101
|
Li M, Riddle S, Kumar S, Poczobutt J, McKeon BA, Frid MG, Ostaff M, Reisz JA, Nemkov T, Fini MA, Laux A, Hu CJ, El Kasmi KC, D’Alessandro A, Brown RD, Zhang H, Stenmark KR. Microenvironmental Regulation of Macrophage Transcriptomic and Metabolomic Profiles in Pulmonary Hypertension. Front Immunol 2021; 12:640718. [PMID: 33868271 PMCID: PMC8044406 DOI: 10.3389/fimmu.2021.640718] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022] Open
Abstract
The recruitment and subsequent polarization of inflammatory monocytes/macrophages in the perivascular regions of pulmonary arteries is a key feature of pulmonary hypertension (PH). However, the mechanisms driving macrophage polarization within the adventitial microenvironment during PH progression remain unclear. We previously established that reciprocal interactions between fibroblasts and macrophages are essential in driving the activated phenotype of both cell types although the signals involved in these interactions remain undefined. We sought to test the hypothesis that adventitial fibroblasts produce a complex array of metabolites and proteins that coordinately direct metabolomic and transcriptomic re-programming of naïve macrophages to recapitulate the pathophysiologic phenotype observed in PH. Media conditioned by pulmonary artery adventitial fibroblasts isolated from pulmonary hypertensive (PH-CM) or age-matched control (CO-CM) calves were used to activate bone marrow derived macrophages. RNA-Seq and mass spectrometry-based metabolomics analyses were performed. Fibroblast conditioned medium from patients with idiopathic pulmonary arterial hypertension or controls were used to validate transcriptional findings. The microenvironment was targeted in vitro using a fibroblast-macrophage co-culture system and in vivo in a mouse model of hypoxia-induced PH. Both CO-CM and PH-CM actively, yet distinctly regulated macrophage transcriptomic and metabolomic profiles. Network integration revealed coordinated rewiring of pro-inflammatory and pro-remodeling gene regulation in concert with altered mitochondrial and intermediary metabolism in response to PH-CM. Pro-inflammation and metabolism are key regulators of macrophage phenotype in vitro, and are closely related to in vivo flow sorted lung interstitial/perivascular macrophages from hypoxic mice. Metabolic changes are accompanied by increased free NADH levels and increased expression of a metabolic sensor and transcriptional co-repressor, C-terminal binding protein 1 (CtBP1), a mechanism shared with adventitial PH-fibroblasts. Targeting the microenvironment created by both cell types with the CtBP1 inhibitor MTOB, inhibited macrophage pro-inflammatory and metabolic re-programming both in vitro and in vivo. In conclusion, coordinated transcriptional and metabolic reprogramming is a critical mechanism regulating macrophage polarization in response to the complex adventitial microenvironment in PH. Targeting the adventitial microenvironment can return activated macrophages toward quiescence and attenuate pathological remodeling that drives PH progression.
Collapse
Affiliation(s)
- Min Li
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Suzette Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sushil Kumar
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Joanna Poczobutt
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - B. Alexandre McKeon
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maria G. Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maureen Ostaff
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mehdi A. Fini
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Aya Laux
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cheng-Jun Hu
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karim C. El Kasmi
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - R. Dale Brown
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
102
|
Wu H, Cui M, Li C, Li H, Dai Y, Cui K, Li Z. Kaempferol Reverses Aerobic Glycolysis via miR-339-5p-Mediated PKM Alternative Splicing in Colon Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:3060-3068. [PMID: 33663206 DOI: 10.1021/acs.jafc.0c07640] [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: 06/12/2023]
Abstract
Colon cancer is an aggressive malignancy with very limited therapeutic approaches. The available therapeutic agents for colon cancer show strong adverse effects and poor effectiveness, indicating the urgent need to identify new therapeutic drugs for this malignancy. Kaempferol, a flavonoid found in a variety of natural foods, exhibits significant inhibitory effects on colon cancer. Here, it was found that kaempferol inhibited the proliferation of human colon cancer cells HCT116 and DLD1 in a dose-dependent manner, and the IC50 values were 63.0 ± 12.9 and 98.3 ± 15.9 μM, respectively. Also, kaempferol treatment delayed G1 phase progression of cell cycle and induced apoptosis. Aerobic glycolysis is the major energy source for various tumor growths, including colon cancer. Indeed, kaempferol treatment impaired glucose consumption, which subsequently led to reduced lactic acid accumulation and ATP production. Mechanistically, kaempferol promoted the expression of miR-339-5p. Further studies identified hnRNPA1 and PTBP1 as two direct targets of miR-339-5p. By directly targeting hnRNPA1 and PTBP1, miR-339-5p reduced the expression of M2-type pyruvate kinase (PKM2) but induced that of PKM1. In conclusion, these data demonstrate that by modulating miR-339-5p-hnRNPA1/PTBP1-PKM2 axis, kaempferol inhibits glycolysis and colon cancer growth, which reveals a new explanation for the molecular mechanism underlying kaempferol anti-tumor.
