellular acidification rate. SUMOylation and protein interaction were determined by immunoprecipitation. RESULTS The involvement of SENP1 (sentrin-specific protease 1)-mediated SUMOylation in mitochondrial remodeling in the pulmonary endothelium was identified in clinical specimens of hypoxia-related PH and was verified in human pulmonary artery endothelial cells under hypoxia. Further analyses in clinical specimens, hypoxic rat and mouse PH models, and human pulmonary artery endothelial cells and human embryonic stem cell-derived endothelial cells revealed that short-term hypoxia-induced SENP1 translocation to endothelial mitochondria to regulate deSUMOylation (the reversible process of SUMOylation) of mitochondrial fission protein FIS1 (mitochondrial fission 1), which facilitated FIS1 assembling with fusion protein MFN2 (mitofusin 2) and mitochondrial gatekeeper VDAC1 (voltage-dependent anion channel 1), and the membrane tethering activity of MFN2 by enhancing its oligomerization. Consequently, FIS1 deSUMOylation maintained the mitochondrial integrity and endoplasmic reticulum-mitochondria calcium communication across mitochondrial-associated membranes, subsequently preserving pulmonary endothelial function and vascular homeostasis. In contrast, prolonged hypoxia disabled the FIS1 deSUMOylation by diminishing the availability of SENP1 in mitochondria via inducing miR (micro RNA)-138 and consequently resulted in mitochondrial dysfunction and metabolic reprogramming in pulmonary endothelium. Functionally, introduction of viral-packaged deSUMOylated FIS1 within pulmonary endothelium in mice improved pulmonary endothelial dysfunction and hypoxic PH development, while knock-in of SUMO (small ubiquitin-like modifier)-conjugated FIS1 in mice exaggerated the diseased cellular and tissue phenotypes. CONCLUSIONS By maintaining endothelial mitochondrial homeostasis, deSUMOylation of FIS1 adaptively preserves pulmonary endothelial function against hypoxic stress and consequently protects against PH. The FIS1 deSUMOylation-SUMOylation transition in pulmonary endothelium is an intrinsic pathogenesis of hypoxic PH.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yuanqing Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yuewen Wang
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, China (Yuewen Wang)
| | - Linge Fan
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yunhui Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Yefeng Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yiran Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Yingyi Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Hongkun Wang
- Institute of Translational Medicine (H.W., P.L.), Hangzhou, China
| | - Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China (Z.P., K.W.)
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Z.P., K.W.)
| | - Zhoubin Li
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Xiaolong Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
| | - Ruizhe Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
| | - Zhen Ge
- School of Pharmaceutical Sciences, Hangzhou Medical College, Zhejiang, China (Z.G.)
| | - Dongwu Lai
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
| | - En Yin Lai
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Ting Chen
- The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China (Z.L., E.Y.-L., T.C.)
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China (Z.P., K.W.)
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Z.P., K.W.)
| | - Ping Liang
- Institute of Translational Medicine (H.W., P.L.), Hangzhou, China
| | - Lingfeng Qin
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Cuiqing Liu
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China (C.L.)
| | - Cong Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
- Cancer Center, Zhejiang University (C.Q., L.Y.), Hangzhou, China
| | - Michael Simons
- Cardiovascular Research Center, Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, CT (X. Zhu, L.Q., M.S.)
| | - Luyang Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, X. Zhu, R.R., D.L., C.Q., L.Y.), Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute (X. Zhou, Y.J., L.F., Yunhui Zhu, Y.C., Yiran Wang, Yingyi Zhu, R.R., C.Q., L.Y.), Hangzhou, China
- Cancer Center, Zhejiang University (C.Q., L.Y.), Hangzhou, China
| |
Collapse
|
14
|
Sun Y, Liu S, Chen C, Yang S, Pei G, Lin M, Wang T, Long J, Yan Q, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N, Yang Y. The mechanism of programmed death and endoplasmic reticulum stress in pulmonary hypertension. Cell Death Discov 2023; 9:78. [PMID: 36841823 PMCID: PMC9968278 DOI: 10.1038/s41420-023-01373-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/27/2023] Open
Abstract
Pulmonary hypertension (PH) was a cardiovascular disease with high morbidity and mortality. PH was a chronic disease with complicated pathogenesis and uncontrollable factors. PH was divided into five groups according to its pathogenesis and clinical manifestations. Although the treatment and diagnosis of PH has made great progress in the past ten years. However, the diagnosis and prognosis of the PAH had a great contrast, which was not conducive to the diagnosis and treatment of PH. If not treated properly, it will lead to right ventricular failure or even death. Therefore, it was necessary to explore the pathogenesis of PH. The problem we urgently need to solve was to find and develop drugs for the treatment of PH. We reviewed the PH articles in the past 10 years or so as well as systematically summarized the recent advance. We summarized the latest research on the key regulatory factors (pyroptosis, apoptosis, necroptosis, ferroptosis, and endoplasmic reticulum stress) involved in PH. To provide theoretical basis and basis for finding new therapeutic targets and research directions of PH.
Collapse
Affiliation(s)
- Yang Sun
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal & Child Health Care, Changsha, P. R. China
| | - Chen Chen
- grid.412643.60000 0004 1757 2902Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, P. R. China
| | - Songwei Yang
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Gang Pei
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Meiyu Lin
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Ting Wang
- grid.501248.aDepartment of Rehabilitation Medicine, Zhuzhou Central Hospital, Zhuzhou, P. R. China
| | - Junpeng Long
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Qian Yan
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Jiao Yao
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yuting Lin
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Fan Yi
- grid.411615.60000 0000 9938 1755Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, P. R. China
| | - Lei Meng
- grid.488482.a0000 0004 1765 5169Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China
| | - Yong Tan
- Department of nephrology, Xiangtan Central Hospital, Xiangtan, P. R. China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China. .,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China.
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, P. R. China.
| |
Collapse
|
15
|
Mao L, Yuan X, Su J, Ma Y, Li C, Chen H, Zhang F. Human Umbilical Vein Endothelial Cells Survive on the Ischemic TCA Cycle under Lethal Ischemic Conditions. J Proteome Res 2022; 21:2385-2396. [PMID: 36074008 PMCID: PMC9552233 DOI: 10.1021/acs.jproteome.2c00255] [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] [Indexed: 11/28/2022]
Abstract
![]()
It is generally believed that vascular endothelial cells
(VECs)
rely on glycolysis instead of the tricarboxylic acid (TCA) cycle under
both normoxic and hypoxic conditions. However, the metabolic pattern
of human umbilical vein endothelial cells (HUVECs) under extreme ischemia
(hypoxia and nutrient deprivation) needs to be elucidated. We initiated
a lethal ischemic model of HUVECs, performed proteomics and bioinformatics,
and verified the metabolic pattern shift of HUVECs. Ischemic HUVECs
displayed extensive aerobic respiration, including upregulation of
the TCA cycle and mitochondrial respiratory chain in mitochondria
and downregulation of glycolysis in cytoplasm. The TCA cycle was enhanced
while the cell viability was decreased through the citrate synthase
pathway when substrates of the TCA cycle (acetate and/or pyruvate)
were added and vice versa when inhibitors of the TCA cycle (palmitoyl-CoA
and/or avidin) were applied. The inconsistency of the TCA cycle level
and cell viability suggested that the extensive TCA cycle can keep
cells alive yet generate toxic substances that reduce cell viability.
The data revealed that HUVECs depend on “ischemic TCA cycle”
instead of glycolysis to keep cells alive under lethal ischemic conditions,
but consideration must be given to relieve cell injury.
Collapse
Affiliation(s)
- Lisha Mao
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Xiaoqi Yuan
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Junlei Su
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing 401147, China
| | - Yaping Ma
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Chaofan Li
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Hongying Chen
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Fugui Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
16
|
Lv C, Kang W, Liu S, Yang P, Nishina Y, Ge S, Bianco A, Ma B. Growth of ZIF-8 Nanoparticles In Situ on Graphene Oxide Nanosheets: A Multifunctional Nanoplatform for Combined Ion-Interference and Photothermal Therapy. ACS NANO 2022; 16:11428-11443. [PMID: 35816172 DOI: 10.1021/acsnano.2c05532] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The regulation of intracellular ions' overload to interrupt normal bioprocesses and cause cell death has been developed as an efficient strategy (named as ion-interference therapy/IIT) to treat cancer. In this study, we design a multifunctional nanoplatform (called BSArGO@ZIF-8 NSs) by in situ growth of metal organic framework nanoparticles (ZIF-8 NPs) onto the graphene oxide (GO) surface, subsequently reduced by ascorbic acid and modified by bovine serum albumin. This nanocomplex causes the intracellular overload of Zn2+, an increase of reactive oxygen species (ROS), and exerts a broad-spectrum lethality to different kinds of cancer cells. BSArGO@ZIF-8 NSs can promote cell apoptosis by initiating bim (a pro-apoptotic protein)-mediated mitochondrial apoptotic events, up-regulating PUMA/NOXA expression, and down-regulating the level of Bid/p53AIP1. Meanwhile, Zn2+ excess triggers cellular dysfunction and mitochondria damage by activating the autophagy signaling pathways and disturbing the intracellular environmental homeostasis. Combined with the photothermal effect of reduced GO (rGO), BSArGO@ZIF-8 NSs mediated ion-interference and photothermal combined therapy leads to effective apoptosis and inhibits cell proliferation and angiogenesis, bringing a higher efficacy in tumor suppression in vivo. This designed Zn-based multifunctional nanoplatform will allow promoting further the development of IIT and the corresponding combined cancer therapy strategy.