Collapse
Affiliation(s)
- Haili Wu
- College of Life Science, Shanxi University, Taiyuan 030006, China
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030006, China
| | - Mimi Cui
- Institute of Biotechnology, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Chenglu Li
- College of Life Science, Shanxi University, Taiyuan 030006, China
| | - Hanqing Li
- College of Life Science, Shanxi University, Taiyuan 030006, China
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030006, China
| | - Yuhao Dai
- College of Life Science, Shanxi University, Taiyuan 030006, China
| | - Kaili Cui
- Institute of Biotechnology, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- College of Life Science, Shanxi University, Taiyuan 030006, China
- Institute of Biotechnology, The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
- Shanxi Key Laboratory for Research and Development of Regional Plants, Taiyuan 030006, China
| |
Collapse
|
103
|
Ho JJD, Man JHS, Schatz JH, Marsden PA. Translational remodeling by RNA-binding proteins and noncoding RNAs. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1647. [PMID: 33694288 DOI: 10.1002/wrna.1647] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/14/2022]
Abstract
Responsible for generating the proteome that controls phenotype, translation is the ultimate convergence point for myriad upstream signals that influence gene expression. System-wide adaptive translational reprogramming has recently emerged as a pillar of cellular adaptation. As classic regulators of mRNA stability and translation efficiency, foundational studies established the concept of collaboration and competition between RNA-binding proteins (RBPs) and noncoding RNAs (ncRNAs) on individual mRNAs. Fresh conceptual innovations now highlight stress-activated, evolutionarily conserved RBP networks and ncRNAs that increase the translation efficiency of populations of transcripts encoding proteins that participate in a common cellular process. The discovery of post-transcriptional functions for long noncoding RNAs (lncRNAs) was particularly intriguing given their cell-type-specificity and historical definition as nuclear-functioning epigenetic regulators. The convergence of RBPs, lncRNAs, and microRNAs on functionally related mRNAs to enable adaptive protein synthesis is a newer biological paradigm that highlights their role as "translatome (protein output) remodelers" and reinvigorates the paradigm of "RNA operons." Together, these concepts modernize our understanding of cellular stress adaptation and strategies for therapeutic development. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications Translation > Translation Regulation Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs.
Collapse
Affiliation(s)
- J J David Ho
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Division of Hematology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jeffrey H S Man
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Respirology, University Health Network, Latner Thoracic Research Laboratories, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan H Schatz
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Division of Hematology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Philip A Marsden
- Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
104
|
Li K, Li Y, Yu Y, Ding J, Huang H, Chu C, Hu L, Yu Y, Cao Y, Xu P, Fulton D, Chen F. Bmi-1 alleviates adventitial fibroblast senescence by eliminating ROS in pulmonary hypertension. BMC Pulm Med 2021; 21:80. [PMID: 33673825 PMCID: PMC7934412 DOI: 10.1186/s12890-021-01439-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/17/2021] [Indexed: 01/19/2023] Open
Abstract
Objectives Pulmonary hypertension (PH) is a life-threatening progressive disease with high mortality in the elderly. However, the pathogenesis of PH has not been fully understood and there is no effective therapy to reverse the disease process. This study aims to determine whether cellular senescence is involved in the development of PH. Methods The rat PH model was established by intraperitoneal injection of monocrotaline and evaluated by pulmonary arteriole wall thickness and right ventricular hypertrophy index. Human lung fibroblasts (HLFs) were treated with CoCl2 or hypoxia to induce cellular senescence in vitro. SA-β-gal staining and the changes of senescent markers were used to examine cellular senescence. The molecular mechanism of cellular senescence was further explored by detecting reactive oxygen species (ROS) levels and culturing cells with a conditioned medium. Results We revealed the cellular senescence of pulmonary adventitial fibroblasts in vivo in the rat PH model. The expression of Bmi-1, an important regulator of senescence, was decreased in the lungs of PH rats and localized in adventitial fibroblasts. The in vitro experiments showed that p16 expression was increased while Bmi-1 expression was decreased after CoCl2 treatment in HLFs. Mechanistically, Bmi-1 could alleviate CoCl2-induced HLFs senescence by eliminating ROS which further promoted the proliferation of pulmonary artery smooth muscle cells by paracrine mode of action of HLFs. Conclusion Bmi-1 alleviates the cellular senescence of pulmonary fibroblasts in PH, which expands the pathogenesis of PH and provides a theoretical basis for targeting senescent cells in the treatment of PH. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-021-01439-0.
Collapse
Affiliation(s)
- Kai Li
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yan Li
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China.
| | - Youjia Yu
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Jingjing Ding
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Huijie Huang
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Chunyan Chu
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Li Hu
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yanfang Yu
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Yue Cao
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - Peng Xu
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, People's Republic of China. .,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, People's Republic of China.