Collapse
Affiliation(s)
- Chunxu Lv
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
| | - Wenyan Kang
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
| | - Shuo Liu
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
| | - Pishan Yang
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
| | - Yuta Nishina
- Graduate School of Natural Science and Technology, Okayama University, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
- Research Core for Interdisciplinary Sciences, Okayama University, Tsushimanaka, Kita-ku, Okayama, 700-8530, Japan
| | - Shaohua Ge
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| | - Baojin Ma
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong 250012, China
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, Strasbourg, 67000, France
| |
Collapse
|
17
|
Predescu DN, Mokhlesi B, Predescu SA. The Impact of Sex Chromosomes in the Sexual Dimorphism of Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:582-594. [PMID: 35114193 PMCID: PMC8978209 DOI: 10.1016/j.ajpath.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/14/2021] [Accepted: 01/11/2022] [Indexed: 02/09/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a sex-biased disease with a poorly understood female prevalence. Emerging research suggests that nonhormonal factors, such as the XX or XY sex chromosome complement and sex bias in gene expression, may also lead to sex-based differences in PAH incidence, penetrance, and progression. Typically, one of females' two X chromosomes is epigenetically silenced to offer a gender-balanced gene expression. Recent data demonstrate that the long noncoding RNA X-inactive specific transcript, essential for X chromosome inactivation and dosage compensation of X-linked gene expression, shows elevated levels in female PAH lung specimens compared with controls. This molecular event leads to incomplete inactivation of the females' second X chromosome, abnormal expression of X-linked gene(s) involved in PAH pathophysiology, and a pulmonary artery endothelial cell (PAEC) proliferative phenotype. Moreover, the pathogenic proliferative p38 mitogen-activated protein kinase/ETS transcription factor ELK1 (Elk1)/cFos signaling is mechanistically linked to the sexually dimorphic proliferative response of PAECs in PAH. Apprehending the complicated relationship between long noncoding RNA X-inactive specific transcript and X-linked genes and how this relationship integrates into a sexually dimorphic proliferation of PAECs and PAH sex paradox remain challenging. We highlight herein new findings related to how the sex chromosome complement and sex-differentiated epigenetic mechanisms to control gene expression are decisive players in the sexual dimorphism of PAH. Pharmacologic interventions in the light of the newly elucidated mechanisms are discussed.
Collapse
Affiliation(s)
- Dan N Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois.
| | - Babak Mokhlesi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Sanda A Predescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
18
|
Gomes MT, Bai Y, Potje SR, Zhang L, Lockett AD, Machado RF. Signal Transduction during Metabolic and Inflammatory Reprogramming in Pulmonary Vascular Remodeling. Int J Mol Sci 2022; 23:2410. [PMID: 35269553 PMCID: PMC8910500 DOI: 10.3390/ijms23052410] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by (mal)adaptive remodeling of the pulmonary vasculature, which is associated with inflammation, fibrosis, thrombosis, and neovascularization. Vascular remodeling in PAH is associated with cellular metabolic and inflammatory reprogramming that induce profound endothelial and smooth muscle cell phenotypic changes. Multiple signaling pathways and regulatory loops act on metabolic and inflammatory mediators which influence cellular behavior and trigger pulmonary vascular remodeling in vivo. This review discusses the role of bioenergetic and inflammatory impairments in PAH development.
Collapse
Affiliation(s)
- Marta T. Gomes
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; (Y.B.); (S.R.P.); (A.D.L.)
| | - Yang Bai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; (Y.B.); (S.R.P.); (A.D.L.)
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Simone R. Potje
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; (Y.B.); (S.R.P.); (A.D.L.)
- Department of Biological Science, Minas Gerais State University (UEMG), Passos 37900-106, Brazil
| | - Lu Zhang
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China;
| | - Angelia D. Lockett
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; (Y.B.); (S.R.P.); (A.D.L.)
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; (Y.B.); (S.R.P.); (A.D.L.)
| |
Collapse
|
19
|
Liang S, Yegambaram M, Wang T, Wang J, Black SM, Tang H. Mitochondrial Metabolism, Redox, and Calcium Homeostasis in Pulmonary Arterial Hypertension. Biomedicines 2022; 10:biomedicines10020341. [PMID: 35203550 PMCID: PMC8961787 DOI: 10.3390/biomedicines10020341] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by elevated pulmonary arterial pressure due to increased pulmonary vascular resistance, secondary to sustained pulmonary vasoconstriction and excessive obliterative pulmonary vascular remodeling. Work over the last decade has led to the identification of a critical role for metabolic reprogramming in the PAH pathogenesis. It is becoming clear that in addition to its role in ATP generation, the mitochondrion is an important organelle that regulates complex and integrative metabolic- and signal transduction pathways. This review focuses on mitochondrial metabolism alterations that occur in deranged pulmonary vessels and the right ventricle, including abnormalities in glycolysis and glucose oxidation, fatty acid oxidation, glutaminolysis, redox homeostasis, as well as iron and calcium metabolism. Further understanding of these mitochondrial metabolic mechanisms could provide viable therapeutic approaches for PAH patients.
Collapse
Affiliation(s)
- Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Manivannan Yegambaram
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
| | - Ting Wang
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
| | - Stephen M. Black
- Center for Translational Science, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA; (M.Y.); (T.W.)
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Port St. Lucie, FL 34987, USA
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Port St. Lucie, FL 34987, USA
- Correspondence: (S.M.B.); (H.T.)
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (S.L.); (J.W.)
- Correspondence: (S.M.B.); (H.T.)
| |
Collapse
|
20
|
Thomas S, Manivannan S, Garg V, Lilly B. Single-Cell RNA Sequencing Reveals Novel Genes Regulated by Hypoxia in the Lung Vasculature. J Vasc Res 2022; 59:163-175. [PMID: 35294950 PMCID: PMC9117417 DOI: 10.1159/000522340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/25/2022] [Indexed: 11/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic progressive disease with significant morbidity and mortality. The disease is characterized by vascular remodeling that includes increased muscularization of distal blood vessels and vessel stiffening associated with changes in extracellular matrix deposition. In humans, chronic hypoxia causes PAH, and hypoxia-induced rodent models of PAH have been used for years to study the disease. With the development of single-cell RNA sequencing technology, it is now possible to examine hypoxia-dependent transcriptional changes in vivo at a cell-specific level. In this study, we used single-cell RNA sequencing to compare lungs from wild-type (Wt) mice exposed to hypoxia for 28 days to normoxia-treated control mice. We additionally examined mice deficient for Notch3, a smooth muscle-enriched gene linked to PAH. Data analysis revealed that hypoxia promoted cell number changes in immune and endothelial cell types in the lung, activated the innate immunity pathway, and resulted in specific changes in gene expression in vascular cells. Surprisingly, we found limited differences in lungs from mice deficient for Notch3 compared to Wt controls. These findings provide novel insight into the effects of chronic hypoxia exposure on gene expression and cell phenotypes in vivo and identify unique changes to cells of the vasculature.
Collapse
Affiliation(s)
- Shelby Thomas
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sathiyanarayanan Manivannan
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Vidu Garg
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
21
|
Research Progress on Pulmonary Arterial Hypertension and the Role of the Angiotensin Converting Enzyme 2-Angiotensin-(1-7)-Mas Axis in Pulmonary Arterial Hypertension. Cardiovasc Drugs Ther 2022; 36:363-370. [PMID: 33394361 PMCID: PMC7779643 DOI: 10.1007/s10557-020-07114-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2020] [Indexed: 01/31/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease with a complex aetiology and high mortality. Functional and structural changes in the small pulmonary arteries lead to elevated pulmonary arterial pressure, resulting in right heart failure. The pathobiology of PAH is not fully understood, and novel treatment targets in PAH are desperately needed. The renin-angiotensin system is critical for maintaining homeostasis of the cardiovascular system. The system consists of the angiotensin converting enzyme (ACE)-angiotensin (Ang) II-angiotensin type 1 receptor (AT1R) axis and the ACE2-Ang-(1-7)-Mas receptor axis. The former, the ACE-Ang II-AT1R axis, is involved in vasoconstrictive and hypertensive actions along with cardiac and vascular remodelling. The latter, the ACE2-Ang-(1-7)-Mas axis, generally mediates counterbalancing effects against those mediated by the ACE-Ang II-AT1R axis. Based on established functions, the ACE2-Ang-(1-7)-Mas axis may represent a novel target for the treatment of PAH. This review focuses on recent advances in pulmonary circulation science and the role of the ACE2-Ang-(1-7)-Mas axis in PAH.
Collapse
|
22
|
Wang Z, Chen J, Babicheva A, Jain PP, Rodriguez M, Ayon RJ, Ravellette KS, Wu L, Balistrieri F, Tang H, Wu X, Zhao T, Black SM, Desai AA, Garcia JGN, Sun X, Shyy JYJ, Valdez-Jasso D, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Endothelial upregulation of mechanosensitive channel Piezo1 in pulmonary hypertension. Am J Physiol Cell Physiol 2021; 321:C1010-C1027. [PMID: 34669509 PMCID: PMC8714987 DOI: 10.1152/ajpcell.00147.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 12/16/2022]
Abstract
Piezo is a mechanosensitive cation channel responsible for stretch-mediated Ca2+ and Na+ influx in multiple types of cells. Little is known about the functional role of Piezo1 in the lung vasculature and its potential pathogenic role in pulmonary arterial hypertension (PAH). Pulmonary arterial endothelial cells (PAECs) are constantly under mechanic stretch and shear stress that are sufficient to activate Piezo channels. Here, we report that Piezo1 is significantly upregulated in PAECs from patients with idiopathic PAH and animals with experimental pulmonary hypertension (PH) compared with normal controls. Membrane stretch by decreasing extracellular osmotic pressure or by cyclic stretch (18% CS) increases Ca2+-dependent phosphorylation (p) of AKT and ERK, and subsequently upregulates expression of Notch ligands, Jagged1/2 (Jag-1 and Jag-2), and Delta like-4 (DLL4) in PAECs. siRNA-mediated downregulation of Piezo1 significantly inhibited the stretch-mediated pAKT increase and Jag-1 upregulation, whereas downregulation of AKT by siRNA markedly attenuated the stretch-mediated Jag-1 upregulation in human PAECs. Furthermore, the mRNA and protein expression level of Piezo1 in the isolated pulmonary artery, which mainly contains pulmonary arterial smooth muscle cells (PASMCs), from animals with severe PH was also significantly higher than that from control animals. Intraperitoneal injection of a Piezo1 channel blocker, GsMTx4, ameliorated experimental PH in mice. Taken together, our study suggests that membrane stretch-mediated Ca2+ influx through Piezo1 is an important trigger for pAKT-mediated upregulation of Jag-1 in PAECs. Upregulation of the mechanosensitive channel Piezo1 and the resultant increase in the Notch ligands (Jag-1/2 and DLL4) in PAECs may play a critical pathogenic role in the development of pulmonary vascular remodeling in PAH and PH.
Collapse
Affiliation(s)
- Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Keeley S Ravellette
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Linda Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Francesca Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Haiyang Tang
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomin Wu
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Stephen M Black
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ankit A Desai
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
- Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Joe G N Garcia
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | | | - Ayako Makino
- Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
- Departments of Medicine and Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
23
|
Kuropatkina TA, Pankova NV, Medvedeva NA, Medvedev OS. Ubiquinol ameliorates endothelial dysfunction and increases expression of miRNA-34a in a rat model of pulmonary hypertension. RESEARCH RESULTS IN PHARMACOLOGY 2021. [DOI: 10.3897/rrpharmacology.7.67291] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Introduction: In this research, we evaluate the effect of intravenously administrated solubilized ubiquinol on 4-week monocrotalin-induced pulmonary hypertension (PH) in rats.
Materials and methods: To reproduce the model, some male Wistar rats were subcutaneously injected with alcohol solution of monocrotaline 60 mg/kg and the rest – with alcohol solution (Control). Those with monocrotaline (MCT) were divided into 3 groups. They underwent intravenous administration of 1% ubiquinol solution 30 mg/kg (MCT-Ubiquinol), the vehicle (MCT-Vehicle) and saline (MCT-saline) three times on days 7, 14 and 21, depending on the group. The hemodynamic parameters were measured in anesthetized rats on day 29. Right ventricle hypertrophy, pulmonary arteries reactivity and expression of miRNA-21 and miRNA-34a were estimated after euthanasia.
Results and discussion: All MCT-groups demonstrated an increase in right ventricle systolic pressure and hypertrophy in comparison with the control group. An increase in lung weight was shown in MCT-Vehicle and MCT-Saline; however, the MCT-Ubiquinol indicators did not differ from those of the Control. There was an increased vasodilatation response to acetylcholine at concentrations of 1*10-6M and 1*10-5M in MCT-Ubiquinol in contrast to the other two MCT-groups. A significantly lower level of expression of miRNA-34a was observed in MCT-Ubiquinol.
Conclusion: Our findings suggest that a triple ubiquinol injection influences pulmonary changes and endothelium-depended vasodilatation, which contributes to pulmonary vascular tone and reactivity. A decrease in miRNA-34a expression in MCT-Ubiquinol group demonstrates the ubiquinol anti-inflammatory properties.
Collapse
|
24
|
Single-cell transcriptomic profile of human pulmonary artery endothelial cells in health and pulmonary arterial hypertension. Sci Rep 2021; 11:14714. [PMID: 34282213 PMCID: PMC8289993 DOI: 10.1038/s41598-021-94163-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/16/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an insidious disease characterized by severe remodeling of the pulmonary vasculature caused in part by pathologic changes of endothelial cell functions. Although heterogeneity of endothelial cells across various vascular beds is well known, the diversity among endothelial cells in the healthy pulmonary vascular bed and the pathologic diversity among pulmonary arterial endothelial cells (PAEC) in PAH is unknown and previously unexplored. Here single-cell RNA sequencing technology was used to decipher the cellular heterogeneity among PAEC in the human pulmonary arteries isolated from explanted lungs from three patients with PAH undergoing lung transplantation and three healthy donor lungs not utilized for transplantation. Datasets of 36,368 PAH individual endothelial cells and 36,086 healthy cells were analyzed using the SeqGeq bioinformatics program. Total population differential gene expression analyses identified 629 differentially expressed genes between PAH and controls. Gene Ontology and Canonical Ingenuity analysis revealed pathways that are known to be involved in pathogenesis, as well as unique new pathways. At the individual cell level, dimensionality reduction followed by density based clustering revealed the presence of eight unique PAEC clusters that were typified by proliferative, angiogenic or quiescent phenotypes. While control and PAH harbored many similar subgroups of endothelial cells, PAH had greater proportions of angiogenic and proliferative subsets. These findings identify that only specific subgroups of PAH PAEC have gene expression different than healthy PAEC, and suggest these subpopulations lead to the pathologic functions leading to remodeling.
Collapse
|
25
|
Xiao W, Oldham WM, Priolo C, Pandey AK, Loscalzo J. Immunometabolic Endothelial Phenotypes: Integrating Inflammation and Glucose Metabolism. Circ Res 2021; 129:9-29. [PMID: 33890812 PMCID: PMC8221540 DOI: 10.1161/circresaha.120.318805] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/20/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - William M. Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine (W.M.O., C.P.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Carmen Priolo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine (W.M.O., C.P.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Arvind K. Pandey
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine (W.X., A.K.P., J.L.), Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Ma Y, Chen SS, Jiang F, Ma RY, Wang HL. Bioinformatic analysis and validation of microRNA-508-3p as a protective predictor by targeting NR4A3/MEK axis in pulmonary arterial hypertension. J Cell Mol Med 2021; 25:5202-5219. [PMID: 33942991 PMCID: PMC8178270 DOI: 10.1111/jcmm.16523] [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] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) featured a debilitating progressive disorder. Here, we intend to determine diagnosis‐valuable biomarkers for PAH and decode the fundamental mechanisms of the biological function of these markers. Two mRNA microarray profiles (GSE70456 and GSE117261) and two microRNA microarray profiles (GSE55427 and GSE67597) were mined from the Gene Expression Omnibus platform. Then, we identified the differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs), respectively. Besides, we investigated online miRNA prediction tools to screen the target gene of DEMs. In this study, 185 DEGs and three common DEMs were screened as well as 1266 target genes of the three DEMs were identified. Next, 16 overlapping dysregulated genes from 185 DEGs and 1266 target gene were obtained. Meanwhile, we constructed the miRNA gene regulatory network and determined miRNA‐508‐3p‐NR4A3 pair for deeper exploring. Experiment methods verified the functional expression of miR‐508‐3p in PAH and its signalling cascade. We observed that ectopic miR‐508‐3p expression promotes proliferation and migration of pulmonary artery smooth muscle cell (PASMC). Bioinformatic, dual‐luciferase assay showed NR4A3 represents directly targeted gene of miR‐508‐3p. Mechanistically, we demonstrated that down‐regulation of miR‐508‐3p advances PASMC proliferation and migration via inducing NR4A3 to activate MAPK/ERK kinase signalling pathway. Altogether, our research provides a promising diagnosis of predictor and therapeutic avenues for patients in PAH.
Collapse
Affiliation(s)
- Yi Ma
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Shu-Shu Chen
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Fen Jiang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ru-Yi Ma
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China
| | - Huan-Liang Wang
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, China.,Shenzhen Research Institute of Shandong University, Shenzhen, China
| |
Collapse
|
27
|
Zemskova M, McClain N, Niihori M, Varghese MV, James J, Rafikov R, Rafikova O. Necrosis-Released HMGB1 (High Mobility Group Box 1) in the Progressive Pulmonary Arterial Hypertension Associated With Male Sex. Hypertension 2020; 76:1787-1799. [PMID: 33012199 DOI: 10.1161/hypertensionaha.120.16118] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Damage-associated molecular patterns, such as HMGB1 (high mobility group box 1), play a well-recognized role in the development of pulmonary arterial hypertension (PAH), a progressive fatal disease of the pulmonary vasculature. However, the contribution of the particular type of vascular cells, type of cell death, or the form of released HMGB1 in PAH remains unclear. Moreover, although male patients with PAH show a higher level of circulating HMGB1, its involvement in the severe PAH phenotype reported in males is unknown. In this study, we aimed to investigate the sources and active forms of HMGB1 released from damaged vascular cells and their contribution to the progressive type of PAH in males. Our results showed that HMGB1 is released by either pulmonary artery human endothelial cells or human pulmonary artery smooth muscle cells that underwent necrotic cell death, although only human pulmonary artery smooth muscle cells produce HMGB1 during apoptosis. Moreover, only human pulmonary artery smooth muscle cell death induced a release of dimeric HMGB1, found to be mitochondrial reactive oxygen species dependent, and TLR4 (toll-like receptor 4) activation. The modified Sugen/Hypoxia rat model replicates the human sexual dimorphism in PAH severity (right ventricle systolic pressure in males versus females 54.7±2.3 versus 44.6±2 mm Hg). By using this model, we confirmed that necroptosis and necrosis are the primary sources of circulating HMGB1 in the male rats, although only necrosis increased circulation of HMGB1 dimers. Attenuation of necrosis but not apoptosis or necroptosis prevented TLR4 activation in males and blunted the sex differences in PAH severity. We conclude that necrosis, through the release of HMGB1 dimers, predisposes males to a progressive form of PAH.
Collapse
Affiliation(s)
- Marina Zemskova
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Nolan McClain
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Maki Niihori
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Mathews V Varghese
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Joel James
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Ruslan Rafikov
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| | - Olga Rafikova
- From the Division of Endocrinology, Department of Medicine, University of Arizona College of Medicine, Tucson
| |
Collapse
|
28
|
Zhang JR, Sun HJ. LncRNAs and circular RNAs as endothelial cell messengers in hypertension: mechanism insights and therapeutic potential. Mol Biol Rep 2020; 47:5535-5547. [PMID: 32567025 DOI: 10.1007/s11033-020-05601-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Endothelial cells are major constituents in the vasculature, and they act as important players in vascular homeostasis via secretion/release of vasodilators and vasoconstrictors. In healthy arteries, endothelial cells play a key role in the regulation of vascular tone, cellular adhesion, and angiogenesis. A shift in the functions of the blood vessels toward vasoconstriction, proinflammatory state, oxidative stress and deficiency of nitric oxide (NO) might lead to endothelial dysfunction, a key event implicated in the pathophysiology of cardiovascular metabolic diseases, including diabetes, atherosclerosis, arterial hypertension and pulmonary arterial hypertension (PAH). Thus, reversibility of endothelial dysfunction may be beneficial for maintaining vascular homeostasis. In recent years, accumulative evidence has documented that noncoding RNAs (ncRNAs) are critically involved in endothelial homeostasis. Specifically, long noncoding RNAs (lncRNAs) and circular RNAs are highly expressed in endothelial cells where they serve as important mediators in normal endothelial functions. Dysregulation of lncRNAs and circular RNAs has been tightly associated with hypertension-related endothelial dysfunction. In this review, we will summarize the current progression and underlying mechanisms of lncRNA and circular RNA in endothelial cell biology under hypertensive conditions. We will also highlight their potential as biomarkers or therapeutic targets for hypertension and its associated endothelial dysfunction.