| |
Collapse
|
105
|
James J, Zemskova M, Eccles CA, Varghese MV, Niihori M, Barker NK, Luo M, Mandarino LJ, Langlais PR, Rafikova O, Rafikov R. Single Mutation in the NFU1 Gene Metabolically Reprograms Pulmonary Artery Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2021; 41:734-754. [PMID: 33297749 PMCID: PMC7837686 DOI: 10.1161/atvbaha.120.314655] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE NFU1 is a mitochondrial iron-sulfur scaffold protein, involved in iron-sulfur assembly and transfer to complex II and LAS (lipoic acid synthase). Patients with the point mutation NFU1G208C and CRISPR/CAS9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9)-generated rats develop mitochondrial dysfunction leading to pulmonary arterial hypertension. However, the mechanistic understanding of pulmonary vascular proliferation due to a single mutation in NFU1 remains unresolved. Approach and Results: Quantitative proteomics of isolated mitochondria showed the entire phenotypic transformation of NFU1G206C rats with a disturbed mitochondrial proteomic landscape, involving significant changes in the expression of 208 mitochondrial proteins. The NFU1 mutation deranged the expression pattern of electron transport proteins, resulting in a significant decrease in mitochondrial respiration. Reduced reliance on mitochondrial respiration amplified glycolysis in pulmonary artery smooth muscle cell (PASMC) and activated GPD (glycerol-3-phosphate dehydrogenase), linking glycolysis to oxidative phosphorylation and lipid metabolism. Decreased PDH (pyruvate dehydrogenase) activity due to the lipoic acid shortage is compensated by increased fatty acid metabolism and oxidation. PASMC became dependent on extracellular fatty acid sources due to upregulated transporters such as CD36 (cluster of differentiation 36) and CPT (carnitine palmitoyltransferase)-1. Finally, the NFU1 mutation produced a dysregulated antioxidant system in the mitochondria, leading to increased reactive oxygen species levels. PASMC from NFU1 rats showed apoptosis resistance, increased anaplerosis, and attained a highly proliferative phenotype. Attenuation of mitochondrial reactive oxygen species by mitochondrial-targeted antioxidant significantly decreased PASMC proliferation. CONCLUSIONS The alteration in iron-sulfur metabolism completely transforms the proteomic landscape of the mitochondria, leading toward metabolic plasticity and redistribution of energy sources to the acquisition of a proliferative phenotype by the PASMC.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Proliferation
- Cells, Cultured
- Cellular Reprogramming
- Energy Metabolism
- Fatty Acids/metabolism
- Female
- Mitochondria, Liver/genetics
- Mitochondria, Liver/metabolism
- Mitochondria, Liver/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Point Mutation
- Proteome
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Rats
Collapse
Affiliation(s)
- Joel James
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Marina Zemskova
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Cody A. Eccles
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Mathews V. Varghese
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Maki Niihori
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Natalie K. Barker
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Moulun Luo
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Lawrence J. Mandarino
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Olga Rafikova
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| | - Ruslan Rafikov
- Department of Medicine, Division of Endocrinology, University of Arizona College of Medicine, Tucson
| |
Collapse
|
106
|
Puckett DL, Alquraishi M, Chowanadisai W, Bettaieb A. The Role of PKM2 in Metabolic Reprogramming: Insights into the Regulatory Roles of Non-Coding RNAs. Int J Mol Sci 2021; 22:1171. [PMID: 33503959 PMCID: PMC7865720 DOI: 10.3390/ijms22031171] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
Pyruvate kinase is a key regulator in glycolysis through the conversion of phosphoenolpyruvate (PEP) into pyruvate. Pyruvate kinase exists in various isoforms that can exhibit diverse biological functions and outcomes. The pyruvate kinase isoenzyme type M2 (PKM2) controls cell progression and survival through the regulation of key signaling pathways. In cancer cells, the dimer form of PKM2 predominates and plays an integral role in cancer metabolism. This predominance of the inactive dimeric form promotes the accumulation of phosphometabolites, allowing cancer cells to engage in high levels of synthetic processing to enhance their proliferative capacity. PKM2 has been recognized for its role in regulating gene expression and transcription factors critical for health and disease. This role enables PKM2 to exert profound regulatory effects that promote cancer cell metabolism, proliferation, and migration. In addition to its role in cancer, PKM2 regulates aspects essential to cellular homeostasis in non-cancer tissues and, in some cases, promotes tissue-specific pathways in health and diseases. In pursuit of understanding the diverse tissue-specific roles of PKM2, investigations targeting tissues such as the kidney, liver, adipose, and pancreas have been conducted. Findings from these studies enhance our understanding of PKM2 functions in various diseases beyond cancer. Therefore, there is substantial interest in PKM2 modulation as a potential therapeutic target for the treatment of multiple conditions. Indeed, a vast plethora of research has focused on identifying therapeutic strategies for targeting PKM2. Recently, targeting PKM2 through its regulatory microRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) has gathered increasing interest. Thus, the goal of this review is to highlight recent advancements in PKM2 research, with a focus on PKM2 regulatory microRNAs and lncRNAs and their subsequent physiological significance.
Collapse
Affiliation(s)
- Dexter L. Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Winyoo Chowanadisai
- Department of Nutrition, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| |
Collapse
|
107
|
Rai N, Shihan M, Seeger W, Schermuly RT, Novoyatleva T. Genetic Delivery and Gene Therapy in Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22031179. [PMID: 33503992 PMCID: PMC7865388 DOI: 10.3390/ijms22031179] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive complex fatal disease of multiple etiologies. Hyperproliferation and resistance to apoptosis of vascular cells of intimal, medial, and adventitial layers of pulmonary vessels trigger excessive pulmonary vascular remodeling and vasoconstriction in the course of pulmonary arterial hypertension (PAH), a subgroup of PH. Multiple gene mutation/s or dysregulated gene expression contribute to the pathogenesis of PAH by endorsing the proliferation and promoting the resistance to apoptosis of pulmonary vascular cells. Given the vital role of these cells in PAH progression, the development of safe and efficient-gene therapeutic approaches that lead to restoration or down-regulation of gene expression, generally involved in the etiology of the disease is the need of the hour. Currently, none of the FDA-approved drugs provides a cure against PH, hence innovative tools may offer a novel treatment paradigm for this progressive and lethal disorder by silencing pathological genes, expressing therapeutic proteins, or through gene-editing applications. Here, we review the effectiveness and limitations of the presently available gene therapy approaches for PH. We provide a brief survey of commonly existing and currently applicable gene transfer methods for pulmonary vascular cells in vitro and describe some more recent developments for gene delivery existing in the field of PH in vivo.