Collapse
Affiliation(s)
- Ji-Ru Zhang
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Wuxi, 214062, People's Republic of China
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, People's Republic of China. .,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
29
|
Zeineh N, Denora N, Laquintana V, Franco M, Weizman A, Gavish M. Efficaciousness of Low Affinity Compared to High Affinity TSPO Ligands in the Inhibition of Hypoxic Mitochondrial Cellular Damage Induced by Cobalt Chloride in Human Lung H1299 Cells. Biomedicines 2020; 8:biomedicines8050106. [PMID: 32370132 PMCID: PMC7277862 DOI: 10.3390/biomedicines8050106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/25/2022] Open
Abstract
The 18 kDa translocator protein (TSPO) plays an important role in apoptotic cell death, including apoptosis induced by the hypoxia mimicking agent cobalt chloride (CoCl2). In this study, the protective effects of a high (CB86; Ki = 1.6 nM) and a low (CB204; Ki = 117.7 nM) affinity TSPO ligands were investigated in H1299 lung cancer cell line exposed to CoCl2. The lung cell line H1299 was chosen in the present study since they express TSPO and able to undergo programmed cell death. The examined cell death markers included: ATP synthase reversal, reactive oxygen species (ROS) generation, mitochondrial membrane potential (Δψm) depolarization, cellular toxicity, and cellular viability. Pretreatment of the cells with the low affinity ligand CB204 at a concentration of 100 µM suppressed significantly (p < 0.05 for all) CoCl2-induced cellular cytotoxicity (100%), ATP synthase reversal (67%), ROS generation (82%), Δψm depolarization (100%), reduction in cellular density (97%), and also increased cell viability (85%). Furthermore, the low affinity TSPO ligand CB204, was harmless when given by itself at 100 µM. In contrast, the high affinity ligand (CB86) was significantly effective only in the prevention of CoCl2–induced ROS generation (39%, p < 0.001), and showed significant cytotoxic effects when given alone at 100 µM, as reflected in alterations in ADP/ATP ratio, oxidative stress, mitochondrial membrane potential depolarization and cell death. It appears that similar to previous studies on brain-derived cells, the relatively low affinity for the TSPO target enhances the potency of TSPO ligands in the protection from hypoxic cell death. Moreover, the high affinity TSPO ligand CB86, but not the low affinity ligand CB204, was lethal to the lung cells at high concentration (100 µM). The low affinity TSPO ligand CB204 may be a candidate for the treatment of pulmonary diseases related to hypoxia, such as pulmonary ischemia and chronic obstructive pulmonary disease COPD.
Collapse
Affiliation(s)
- Nidal Zeineh
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel;
| | - Nunzio Denora
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (N.D.); (V.L.); (M.F.)
- Institute for Chemical and Physical Processes (IPCF)-CNR SS Bari, Via Orabona 4, 70126 Bari, Italy
| | - Valentino Laquintana
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (N.D.); (V.L.); (M.F.)
| | - Massimo Franco
- Department of Pharmacy–Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (N.D.); (V.L.); (M.F.)
| | - Abraham Weizman
- Research Unit at Geha Mental Health Center and Laboratory of Biological Psychiatry at Felsenstein Medical Research Center, Petah Tikva 4910002, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Moshe Gavish
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion Institute of Technology, Haifa 31096, Israel;
- Correspondence: ; Tel.: +972-4829-5275; Fax: +972-4829-5330
| |
Collapse
|
30
|
McDowell RE, Aulak KS, Almoushref A, Melillo CA, Brauer BE, Newman JE, Tonelli AR, Dweik RA. Platelet glycolytic metabolism correlates with hemodynamic severity in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2020; 318:L562-L569. [PMID: 32022593 DOI: 10.1152/ajplung.00389.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Group 1 pulmonary hypertension (PH), i.e., pulmonary arterial hypertension (PAH), is associated with a metabolic shift favoring glycolysis in cells comprising the lung vasculature as well as skeletal muscle and right heart. We sought to determine whether this metabolic switch is also detectable in circulating platelets from PAH patients. We used Seahorse Extracellular Flux to measure bioenergetics in platelets isolated from group 1 PH (PAH), group 2 PH, patients with dyspnea and normal pulmonary artery pressures, and healthy controls. We show that platelets from group 1 PH patients exhibit enhanced basal glycolysis and lower glycolytic reserve compared with platelets from healthy controls but do not differ from platelets of group 2 PH or dyspnea patients without PH. Although we were unable to identify a glycolytic phenotype unique to platelets from PAH patients, we found that platelet glycolytic metabolism correlated with hemodynamic severity only in group 1 PH patients, supporting the known link between PAH pathology and altered glycolytic metabolism and extending this association to ex vivo platelets. Pulmonary artery pressure and pulmonary vascular resistance in patients with group 1 PH were directly associated with basal platelet glycolysis and inversely associated with maximal and reserve glycolysis, suggesting that PAH progression reduces the capacity for glycolysis even while demanding an increase in glycolytic metabolism. Therefore, platelets may provide an easy-to-harvest, real-time window into the metabolic shift occurring in the lung vasculature and represent a useful surrogate for interrogating the glycolytic shift central to PAH pathology.
Collapse
Affiliation(s)
- Ruth E McDowell
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Kulwant S Aulak
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Allaa Almoushref
- Department of Pulmonary, Allergy, and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Celia A Melillo
- Department of Pulmonary, Allergy, and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Brittany E Brauer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jennie E Newman
- Department of Pulmonary, Allergy, and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Adriano R Tonelli
- Department of Pulmonary, Allergy, and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Raed A Dweik
- Department of Pulmonary, Allergy, and Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
31
|
Arwood MJ, Vahabi N, Lteif C, Sharma RK, Machado RF, Duarte JD. Transcriptome-wide analysis associates ID2 expression with combined pre- and post-capillary pulmonary hypertension. Sci Rep 2019; 9:19572. [PMID: 31862991 PMCID: PMC6925238 DOI: 10.1038/s41598-019-55700-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/25/2019] [Indexed: 01/11/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) patients who develop pulmonary hypertension (PH) have an increased risk of death, with combined pre- and post-capillary PH (CpcPH) having the highest risk. However, the mechanism behind PH development in HFpEF is poorly understood. We aimed to identify transcriptomic associations with PH development in HFpEF. Blood was collected from 30 HFpEF patients: 10 without PH, 10 with isolated post-capillary PH, and 10 with CpcPH. Gene expression measurements were completed using transcriptome-wide RNA sequencing. Gene expression differences were compared using a quasi-likelihood method adjusting for age, sex, race, and smoking-status. Biological pathways were compared using global gene expression differences. A replication in 34 additional heart failure patients and a validation in lung tissue from a representative mouse model were completed using quantitative PCR. Six differentially expressed genes were identified when comparing transcriptomics between subjects with CpcPH and those without PH. When tested in additional subjects, only the association with ID2 replicated. Consistent with clinical findings, Id2 expression was also upregulated in mice with HFpEF and PH. Pathway analysis identified proliferative and mitochondrial pathways associated with CpcPH. Thus, these patients may possess systemic pathophysiological differences similar to those observed in pulmonary arterial hypertension patients.
Collapse
Affiliation(s)
- Meghan J Arwood
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Nasim Vahabi
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Christelle Lteif
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Ravindra K Sharma
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Roberto F Machado
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University, Indianapolis, IN, USA
| | - Julio D Duarte
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
32
|
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
|
33
|
Xu W, Comhair SAA, Chen R, Hu B, Hou Y, Zhou Y, Mavrakis LA, Janocha AJ, Li L, Zhang D, Willard BB, Asosingh K, Cheng F, Erzurum SC. Integrative proteomics and phosphoproteomics in pulmonary arterial hypertension. Sci Rep 2019; 9:18623. [PMID: 31819116 PMCID: PMC6901481 DOI: 10.1038/s41598-019-55053-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial endothelial cells (PAEC) are mechanistically linked to origins of pulmonary arterial hypertension (PAH). Here, global proteomics and phosphoproteomics of PAEC from PAH (n = 4) and healthy lungs (n = 5) were performed using LC-MS/MS to confirm known pathways and identify new areas of investigation in PAH. Among PAH and control cells, 170 proteins and 240 phosphopeptides were differentially expressed; of these, 45 proteins and 18 phosphopeptides were located in the mitochondria. Pathologic pathways were identified with integrative bioinformatics and human protein-protein interactome network analyses, then confirmed with targeted proteomics in PAH PAEC and non-targeted metabolomics and targeted high-performance liquid chromatography of metabolites in plasma from PAH patients (n = 30) and healthy controls (n = 12). Dysregulated pathways in PAH include accelerated one carbon metabolism, abnormal tricarboxylic acid (TCA) cycle flux and glutamate metabolism, dysfunctional arginine and nitric oxide pathways, and increased oxidative stress. Functional studies in cells confirmed abnormalities in glucose metabolism, mitochondrial oxygen consumption, and production of reactive oxygen species in PAH. Altogether, the findings indicate that PAH is typified by changes in metabolic pathways that are primarily found in mitochondria.