Collapse
Affiliation(s)
- Nabham Rai
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Mazen Shihan
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
| | - Ralph T. Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
| | - Tatyana Novoyatleva
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University of Giessen, Aulweg 130, 35392 Giessen, Germany; (N.R.); (M.S.); (W.S.); (R.T.S.)
- Correspondence:
| |
Collapse
|
108
|
Santos-Ferreira CA, Abreu MT, Marques CI, Gonçalves LM, Baptista R, Girão HM. Micro-RNA Analysis in Pulmonary Arterial Hypertension: Current Knowledge and Challenges. ACTA ACUST UNITED AC 2020; 5:1149-1162. [PMID: 33294743 PMCID: PMC7691282 DOI: 10.1016/j.jacbts.2020.07.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 01/18/2023]
Abstract
The role of miRNAs in PAH is fast expanding, and it is increasingly difficult to identify which molecules have the highest translational potential. This review discusses the challenges in miRNA analysis and interpretation in PAH and highlights 4 promising miRNAs in this field. Additional pre-clinical studies and clinical trials are urgently needed to bring miRNAs from the bench to the bedside soon.
Pulmonary arterial hypertension (PAH) is a rare, chronic disease of the pulmonary vasculature that is associated with poor outcomes. Its pathogenesis is multifactorial and includes micro-RNA (miRNA) deregulation. The understanding of the role of miRNAs in PAH is expanding quickly, and it is increasingly difficult to identify which miRNAs have the highest translational potential. This review summarizes the current knowledge of miRNA expression in PAH, discusses the challenges in miRNA analysis and interpretation, and highlights 4 promising miRNAs in this field (miR-29, miR-124, miR-140, and miR-204).
Collapse
Key Words
- BMPR2, bone morphogenetic protein receptor type 2
- EPC, endothelial progenitor cell
- HIF, hypoxia-inducible factor
- HPAH, hereditary pulmonary arterial hypertension
- MCT, monocrotaline
- PAAF, pulmonary arterial adventitial fibroblast
- PAEC, pulmonary artery endothelial cell
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cells
- PH, pulmonary hypertension
- RV, right ventricle
- SU/Hx/Nx, association of Sugen 5416 with chronic hypoxia followed by normoxia
- WHO, World Health Organization
- animal model
- lncRNA, long noncoding RNA
- mRNA, messenger RNA
- miRNA, micro-RNA
- micro-RNA
- microarray
- ncRNAs, noncoding RNAs
- pulmonary arterial hypertension
Collapse
Affiliation(s)
- Cátia A Santos-Ferreira
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Mónica T Abreu
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Carla I Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Lino M Gonçalves
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| | - Rui Baptista
- Cardiology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal.,Cardiology Department, Centro Hospitalar Entre Douro e Vouga, Santa Maria de Feira, Portugal
| | - Henrique M Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology, Coimbra, Portugal.,Clinical Academic Centre of Coimbra, Coimbra, Portugal
| |
Collapse
|
109
|
Lee LY, Pandey AK, Maron BA, Loscalzo J. Network medicine in Cardiovascular Research. Cardiovasc Res 2020; 117:2186-2202. [PMID: 33165538 DOI: 10.1093/cvr/cvaa321] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/08/2020] [Accepted: 10/30/2020] [Indexed: 12/21/2022] Open
Abstract
The ability to generate multi-omics data coupled with deeply characterizing the clinical phenotype of individual patients promises to improve understanding of complex cardiovascular pathobiology. There remains an important disconnection between the magnitude and granularity of these data and our ability to improve phenotype-genotype correlations for complex cardiovascular diseases. This shortcoming may be due to limitations associated with traditional reductionist analytical methods, which tend to emphasize a single molecular event in the pathogenesis of diseases more aptly characterized by crosstalk between overlapping molecular pathways. Network medicine is a rapidly growing discipline that considers diseases as the consequences of perturbed interactions between multiple interconnected biological components. This powerful integrative approach has enabled a number of important discoveries in complex disease mechanisms. In this review, we introduce the basic concepts of network medicine and highlight specific examples by which this approach has accelerated cardiovascular research. We also review how network medicine is well-positioned to promote rational drug design for patients with cardiovascular diseases, with particular emphasis on advancing precision medicine.