Collapse
Affiliation(s)
- Weiling Xu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| | - Suzy A A Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ruoying Chen
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Bo Hu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yuan Hou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yadi Zhou
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Lori A Mavrakis
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Allison J Janocha
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ling Li
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Dongmei Zhang
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Belinda B Willard
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Kewal Asosingh
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Feixiong Cheng
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America. .,Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, United States of America.
| |
Collapse
|
34
|
Suresh K, Servinsky L, Jiang H, Bigham Z, Zaldumbide J, Huetsch JC, Kliment C, Acoba MG, Kirsch BJ, Claypool SM, Le A, Damarla M, Shimoda LA. Regulation of mitochondrial fragmentation in microvascular endothelial cells isolated from the SU5416/hypoxia model of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 317:L639-L652. [PMID: 31461316 PMCID: PMC6879901 DOI: 10.1152/ajplung.00396.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a morbid disease characterized by progressive right ventricle (RV) failure due to elevated pulmonary artery pressures (PAP). In PAH, histologically complex vaso-occlusive lesions in the pulmonary vasculature contribute to elevated PAP. However, the mechanisms underlying dysfunction of the microvascular endothelial cells (MVECs) that comprise a significant portion of these lesions are not well understood. We recently showed that MVECs isolated from the Sugen/hypoxia (SuHx) rat experimental model of PAH (SuHx-MVECs) exhibit increases in migration/proliferation, mitochondrial reactive oxygen species (ROS; mtROS) production, intracellular calcium levels ([Ca2+]i), and mitochondrial fragmentation. Furthermore, quenching mtROS with the targeted antioxidant MitoQ attenuated basal [Ca2+]i, migration and proliferation; however, whether increased mtROS-induced [Ca2+]i entry affected mitochondrial morphology was not clear. In this study, we sought to better understand the relationship between increased ROS, [Ca2+]i, and mitochondrial morphology in SuHx-MVECs. We measured changes in mitochondrial morphology at baseline and following inhibition of mtROS, with the targeted antioxidant MitoQ, or transient receptor potential vanilloid-4 (TRPV4) channels, which we previously showed were responsible for mtROS-induced increases in [Ca2+]i in SuHx-MVECs. Quenching mtROS or inhibiting TRPV4 attenuated fragmentation in SuHx-MVECs. Conversely, inducing mtROS production in MVECs from normoxic rats (N-MVECs) increased fragmentation. Ca2+ entry induced by the TRPV4 agonist GSK1017920A was significantly increased in SuHx-MVECs and was attenuated with MitoQ treatment, indicating that mtROS contributes to increased TRPV4 activity in SuHx-MVECs. Basal and maximal respiration were depressed in SuHx-MVECs, and inhibiting mtROS, but not TRPV4, improved respiration in these cells. Collectively, our data show that, in SuHx-MVECs, mtROS production promotes TRPV4-mediated increases in [Ca2+]i, mitochondrial fission, and decreased mitochondrial respiration. These results suggest an important role for mtROS in driving MVEC dysfunction in PAH.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura Servinsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zahna Bigham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joel Zaldumbide
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John C Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Corrine Kliment
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michelle G Acoba
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian J Kirsch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
35
|
Tan H, Yao H, Lie Z, Chen G, Lin S, Zhang Y. MicroRNA‑30a‑5p promotes proliferation and inhibits apoptosis of human pulmonary artery endothelial cells under hypoxia by targeting YKL‑40. Mol Med Rep 2019; 20:236-244. [PMID: 31115541 PMCID: PMC6579982 DOI: 10.3892/mmr.2019.10251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 04/08/2019] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal and currently incurable cardiopulmonary disease. Numerous microRNAs (miRNAs) serve important roles in the development of PAH. While the expression of miR-30a-5p was downregulated in the lung tissue of rats in a pulmonary hypertension rat model, the expression pattern and function of miR-30a-5p in human PAH remain unclear. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to examine miR-30a-5p and chitinase-3-like protein 1 (YKL-40) mRNA expression levels. The expression levels of YKL-40 and apoptosis-associated proteins were measured by western blot analysis. Cell proliferation assays and flow cytometry analysis were performed to examine cell proliferation and apoptosis, respectively. The association between miR-30a-5p and YKL-40 was determined by a luciferase reporter assay, RT-qPCR and western blot analysis. The relative expression levels of miR-30a-5p in plasma were increased in patients with PAH [median=13.23 (25th percentile=6.388, 75th percentile=21.91)] compared with normal controls [median=2.25 (25th percentile=1.4, 75th percentile=3.7). The expression of miR-30a-5p was significantly downregulated while the protein expression of YKL-40 was significantly upregulated in hypoxia-induced human pulmonary artery endothelial cells (HPAECs) when compared with the hypoxia-induced group at 0 h. miR-30a-5p overexpression promoted HPAEC growth and inhibited apoptosis of HPAECs under hypoxia. A miR-30a-5p mimic decreased the luciferase activity of a luciferase reporter construct containing YKL-40 3′-untranslated region and also decreased YKL-40 protein expression. YKL-40 overexpression partly alleviated the effects of miR-30a-5p upregulation on proliferation and apoptosis of HPAECs under hypoxia. In conclusion, the data indicated that miR-30a-5p promoted cell growth and inhibited apoptosis of HPAECs under hypoxia by targeting YKL-40. Therefore, the miR-30a-5p/YKL-40 axis may provide a potential target for the development of novel PAH therapies.
Collapse
Affiliation(s)
- Hong Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Hua Yao
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Zhenbang Lie
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Guo Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Shuguang Lin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| | - Ying Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, P.R. China
| |
Collapse
|
36
|
Carman BL, Predescu DN, Machado R, Predescu SA. Plexiform Arteriopathy in Rodent Models of Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1133-1144. [PMID: 30926336 DOI: 10.1016/j.ajpath.2019.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 12/11/2022]
Abstract
As time progresses, our understanding of disease pathology is propelled forward by technological advancements. Much of the advancements that aid in understanding disease mechanics are based on animal studies. Unfortunately, animal models often fail to recapitulate the entirety of the human disease. This is especially true with animal models used to study pulmonary arterial hypertension (PAH), a disease with two distinct phases. The first phase is defined by nonspecific medial and adventitial thickening of the pulmonary artery and is commonly reproduced in animal models, including the classic models (ie, hypoxia-induced pulmonary hypertension and monocrotaline lung injury model). However, many animal models, including the classic models, fail to capture the progressive, or second, phase of PAH. This is a stage defined by plexogenic arteriopathy, resulting in obliteration and occlusion of the small- to mid-sized pulmonary vessels. Each of these two phases results in severe pulmonary hypertension that directly leads to right ventricular hypertrophy, decompensated right-sided heart failure, and death. Fortunately, newly developed animal models have begun to address the second, more severe, side of PAH and aid in our ability to develop new therapeutics. Moreover, p38 mitogen-activated protein kinase activation emerges as a central molecular mediator of plexiform lesions in both experimental models and human disease. Therefore, this review will focus on plexiform arteriopathy in experimental animal models of PAH.
Collapse
Affiliation(s)
- Brandon L Carman
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Dan N Predescu
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois
| | - Roberto Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Sanda A Predescu
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, Rush Medical College, Chicago, Illinois.
| |
Collapse
|
37
|
Smolders VF, Zodda E, Quax PHA, Carini M, Barberà JA, Thomson TM, Tura-Ceide O, Cascante M. Metabolic Alterations in Cardiopulmonary Vascular Dysfunction. Front Mol Biosci 2019; 5:120. [PMID: 30723719 PMCID: PMC6349769 DOI: 10.3389/fmolb.2018.00120] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/31/2018] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide. CVD comprise a range of diseases affecting the functionality of the heart and blood vessels, including acute myocardial infarction (AMI) and pulmonary hypertension (PH). Despite their different causative mechanisms, both AMI and PH involve narrowed or blocked blood vessels, hypoxia, and tissue infarction. The endothelium plays a pivotal role in the development of CVD. Disruption of the normal homeostasis of endothelia, alterations in the blood vessel structure, and abnormal functionality are essential factors in the onset and progression of both AMI and PH. An emerging theory proposes that pathological blood vessel responses and endothelial dysfunction develop as a result of an abnormal endothelial metabolism. It has been suggested that, in CVD, endothelial cell metabolism switches to higher glycolysis, rather than oxidative phosphorylation, as the main source of ATP, a process designated as the Warburg effect. The evidence of these alterations suggests that understanding endothelial metabolism and mitochondrial function may be central to unveiling fundamental mechanisms underlying cardiovascular pathogenesis and to identifying novel critical metabolic biomarkers and therapeutic targets. Here, we review the role of the endothelium in the regulation of vascular homeostasis and we detail key aspects of endothelial cell metabolism. We also describe recent findings concerning metabolic endothelial cell alterations in acute myocardial infarction and pulmonary hypertension, their relationship with disease pathogenesis and we discuss the future potential of pharmacological modulation of cellular metabolism in the treatment of cardiopulmonary vascular dysfunction. Although targeting endothelial cell metabolism is still in its infancy, it is a promising strategy to restore normal endothelial functions and thus forestall or revert the development of CVD in personalized multi-hit interventions at the metabolic level.
Collapse
Affiliation(s)
- Valérie Françoise Smolders
- Department of Biochemistry and Molecular Biology and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Erika Zodda
- Department of Biochemistry and Molecular Biology and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paul H. A. Quax
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Marina Carini
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Timothy M. Thomson
- Institute for Molecular Biology of Barcelona, National Research Council (IBMB-CSIC), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biology and Institute of Biomedicine (IBUB), Faculty of Biology, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| |
Collapse
|
38
|
Cheong HI, Farha S, Park MM, Thomas JD, Saygin D, Comhair SAA, Sharp J, Highland KB, Tang WHW, Erzurum SC. Endothelial Phenotype Evoked by Low Dose Carvedilol in Pulmonary Hypertension. Front Cardiovasc Med 2018; 5:180. [PMID: 30619887 PMCID: PMC6299019 DOI: 10.3389/fcvm.2018.00180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/30/2018] [Indexed: 01/06/2023] Open
Abstract
Background: The therapeutic benefits of β-blockers are well established in left heart failure. The Pulmonary Arterial Hypertension Treatment with Carvedilol for Heart Failure [PAHTCH] study showed safety and possible benefit of carvedilol in pulmonary arterial hypertension (PAH) associated right heart failure over 6 months. This study aims at evaluating the short-term cardiovascular effects and early mechanistic biomarkers of carvedilol therapy. Methods: Thirty patients with pulmonary hypertension (PH) received low dose carvedilol (3.125 mg twice daily) for 1 week prior to randomization to placebo, low-dose, or dose-escalating carvedilol therapy. Echocardiography was performed at baseline and 1 week. Exercise capacity was assessed by 6 min walk distance (6MWD). The L-arginine/nitric oxide pathway and other biological markers of endothelial function were measured. Results: All participants tolerated 1 week of carvedilol without adverse effects. After 1 week of carvedilol, 6MWD and heart rate at peak exercise did not vary (both p > 0.1). Heart rate at rest and 1 min post walk dropped significantly (both p < 0.05) with a trend for increase in heart rate recovery (p = 0.08). Right ventricular systolic pressure (RVSP) decreased by an average of 13 mmHg (p = 0.002). Patients who had a decrease in RVSP of more than 10 mm Hg were defined as responders (n = 17), and those with a lesser drop as non-responders (n = 13). Responders had a significant drop in pulmonary vascular resistance (PVR) after 1 week of carvedilol (p = 0.004). In addition, responders had a greater decrease in heart rate at rest and 1 min post walk compared to non-responders (both p < 0.05). Responders had higher plasma arginine and global bioavailability of arginine at baseline compared to non-responders (p = 0.03 and p = 0.05, respectively). After 1 week of carvedilol, responders had greater increase in urinary nitrate (p = 0.04). Responders treated with carvedilol had a sustained drop in RVSP and PVR after 6 months of carvedilol with no change in cardiac output. Conclusions: Low-dose carvedilol for 1 week can potentially identify a PH responder phenotype that may benefit from β-blockers that is associated with less endothelial dysfunction. Clinical Trial Registration:http://www.clinicaltrials.gov. identifier: NCT01586156.
Collapse
Affiliation(s)
- Hoi I Cheong
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Samar Farha
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Margaret M Park
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, United States
| | - James D Thomas
- Heart and Vascular Institute, Northwestern University Hospital, Chicago, IL, United States
| | - Didem Saygin
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Suzy A A Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jacqueline Sharp
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, United States
| | | | - W H Wilson Tang
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Serpil C Erzurum
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
39
|
Karmouty-Quintana H, Guignabert C, Kwapiszewska G, Ormiston ML. Editorial: Molecular Mechanisms in Pulmonary Hypertension and Right Ventricle Dysfunction. Front Physiol 2018; 9:1777. [PMID: 30618793 PMCID: PMC6298242 DOI: 10.3389/fphys.2018.01777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/23/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Christophe Guignabert
- Institut National de la Santé et de la Recherche Médicale UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Graz, Austria
| | - Mark L Ormiston
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.,Department of Medicine, Queen's University, Kingston, ON, Canada.,Department of Surgery, Queen's University, Kingston, ON, Canada
| |
Collapse
|
40
|
Wang Z, Yang K, Zheng Q, Zhang C, Tang H, Babicheva A, Jiang Q, Li M, Chen Y, Carr SG, Wu K, Zhang Q, Balistrieri A, Wang C, Song S, Ayon RJ, Desai AA, Black SM, Garcia JGN, Makino A, Yuan JXJ, Lu W, Wang J. Divergent changes of p53 in pulmonary arterial endothelial and smooth muscle cells involved in the development of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 316:L216-L228. [PMID: 30358436 DOI: 10.1152/ajplung.00538.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The tumor-suppressive role of p53, a transcription factor that regulates the expression of many genes, has been linked to cell cycle arrest, apoptosis, and senescence. The noncanonical function or the pathogenic role of p53 has more recently been implicated in pulmonary vascular disease. We previously reported that rapid nuclear accumulation of hypoxia-inducible factor (HIF)-1α in pulmonary arterial smooth muscle cells (PASMCs) upregulates transient receptor potential channels and enhances Ca2+ entry to increase cytosolic Ca2+ concentration ([Ca2+]cyt). Also, we observed differences in HIF-1α/2α expression in PASMCs and pulmonary arterial endothelial cells (PAECs). Here we report that p53 is increased in PAECs, but decreased in PASMCs, isolated from mice with hypoxia-induced pulmonary hypertension (PH) and rats with monocrotaline (MCT)-induced PH (MCT-PH). The increased p53 in PAECs from rats with MCT-PH is associated with an increased ratio of Bax/Bcl-2, while the decreased p53 in PASMCs is associated with an increased HIF-1α. Furthermore, p53 is downregulated in PASMCs isolated from patients with idiopathic pulmonary arterial hypertension compared with PASMCs from normal subjects. Overexpression of p53 in normal PASMCs inhibits store-operated Ca2+ entry (SOCE) induced by passive depletion of intracellularly stored Ca2+ in the sarcoplasmic reticulum, while downregulation of p53 enhances SOCE. These data indicate that differentially regulated expression of p53 and HIF-1α/2α in PASMCs and PAECs and the cross talk between p53 and HIF-1α/2α in PASMCs and PAECs may play an important role in the development of PH via, at least in part, induction of PAEC apoptosis and PASMC proliferation.
Collapse
Affiliation(s)
- Ziyi Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Qiuyu Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Aleksandra Babicheva
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Qian Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Meichan Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Shane G Carr
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Kang Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Qian Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Angela Balistrieri
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Christina Wang
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ankit A Desai
- Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona
| | - Stephen M Black
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Joe G N Garcia
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine , Tucson, Arizona
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou , China.,Division of Translational and Regenerative Medicine , Tucson, Arizona.,Department of Medicine, The University of Arizona College of Medicine , Tucson, Arizona.,Division of Pulmonary and Critical Care Medicine, The People's Hospital of Inner Mongolia, Huhhot, Inner Mongolia, China
| |
Collapse
|
41
|
Kuebler WM, Nicolls MR, Olschewski A, Abe K, Rabinovitch M, Stewart D, Chan SY, Morrell NW, Archer SL, Spiekerkoetter E. A pro-con debate: current controversies in PAH pathogenesis at the American Thoracic Society International Conference in 2017. Am J Physiol Lung Cell Mol Physiol 2018; 315:L502-L516. [PMID: 29877097 PMCID: PMC6230875 DOI: 10.1152/ajplung.00150.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/22/2018] [Accepted: 06/02/2018] [Indexed: 12/16/2022] Open
Abstract
The following review summarizes the pro-con debate about current controversies regarding the pathogenesis of pulmonary arterial hypertension (PAH) that took place at the American Thoracic Society Conference in May 2017. Leaders in the field of PAH research discussed the importance of the immune system, the role of hemodynamic stress and endothelial apoptosis, as well as bone morphogenetic protein receptor-2 signaling in PAH pathogenesis. Whereas this summary does not intend to resolve obvious conflicts in opinion, we hope that the presented arguments entice further discussions and draw a new generation of enthusiastic researchers into this vibrant field of science to bridge existing gaps for a better understanding and therapy of this fatal disease.
Collapse
Affiliation(s)
- Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitaetsmedizin Berlin, Berlin , Germany
- Keenan Research Centre for Biomedical Science at Saint Michael's , Toronto, Ontario , Canada
- Department of Surgery, University of Toronto , Toronto, Ontario , Canada
- Department of Physiology, University of Toronto , Toronto, Ontario , Canada
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care, Department of Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University , Stanford, California
| | - Andrea Olschewski
- Ludwig Boltzmann Institute, Lung Vascular Research, Medical University of Graz , Graz , Austria
- Johannes Kepler University Linz, Medicine Rectorate, Linz, Austria
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences , Fukuoka , Japan
| | - Marlene Rabinovitch
- Division of Cardiology, Department of Pediatrics, Stanford University School of Medicine , Stanford, California
| | - Duncan Stewart
- Division of Cardiology, Department of Medicine, Ottawa Hospital Research Institute , Ottawa, Ontario , Canada
| | - Stephen Y Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Nicholas W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, University of Cambridge , Cambridge , United Kingdom
| | - Stephen L Archer
- Department of Medicine, Queen's University , Kingston, Ontario , Canada
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care, Department of Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University , Stanford, California
| |
Collapse
|
42
|
Culley MK, Chan SY. Mitochondrial metabolism in pulmonary hypertension: beyond mountains there are mountains. J Clin Invest 2018; 128:3704-3715. [PMID: 30080181 DOI: 10.1172/jci120847] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pulmonary hypertension (PH) is a heterogeneous and fatal disease of the lung vasculature, where metabolic and mitochondrial dysfunction may drive pathogenesis. Similar to the Warburg effect in cancer, a shift from mitochondrial oxidation to glycolysis occurs in diseased pulmonary vessels and the right ventricle. However, appreciation of metabolic events in PH beyond the Warburg effect is only just emerging. This Review discusses molecular, translational, and clinical concepts centered on the mitochondria and highlights promising, controversial, and challenging areas of investigation. If we can move beyond the "mountains" of obstacles in this field and elucidate these fundamental tenets of pulmonary vascular metabolism, such work has the potential to usher in much-needed diagnostic and therapeutic approaches for the mitochondrial and metabolic management of PH.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
43
|
Suresh K, Servinsky L, Jiang H, Bigham Z, Yun X, Kliment C, Huetsch J, Damarla M, Shimoda LA. Reactive oxygen species induced Ca 2+ influx via TRPV4 and microvascular endothelial dysfunction in the SU5416/hypoxia model of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 314:L893-L907. [PMID: 29388466 PMCID: PMC6008124 DOI: 10.1152/ajplung.00430.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/05/2018] [Accepted: 01/24/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a lethal disease characterized by elevations in pulmonary arterial pressure, in part due to formation of occlusive lesions in the distal arterioles of the lung. These complex lesions may comprise multiple cell types, including endothelial cells (ECs). To better understand the molecular mechanisms underlying EC dysfunction in PAH, lung microvascular endothelial cells (MVECs) were isolated from normoxic rats (N-MVECs) and rats subjected to SU5416 plus hypoxia (SuHx), an experimental model of PAH. Compared with N-MVECs, MVECs isolated from SuHx rats (SuHx-MVECs) appeared larger and more spindle shaped morphologically and expressed canonical smooth muscle cell markers smooth muscle-specific α-actin and myosin heavy chain in addition to endothelial markers such as Griffonia simplicifolia and von Willebrand factor. SuHx-MVEC mitochondria were dysfunctional, as evidenced by increased fragmentation/fission, decreased oxidative phosphorylation, and increased reactive oxygen species (ROS) production. Functionally, SuHx-MVECs exhibited increased basal levels of intracellular calcium concentration ([Ca2+]i) and enhanced migratory and proliferative capacity. Treatment with global (TEMPOL) or mitochondria-specific (MitoQ) antioxidants decreased ROS levels and basal [Ca2]i in SuHx-MVECs. TEMPOL and MitoQ also decreased migration and proliferation in SuHx-MVECs. Additionally, inhibition of ROS-induced Ca2+ entry via pharmacologic blockade of transient receptor potential vanilloid-4 (TRPV4) attenuated [Ca2]i, migration, and proliferation. These findings suggest a role for mitochondrial ROS-induced Ca2+ influx via TRPV4 in promoting abnormal migration and proliferation in MVECs in this PAH model.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Laura Servinsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Zahna Bigham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Xin Yun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Corrine Kliment
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - John Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine , Baltimore, Maryland
| |
Collapse
|
44
|
Du J, Xu Z, Liu Q, Yang Y, Qian H, Hu M, Fan Y, Li Q, Yao W, Li H, Qian G, He B, Zhou D, Mao C, Wang G. ATG101 Single-Stranded Antisense RNA-Loaded Triangular DNA Nanoparticles Control Human Pulmonary Endothelial Growth via Regulation of Cell Macroautophagy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:42544-42555. [PMID: 29154530 DOI: 10.1021/acsami.7b13504] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Autophagy plays a key role in pulmonary vascular remodeling via regulation of apoptosis and hyperproliferation of pulmonary arterial endothelial cells, which are the subject of increased attention. Autophagy-related 101 (ATG101) is an essential gene for the initiation of autophagy. Although the structure of ATG101 has been well-characterized, its exact biological function in autophagy is still unknown. In this study, an ATG101 single-stranded antisense RNA-loaded DNA triangular nanoparticle (ssATG101-TNP) is constructed to knock down the ATG101 gene expression. ssATG101-TNP can be effectively transfected into human pulmonary arterial endothelial cells (HPAECs) in time- and dose-dependent manners. Knockdown of ATG101 promotes cell apoptosis as well as inhibits cell autophagy and proliferation with hypoxic stimulation. Additionally, the hedgehog/Gli signal pathway is involved in ATG101-mediated macroautophagy and HPAEC proliferation. This study found that ATG101, an important member of the autophagy gene family, can regulate cell macroautophagy, apoptosis, and growth in HPAECs. ssATG101-TNP is demonstrated to be a nontoxic, highly efficient, gene-delivery vehicle for HPAECs. These findings also suggest that ATG101 might be a potential therapeutic target in diseases involving endothelial injury.