Collapse
Affiliation(s)
- Laurel Y Lee
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Arvind K Pandey
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.,Department of Cardiology, Boston VA Healthcare System, Boston, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
110
|
Zheng D, Jiang Y, Qu C, Yuan H, Hu K, He L, Chen P, Li J, Tu M, Lin L, Chen H, Lin Z, Lin W, Fan J, Cheng G, Hong J. Pyruvate Kinase M2 Tetramerization Protects against Hepatic Stellate Cell Activation and Liver Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2267-2281. [PMID: 32805235 PMCID: PMC7786052 DOI: 10.1016/j.ajpath.2020.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is an increasing health problem worldwide, for which no effective antifibrosis drugs are available. Although the involvement of aerobic glycolysis in hepatic stellate cell (HSC) activation has been reported, the role of pyruvate kinase M2 (PKM2) in liver fibrogenesis still remains unknown. We examined PKM2 expression and location in liver tissues and primary hepatic cells. The in vitro and in vivo effects of a PKM2 antagonist (shikonin) and its allosteric agent (TEPP-46) on liver fibrosis were investigated in HSCs and liver fibrosis mouse model. Chromatin immunoprecipitation sequencing and immunoprecipitation were performed to identify the relevant molecular mechanisms. PKM2 expression was significantly up-regulated in both mouse and human fibrotic livers compared with normal livers, and mainly detected in activated, rather than quiescent, HSCs. PKM2 knockdown markedly inhibited the activation and proliferation of HSCs in vitro. Interestingly, the PKM2 dimer, rather than the tetramer, induced HSC activation. PKM2 tetramerization induced by TEPP-46 effectively inhibited HSC activation, reduced aerobic glycolysis, and decreased MYC and CCND1 expression via regulating histone H3K9 acetylation in activated HSCs. TEPP-46 and shikonin dramatically attenuated liver fibrosis in vivo. Our findings demonstrate a nonmetabolic role of PKM2 in liver fibrosis. PKM2 tetramerization or suppression could prevent HSC activation and protects against liver fibrosis.
Collapse
Affiliation(s)
- Dandan Zheng
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuchuan Jiang
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chen Qu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Yuan
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lu He
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Peng Chen
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jinying Li
- Department of Gastroenterology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Mengxian Tu
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hengxing Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zelong Lin
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wenyu Lin
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jun Fan
- Departments of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, China
| | - Guohua Cheng
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jian Hong
- Department of Abdominal Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Pathophysiology, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
111
|
Abstract
INTRODUCTION Myofibroblasts are the primary executor and influencer in lung fibrosis. Latest studies on lung myofibroblast pathobiology have significantly advanced the understanding of the pathogenesis of lung fibrosis and shed new light on strategies targeting these cells to treat this disease. AREAS COVERED This article reviewed the most recent progresses, mainly within the last 5 years, on the definition, origin, activity regulation, and targeting of lung myofibroblasts in lung fibrosis. We did a literature search on PubMed using the keywords below from the dates 2010 to 2020. EXPERT OPINION With the improved cell lineage characterization and the advent of scRNA-seq, the field is having much better picture of the lung myofibroblast origin and mesenchymal heterogeneity. Additionally, cellular metabolism has emerged as a key regulation of lung myofibroblast pathogenic phenotype and is a promising therapeutic target for treating a variety of lung fibrotic disorders.
Collapse
Affiliation(s)
- Dingyuan Jiang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital; National Clinical Research Center for Respiratory Diseases , Beijing, China
| | - Tapan Dey
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama, USA
| |
Collapse
|
112
|
Chen S, Yan D, Qiu A. The role of macrophages in pulmonary hypertension: Pathogenesis and targeting. Int Immunopharmacol 2020; 88:106934. [PMID: 32889242 DOI: 10.1016/j.intimp.2020.106934] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/22/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
Abstract
Pulmonary hypertension (PH) is a pathophysiological disorder that can complicate most cardiovascular and respiratory diseases and may involve multiple clinical conditions, but its pathogenesis is poorly understood. Despite recent developments in the management of PH, there is an urgent need for new ways to effectively treat PH and reduce the risk of further complications. Recent studies have shown that dysregulated immunity underlies the development of PH. Myeloid cells, including monocytes and macrophages, participate in immune homeostasis and the adaptive immune response, but the function and production of these cells in PH is not well understood. A prominent pathological feature of pH is the accumulation of macrophages near the arterioles of the lung, indicating that pulmonary inflammation mediated by lung perivascular macrophages is a key driver of pulmonary remodelling, which leads to increased right ventricular systolic pressure. An improved understanding of the roles macrophages play in immune responses associated with PH may lead to new therapeutic targets. In this review, we highlight the relationship between macrophages and PH, the molecular mechanisms involved, and the recent advances in targeting these processes to treat PH.
Collapse
Affiliation(s)
- Shanshan Chen
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Dongmei Yan
- Department of Clinical Laboratory, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China
| | - Aimin Qiu
- Department of Respiratory and Critical Care Medicine, Yancheng Third People's Hospital, The Affiliated Yancheng Hospital of Southeast University Medical College, Jiangsu, China.