Collapse
Affiliation(s)
- Juan Du
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Zhi Xu
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Qian Liu
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Yu Yang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Hang Qian
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Mingdong Hu
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Ye Fan
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Qi Li
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Wei Yao
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Hongli Li
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Guisheng Qian
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Binfeng He
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| | - Deshan Zhou
- Department of Histology and Embryology, Capital Medical University , Beijing 100069, China
| | - Chengde Mao
- Department of Chemistry, Purdue University , West Lafayette, Indiana 47907, United States
| | - Guansong Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University , Chongqing 400037, China
| |
Collapse
|
45
|
van de Pol V, Kurakula K, DeRuiter MC, Goumans MJ. Thoracic Aortic Aneurysm Development in Patients with Bicuspid Aortic Valve: What Is the Role of Endothelial Cells? Front Physiol 2017; 8:938. [PMID: 29249976 PMCID: PMC5714935 DOI: 10.3389/fphys.2017.00938] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022] Open
Abstract
Bicuspid aortic valve (BAV) is the most common type of congenital cardiac malformation. Patients with a BAV have a predisposition for the development of thoracic aortic aneurysm (TAA). This pathological aortic dilation may result in aortic rupture, which is fatal in most cases. The abnormal aortic morphology of TAAs results from a complex series of events that alter the cellular structure and extracellular matrix (ECM) composition of the aortic wall. Because the major degeneration is located in the media of the aorta, most studies aim to unravel impaired smooth muscle cell (SMC) function in BAV TAA. However, recent studies suggest that endothelial cells play a key role in both the initiation and progression of TAAs by influencing the medial layer. Aortic endothelial cells are activated in BAV mediated TAAs and have a substantial influence on ECM composition and SMC phenotype, by secreting several key growth factors and matrix modulating enzymes. In recent years there have been significant advances in the genetic and molecular understanding of endothelial cells in BAV associated TAAs. In this review, the involvement of the endothelial cells in BAV TAA pathogenesis is discussed. Endothelial cell functioning in vessel homeostasis, flow response and signaling will be highlighted to give an overview of the importance and the under investigated potential of endothelial cells in BAV-associated TAA.
Collapse
Affiliation(s)
- Vera van de Pol
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Kondababu Kurakula
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
46
|
Caruso P, Dunmore BJ, Schlosser K, Schoors S, Dos Santos C, Perez-Iratxeta C, Lavoie JR, Zhang H, Long L, Flockton AR, Frid MG, Upton PD, D'Alessandro A, Hadinnapola C, Kiskin FN, Taha M, Hurst LA, Ormiston ML, Hata A, Stenmark KR, Carmeliet P, Stewart DJ, Morrell NW. Identification of MicroRNA-124 as a Major Regulator of Enhanced Endothelial Cell Glycolysis in Pulmonary Arterial Hypertension via PTBP1 (Polypyrimidine Tract Binding Protein) and Pyruvate Kinase M2. Circulation 2017; 136:2451-2467. [PMID: 28971999 DOI: 10.1161/circulationaha.117.028034] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/08/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by abnormal growth and enhanced glycolysis of pulmonary artery endothelial cells. However, the mechanisms underlying alterations in energy production have not been identified. METHODS Here, we examined the miRNA and proteomic profiles of blood outgrowth endothelial cells (BOECs) from patients with heritable PAH caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2) gene and patients with idiopathic PAH to determine mechanisms underlying abnormal endothelial glycolysis. We hypothesized that in BOECs from patients with PAH, the downregulation of microRNA-124 (miR-124), determined with a tiered systems biology approach, is responsible for increased expression of the splicing factor PTBP1 (polypyrimidine tract binding protein), resulting in alternative splicing of pyruvate kinase muscle isoforms 1 and 2 (PKM1 and 2) and consequently increased PKM2 expression. We questioned whether this alternative regulation plays a critical role in the hyperglycolytic phenotype of PAH endothelial cells. RESULTS Heritable PAH and idiopathic PAH BOECs recapitulated the metabolic abnormalities observed in pulmonary artery endothelial cells from patients with idiopathic PAH, confirming a switch from oxidative phosphorylation to aerobic glycolysis. Overexpression of miR-124 or siRNA silencing of PTPB1 restored normal proliferation and glycolysis in heritable PAH BOECs, corrected the dysregulation of glycolytic genes and lactate production, and partially restored mitochondrial respiration. BMPR2 knockdown in control BOECs reduced the expression of miR-124, increased PTPB1, and enhanced glycolysis. Moreover, we observed reduced miR-124, increased PTPB1 and PKM2 expression, and significant dysregulation of glycolytic genes in the rat SUGEN-hypoxia model of severe PAH, characterized by reduced BMPR2 expression and endothelial hyperproliferation, supporting the relevance of this mechanism in vivo. CONCLUSIONS Pulmonary vascular and circulating progenitor endothelial cells isolated from patients with PAH demonstrate downregulation of miR-124, leading to the metabolic and proliferative abnormalities in PAH ECs via PTPB1 and PKM1/PKM2. Therefore, the manipulation of this miRNA or its targets could represent a novel therapeutic approach for the treatment of PAH.
Collapse
Affiliation(s)
- Paola Caruso
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Benjamin J Dunmore
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Kenny Schlosser
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Sandra Schoors
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (S.S., P.C.).,Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Belgium (S.S., P.C.)
| | - Claudia Dos Santos
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.S., M.L.O.)
| | - Carol Perez-Iratxeta
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Jessie R Lavoie
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Lu Long
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Amanda R Flockton
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Paul D Upton
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | | | - Charaka Hadinnapola
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Fedir N Kiskin
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Mohamad Taha
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Liam A Hurst
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Mark L Ormiston
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.S., M.L.O.)
| | - Akiko Hata
- University of Colorado, Anschutz Medical Campus, Aurora. Cardiovascular Research Institute, University of California, San Francisco (A.H.)