| |
Collapse
|
113
|
Lei J, Liu X, Song M, Zhou Y, Fan J, Shen X, Xu X, Kapoor I, Zhu G, Wang (王觉进) J. Aberrant Exon 8/8a Splicing by Downregulated PTBP (Polypyrimidine Tract-Binding Protein) 1 Increases Ca V1.2 Dihydropyridine Resistance to Attenuate Vasodilation. Arterioscler Thromb Vasc Biol 2020; 40:2440-2453. [PMID: 32787518 DOI: 10.1161/atvbaha.120.315010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Calcium channel blockers, such as dihydropyridines, are commonly used to inhibit enhanced activity of vascular CaV1.2 channels in hypertension. However, patients who are insensitive to such treatments develop calcium channel blocker-resistant hypertension. The function of CaV1.2 channel is diversified by alternative splicing, and the splicing factor PTBP (polypyrimidine tract-binding protein) 1 influences the utilization of mutually exclusive exon 8/8a of the CaV1.2 channel during neuronal development. Nevertheless, whether and how PTBP1 makes a role in the calcium channel blocker sensitivity of vascular CaV1.2 channels, and calcium channel blocker-induced vasodilation remains unknown. Approach and Results: We detected high expression of PTBP1 and, inversely, low expression of exon 8a in CaV1.2 channels (CaV1.2E8a) in rat arteries. In contrast, the opposite expression patterns were observed in brain and heart tissues. In comparison to normotensive rats, the expressions of PTBP1 and CaV1.2E8a channels were dysregulated in mesenteric arteries of hypertensive rats. Notably, PTBP1 expression was significantly downregulated, and CaV1.2E8a channels were aberrantly increased in dihydropyridine-resistant arteries compared with dihydropyridine-sensitive arteries of rats and human. In rat vascular smooth muscle cells, PTBP1 knockdown resulted in shifting of CaV1.2 exon 8 to 8a. Using patch-clamp recordings, we demonstrated a concomitant reduction of sensitivity of CaV1.2 channels to nifedipine, due to the higher expression of CaV1.2E8a isoform. In vascular myography experiments, small interfering RNA-mediated knockdown of PTBP1 attenuated nifedipine-induced vasodilation of rat mesenteric arteries. CONCLUSIONS PTBP1 finely modulates the sensitivities of CaV1.2 channels to dihydropyridine by shifting the utilization of exon 8/8a and resulting in changes of responses in dihydropyridine-induced vasodilation.
Collapse
Affiliation(s)
- Jianzhen Lei
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Xiaoxin Liu
- Shanghai Chest Hospital, Shanghai Jiaotong University, China (X.L.)
| | - Miaomiao Song
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Yingying Zhou
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Jia Fan
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Xiaowei Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (X.S., X.X.)
| | - Xiaohan Xu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (X.S., X.X.)
| | - Isha Kapoor
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Guoqing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Juejin Wang (王觉进)
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| |
Collapse
|
114
|
Tian L, Xiong PY, Alizadeh E, Lima PDA, Potus F, Mewburn J, Martin A, Chen K, Archer SL. Supra-coronary aortic banding improves right ventricular function in experimental pulmonary arterial hypertension in rats by increasing systolic right coronary artery perfusion. Acta Physiol (Oxf) 2020; 229:e13483. [PMID: 32339403 DOI: 10.1111/apha.13483] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
AIM Pulmonary arterial hypertension (PAH) results in right ventricular (RV) dysfunction owing, in part, to RV ischemia. The relative contribution of RV microvascular rarefaction vs reduced right coronary artery perfusion pressure (RCA-PP) to RV ischemia remains unknown. We hypothesize that increasing RCA-PP improves RV function in PAH by increasing RV systolic perfusion. METHODS Supra-coronary aortic banding (SAB) or sham surgery was performed on male Sprague-Dawley rats. Seven to ten days later, rats received either monocrotaline (MCT; 60 mg/kg) or saline. After 1 month, echocardiography, cardiac catheterization, 99m Tc-sestamibi single-photon emission computed tomography (SPECT) and microsphere infusion studies were performed. The RV was harvested for measurement of hypertrophy (RVH), fibrosis and immunoblotting, and the lung was harvested for pulmonary artery (PA) histology. RESULTS Supra-coronary aortic banding increased systolic pressures in proximal aorta and systolic RCA-PP in SAB + MCT vs MCT rats (114 ± 12 vs 5 ± 9 mm Hg), without altering diastolic RCA-PP. SAB + MCT rats had improved RV function vs MCT rats, evident from their significantly increased cardiac output (CO), RV free wall (RVFW) thickening, tricuspid annular plane systolic excursion (TAPSE) and RV-PA coupling indices. RV-PA coupling indices and CO correlated directly with systolic RCA-PP. RV perfusion was increased in SAB + MCT vs MCT rats and correlated well with CO; whereas microvascular rarefaction was unaltered. SAB + MCT rats had less RVH and fibrosis and lower PA pressures vs MCT rats. SAB + MCT rats had significantly lower RV pyruvate kinase muscle isoform 2/1 ratios than MCT rats, consistent with restoration of oxidative metabolism. CONCLUSION A SAB-induced increase in systolic RCA-PP improves RV perfusion and function in MCT rats. Maintaining systolic RCA perfusion can preserve RV function in PAH.
Collapse
Affiliation(s)
- Lian Tian
- Department of Medicine Queen's University Kingston ON Canada
| | - Ping Yu Xiong
- Department of Medicine Queen's University Kingston ON Canada
- Department of Biomedical and Molecular Sciences Queen's University Kingston ON Canada
| | | | | | - François Potus
- Department of Medicine Queen's University Kingston ON Canada
| | - Jeffrey Mewburn
- Department of Medicine Queen's University Kingston ON Canada
| | - Ashley Martin
- Department of Medicine Queen's University Kingston ON Canada
| | | | - Stephen L. Archer
- Department of Medicine Queen's University Kingston ON Canada
- Queen's CardioPulmonary Unit (QCPU) Kingston ON Canada
| |
Collapse
|
115
|
Pyruvate kinase M2 activation protects against the proliferation and migration of pulmonary artery smooth muscle cells. Cell Tissue Res 2020; 382:585-598. [PMID: 32719938 DOI: 10.1007/s00441-020-03245-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Pyruvate kinase M2 (PKM2), which is encoded by PKM, is a ubiquitously expressed intracellular protein and is associated with proliferation cell phenotype. In PAH patients and PAH models, we found higher levels of PKM2 tyrosine 105 phosphorylation (phospho-PKM2 (Y105)) than in controls, both in vivo and in vitro. Here, we demonstrate that PKM2 stimulates inflammatory and apoptosis signalling pathways in pulmonary artery smooth muscle cells (PASMCs) and promotes PASMC migration and proliferation. PKM2 phosphorylation promoted the dimerization activation and nuclear translocation of STAT3, a transcription factor regulating proliferation, growth, and apoptosis. TLR2, a transmembrane protein receptor involved in both innate and adaptive immune responses, promoted PKM2 phosphorylation in hypoxia-induced PASMCs. Therefore, we hypothesized that PKM2 also affects the proliferation and migration of PASMCs. The proliferation of hypoxia-induced normal human pulmonary artery smooth muscle cells (normal-HPASMCs) was found to be inhibited by TEPP-46 (PKM2 agonist) and PKM2 siRNA using wound healing, 5-ethynyl-2'-deoxyuridine (EdU), and immunofluorescence (Ki67) assays. PASMCs isolated from PAH patients (PAH-HPASMCs) and hypoxia-treated rats (PAH-RPASMCs) also confirmed the above results. TEPP-46 treatment was found to improve hypoxia-induced pulmonary artery remodelling and right heart function in mice, and the link between PKM2 and STAT3 was also confirmed in vivo. In conclusion, PKM2 plays crucial roles in the proliferation and migration of PASMCs.
Collapse
|
116
|
Leopold JA, Maron BA, Loscalzo J. The application of big data to cardiovascular disease: paths to precision medicine. J Clin Invest 2020; 130:29-38. [PMID: 31895052 DOI: 10.1172/jci129203] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Advanced phenotyping of cardiovascular diseases has evolved with the application of high-resolution omics screening to populations enrolled in large-scale observational and clinical trials. This strategy has revealed that considerable heterogeneity exists at the genotype, endophenotype, and clinical phenotype levels in cardiovascular diseases, a feature of the most common diseases that has not been elucidated by conventional reductionism. In this discussion, we address genomic context and (endo)phenotypic heterogeneity, and examine commonly encountered cardiovascular diseases to illustrate the genotypic underpinnings of (endo)phenotypic diversity. We highlight the existing challenges in cardiovascular disease genotyping and phenotyping that can be addressed by the integration of big data and interpreted using novel analytical methodologies (network analysis). Precision cardiovascular medicine will only be broadly applied to cardiovascular patients once this comprehensive data set is subjected to unique, integrative analytical strategies that accommodate molecular and clinical heterogeneity rather than ignore or reduce it.
Collapse
|
117
|
Li Q, Meng L, Liu D. Screening and Identification of Therapeutic Targets for Pulmonary Arterial Hypertension Through Microarray Technology. Front Genet 2020; 11:782. [PMID: 32849793 PMCID: PMC7396553 DOI: 10.3389/fgene.2020.00782] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare but fatal disease characterized by vascular cell proliferation; the pathogenesis of PAH has yet to be fully elucidated. Publicly available genetic data were downloaded from the Gene Expression Omnibus (GEO) database, and gene set enrichment analysis (GSEA) was used to determine significant differences in gene expression between tissues with PAH and healthy lung tissues. Differentially expressed genes (DEGs) were identified using the online tool, GEO2R, and functional annotation of DEGs was performed using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Next, the construction and module analysis of the protein–protein interaction (PPI) network and verification of the expression level of hub genes was performed. Finally, prediction and enrichment analysis of microRNAs associated with the hub genes was carried out. A total of 110 DEGs were detected by screening PAH and healthy lung samples. The expression of nine genes [polo-like kinase 4 (PLK4), centromere protein U, kinesin family member 20B, structural maintenance of chromosome 2 (SMC2), abnormal spindle microtubule assembly, Fanconi Anemia complementation group I, kinesin family member 18A, spindle apparatus coiled-coil protein 1, and MIS18 binding protein 1] was elevated in PAH; this was statistically significant compared with their expression in healthy lung tissue, and they were identified as hub genes. GO and KEGG analysis showed that the variations in DEGs were abundant in DNA-templated transcription, sister chromatid cohesion, mitotic nuclear division, cell proliferation, and regulation of the actin cytoskeleton. In conclusion, this study has successfully identified hub genes and key pathways of PAH, with a total of 110 DEGs and nine hub genes related to PAH, especially the PLK4 and SMC2 genes, thus providing important clues for the in-depth understanding of the molecular mechanism of PAH and providing potential therapeutic targets.