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (S.S., P.C.).,Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Belgium (S.S., P.C.)
| | - Duncan J Stewart
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Nicholas W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| |
Collapse
|
47
|
Zhang H, Wang D, Li M, Plecitá-Hlavatá L, D'Alessandro A, Tauber J, Riddle S, Kumar S, Flockton A, McKeon BA, Frid MG, Reisz JA, Caruso P, El Kasmi KC, Ježek P, Morrell NW, Hu CJ, Stenmark KR. Metabolic and Proliferative State of Vascular Adventitial Fibroblasts in Pulmonary Hypertension Is Regulated Through a MicroRNA-124/PTBP1 (Polypyrimidine Tract Binding Protein 1)/Pyruvate Kinase Muscle Axis. Circulation 2017; 136:2468-2485. [PMID: 28972001 DOI: 10.1161/circulationaha.117.028069] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 09/12/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND An emerging metabolic theory of pulmonary hypertension (PH) suggests that cellular and mitochondrial metabolic dysfunction underlies the pathology of this disease. We and others have previously demonstrated the existence of hyperproliferative, apoptosis-resistant, proinflammatory adventitial fibroblasts from human and bovine hypertensive pulmonary arterial walls (PH-Fibs) that exhibit constitutive reprogramming of glycolytic and mitochondrial metabolism, accompanied by an increased ratio of glucose catabolism through glycolysis versus the tricarboxylic acid cycle. However, the mechanisms responsible for these metabolic alterations in PH-Fibs remain unknown. We hypothesized that in PH-Fibs microRNA-124 (miR-124) regulates PTBP1 (polypyrimidine tract binding protein 1) expression to control alternative splicing of pyruvate kinase muscle (PKM) isoforms 1 and 2, resulting in an increased PKM2/PKM1 ratio, which promotes glycolysis and proliferation even in aerobic environments. METHODS Pulmonary adventitial fibroblasts were isolated from calves and humans with severe PH (PH-Fibs) and from normal subjects. PTBP1 gene knockdown was achieved via PTBP1-siRNA; restoration of miR-124 was performed with miR-124 mimic. TEPP-46 and shikonin were used to manipulate PKM2 glycolytic function. Histone deacetylase inhibitors were used to treat cells. Metabolic products were determined by mass spectrometry-based metabolomics analyses, and mitochondrial function was analyzed by confocal microscopy and spectrofluorometry. RESULTS We detected an increased PKM2/PKM1 ratio in PH-Fibs compared with normal subjects. PKM2 inhibition reversed the glycolytic status of PH-Fibs, decreased their cell proliferation, and attenuated macrophage interleukin-1β expression. Furthermore, normalizing the PKM2/PKM1 ratio in PH-Fibs by miR-124 overexpression or PTBP1 knockdown reversed the glycolytic phenotype (decreased the production of glycolytic intermediates and byproducts, ie, lactate), rescued mitochondrial reprogramming, and decreased cell proliferation. Pharmacological manipulation of PKM2 activity with TEPP-46 and shikonin or treatment with histone deacetylase inhibitors produced similar results. CONCLUSIONS In PH, miR-124, through the alternative splicing factor PTBP1, regulates the PKM2/PKM1 ratio, the overall metabolic, proliferative, and inflammatory state of cells. This PH phenotype can be rescued with interventions at various levels of the metabolic cascade. These findings suggest a more integrated view of vascular cell metabolism, which may open unique therapeutic prospects in targeting the dynamic glycolytic and mitochondrial interactions and between mesenchymal inflammatory cells in PH.
Collapse
Affiliation(s)
- Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Daren Wang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Min Li
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Lydie Plecitá-Hlavatá
- University of Colorado Anschutz Medical Campus, Aurora. Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, Prague (L.P.-H., J.T., P.J.)
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics and Biological Mass Spectrometry Shared Resource (A.D., J.A.R.)
| | - Jan Tauber
- University of Colorado Anschutz Medical Campus, Aurora. Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, Prague (L.P.-H., J.T., P.J.)
| | - Suzette Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Sushil Kumar
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Amanda Flockton
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - B Alexandre McKeon
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics and Biological Mass Spectrometry Shared Resource (A.D., J.A.R.)
| | - Paola Caruso
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., N.W.M.)
| | - Karim C El Kasmi
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition (K.C.E.K.)
| | - Petr Ježek
- University of Colorado Anschutz Medical Campus, Aurora. Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, Prague (L.P.-H., J.T., P.J.)
| | - Nicholas W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., N.W.M.)
| | - Cheng-Jun Hu
- Department of Craniofacial Biology, School of Dental Medicine (C.-J.H.)
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., D.W., M.L., S.R., S.K., A.F., B.A.M., M.G.F., K.R.S.)
| |
Collapse
|
48
|
Hong Z, Chen KH, DasGupta A, Potus F, Dunham-Snary K, Bonnet S, Tian L, Fu J, Breuils-Bonnet S, Provencher S, Wu D, Mewburn J, Ormiston ML, Archer SL. MicroRNA-138 and MicroRNA-25 Down-regulate Mitochondrial Calcium Uniporter, Causing the Pulmonary Arterial Hypertension Cancer Phenotype. Am J Respir Crit Care Med 2017; 195:515-529. [PMID: 27648837 DOI: 10.1164/rccm.201604-0814oc] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is an obstructive vasculopathy characterized by excessive pulmonary artery smooth muscle cell (PASMC) proliferation, migration, and apoptosis resistance. This cancer-like phenotype is promoted by increased cytosolic calcium ([Ca2+]cyto), aerobic glycolysis, and mitochondrial fission. OBJECTIVES To determine how changes in mitochondrial calcium uniporter (MCU) complex (MCUC) function influence mitochondrial dynamics and contribute to PAH's cancer-like phenotype. METHODS PASMCs were isolated from patients with PAH and healthy control subjects and assessed for expression of MCUC subunits. Manipulation of the pore-forming subunit, MCU, in PASMCs was achieved through small interfering RNA knockdown or MCU plasmid-mediated up-regulation, as well as through modulation of the upstream microRNAs (miRs) miR-138 and miR-25. In vivo, nebulized anti-miRs were administered to rats with monocrotaline-induced PAH. MEASUREMENTS AND MAIN RESULTS Impaired MCUC function, resulting from down-regulation of MCU and up-regulation of an inhibitory subunit, mitochondrial calcium uptake protein 1, is central to PAH's pathogenesis. MCUC dysfunction decreases intramitochondrial calcium ([Ca2+]mito), inhibiting pyruvate dehydrogenase activity and glucose oxidation, while increasing [Ca2+]cyto, promoting proliferation, migration, and fission. In PAH PASMCs, increasing MCU decreases cell migration, proliferation, and apoptosis resistance by lowering [Ca2+]cyto, raising [Ca2+]mito, and inhibiting fission. In normal PASMCs, MCUC inhibition recapitulates the PAH phenotype. In PAH, elevated miRs (notably miR-138) down-regulate MCU directly and also by decreasing MCU's transcriptional regulator cAMP response element-binding protein 1. Nebulized anti-miRs against miR-25 and miR-138 restore MCU expression, reduce cell proliferation, and regress established PAH in the monocrotaline model. CONCLUSIONS These results highlight miR-mediated MCUC dysfunction as a unifying mechanism in PAH that can be therapeutically targeted.
Collapse
Affiliation(s)
- Zhigang Hong
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Kuang-Hueih Chen
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Asish DasGupta
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Francois Potus
- 2 Pulmonary Hypertension Research Group of the University Cardiology and Pulmonary Institute of the Quebec Research Centre, Laval University, Quebec City, Quebec, Canada
| | | | - Sebastien Bonnet
- 2 Pulmonary Hypertension Research Group of the University Cardiology and Pulmonary Institute of the Quebec Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Lian Tian
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Jennifer Fu
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Sandra Breuils-Bonnet
- 2 Pulmonary Hypertension Research Group of the University Cardiology and Pulmonary Institute of the Quebec Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Steeve Provencher
- 2 Pulmonary Hypertension Research Group of the University Cardiology and Pulmonary Institute of the Quebec Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Danchen Wu
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Jeffrey Mewburn
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Mark L Ormiston
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| | - Stephen L Archer
- 1 Department of Medicine, Queen's University, Kingston, Ontario, Canada; and
| |
Collapse
|
49
|
Xiong A, Liu Y. Targeting Hypoxia Inducible Factors-1α As a Novel Therapy in Fibrosis. Front Pharmacol 2017; 8:326. [PMID: 28611671 PMCID: PMC5447768 DOI: 10.3389/fphar.2017.00326] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/16/2017] [Indexed: 02/05/2023] Open
Abstract
Fibrosis, characterized by increased extracellular matrix (ECM) deposition, and widespread vasculopathy, has the prominent trait of chronic hypoxia. Hypoxia inducible factors-1α (HIF-1α), a key transcriptional factor in response to this chronic hypoxia, is involved in fibrotic disease, such as Systemic sclerosis (SSc). The implicated function of HIF-1α in fibrosis include stimulation of excessive ECM, vascular remodeling, and futile angiogenesis with further exacerbation of chronic hypoxia and deteriorate pathofibrogenesis. This review will focus on the molecular biological behavior of HIF-1α in regulating progressive fibrosis. Better understanding of the role for HIF-1α-regulated pathways in fibrotic disease will accelerate development of novel therapeutic strategies that target HIF-1α. Such new therapeutic strategies may be particularly effective for treatment of the prototypic, multisystem fibrotic, autoimmune disease SSc.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan UniversityChengdu, China
| |
Collapse
|
50
|
White HA, Jin Y, Chicoine LG, Chen B, Liu Y, Nelin LD. Hypoxic proliferation requires EGFR-mediated ERK activation in human pulmonary microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L649-L656. [PMID: 28188223 DOI: 10.1152/ajplung.00267.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
We have previously shown that hypoxic proliferation of human pulmonary microvascular endothelial cells (hPMVECs) depends on epidermal growth factor receptor (EGFR) activation. To determine downstream signaling leading to proliferation, we tested the hypothesis that hypoxia-induced proliferation in hPMVECs would require EGFR-mediated activation of extracellular signal-regulated kinase (ERK) leading to arginase II induction. To test this hypothesis, hPMVECs were incubated in either normoxia (21% O2, 5% CO2) or hypoxia (1% O2, 5% CO2) and Western blotting was performed for EGFR, arginase II, phosphorylated-ERK (pERK), and total ERK (ERK). Hypoxia led to greater EGFR, pERK, and arginase II protein levels than did normoxia in hPMVECs. To examine the role of EGFR in these hypoxia-induced changes, hPMVECs were transfected with siRNA against EGFR or a scrambled siRNA and placed in hypoxia. Inhibition of EGFR using siRNA attenuated hypoxia-induced pERK and arginase II expression as well as the hypoxia-induced increase in viable cell numbers. hPMVECs were then treated with vehicle, an EGFR inhibitor (AG1478), or an ERK pathway inhibitor (U0126) and placed in hypoxia. Pharmacologic inhibition of EGFR significantly attenuated the hypoxia-induced increase in pERK level. Both AG1478 and U0126 also significantly attenuated the hypoxia-induced increase in viable hPMVECs numbers. hPMVECs were transfected with an adenoviral vector containing arginase II (AdArg2) and overexpression of arginase II rescued the U0126-mediated decrease in viable cell numbers in hypoxic hPMVECs. Our findings suggest that hypoxic activation of EGFR results in phosphorylation of ERK, which is required for hypoxic induction of arginase II and cellular proliferation.
Collapse
Affiliation(s)
- Hilary A White
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Louis G Chicoine
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|