Collapse
Affiliation(s)
- Qing Li
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - LingBing Meng
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Departments of Cardiology, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - DePing Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
118
|
Shi XF, Su YC. Vascular Metabolic Mechanisms of Pulmonary Hypertension. Curr Med Sci 2020; 40:444-454. [PMID: 32681249 DOI: 10.1007/s11596-020-2198-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Pulmonary hypertension (PH) is a severe and progressive disease characterized by increased pulmonary vascular resistance leading to right heart failure and death. In PH, the cellular metabolisms including those of the three major nutrients (carbohydrate, lipid and protein) are aberrant in pulmonary vascular cells. Glucose uptake, glycolysis, insulin resistance, sphingolipid S1P, PGE2, TXA2, leukotrienes and glutaminolysis are upregulated, and phospholipid-prostacyclin and L-arginine-nitric oxide pathway are compromised in lung vascular cells. Fatty acid metabolism is disordered in lung endothelial cells and smooth muscle cells. These molecular mechanisms are integrated to promote PH-specific abnormal vascular cell proliferation and vascular remodeling. This review summarizes the recent advances in the metabolic reprogramming of glucose, fatty acid, and amino acid metabolism in pulmonary vascular remodeling in PH and the mechanisms for how these alterations affect vascular cell fate and impact the course of PH.
Collapse
Affiliation(s)
- Xiao-Fan Shi
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA, 30912, USA
| | - Yun-Chao Su
- Department of Pharmacology & Toxicology, Augusta University, Augusta, GA, 30912, USA. .,Department of Medicine, Augusta University, Augusta, GA, 30912, USA. .,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
119
|
Cool CD, Kuebler WM, Bogaard HJ, Spiekerkoetter E, Nicolls MR, Voelkel NF. The hallmarks of severe pulmonary arterial hypertension: the cancer hypothesis-ten years later. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1115-L1130. [PMID: 32023082 PMCID: PMC9847334 DOI: 10.1152/ajplung.00476.2019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/25/2023] Open
Abstract
Severe forms of pulmonary arterial hypertension (PAH) are most frequently the consequence of a lumen-obliterating angiopathy. One pathobiological model is that the initial pulmonary vascular endothelial cell injury and apoptosis is followed by the evolution of phenotypically altered, apoptosis-resistant, proliferating cells and an inflammatory vascular immune response. Although there may be a vasoconstrictive disease component, the increased pulmonary vascular shear stress in established PAH is caused largely by the vascular wall pathology. In this review, we revisit the "quasi-malignancy concept" of severe PAH and examine to what extent the hallmarks of PAH can be compared with the hallmarks of cancer. The cancer model of severe PAH, based on the growth of abnormal vascular and bone marrow-derived cells, may enable the emergence of novel cell-based PAH treatment strategies.
Collapse
Affiliation(s)
- Carlyne D Cool
- Department of Pathology, University of Colorado, Anschuetz Campus, Aurora, Colorado
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitaetsmedizin, Berlin, Germany
| | - Harm Jan Bogaard
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, California
| | - Norbert F Voelkel
- Amsterdam University Medical Centers, Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
120
|
Zhang H, Brown RD, Stenmark KR, Hu CJ. RNA-Binding Proteins in Pulmonary Hypertension. Int J Mol Sci 2020; 21:ijms21113757. [PMID: 32466553 PMCID: PMC7312837 DOI: 10.3390/ijms21113757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening disease characterized by significant vascular remodeling and aberrant expression of genes involved in inflammation, apoptosis resistance, proliferation, and metabolism. Effective therapeutic strategies are limited, as mechanisms underlying PH pathophysiology, especially abnormal expression of genes, remain unclear. Most PH studies on gene expression have focused on gene transcription. However, post-transcriptional alterations have been shown to play a critical role in inflammation and metabolic changes in diseases such as cancer and systemic cardiovascular diseases. In these diseases, RNA-binding proteins (RBPs) have been recognized as important regulators of aberrant gene expression via post-transcriptional regulation; however, their role in PH is less clear. Identifying RBPs in PH is of great importance to better understand PH pathophysiology and to identify new targets for PH treatment. In this manuscript, we review the current knowledge on the role of dysregulated RBPs in abnormal mRNA gene expression as well as aberrant non-coding RNA processing and expression (e.g., miRNAs) in PH.
Collapse
Affiliation(s)
- Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.Z.); (R.D.B.); (K.R.S.)
| | - R. Dale Brown
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.Z.); (R.D.B.); (K.R.S.)
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.Z.); (R.D.B.); (K.R.S.)
| | - Cheng-Jun Hu
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.Z.); (R.D.B.); (K.R.S.)
- Department of Craniofacial Biology School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Correspondence: ; Tel.: +1-303-724-4576; Fax: +1-303-724-4580
| |
Collapse
|
121
|
Novel Molecular Mechanisms of Pulmonary Hypertension: A Search for Biomarkers and Novel Drug Targets-From Bench to Bed Site. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7265487. [PMID: 32566097 PMCID: PMC7261339 DOI: 10.1155/2020/7265487] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/11/2020] [Indexed: 12/23/2022]
Abstract
Pulmonary hypertension (PH) is defined as increased mean pulmonary artery pressure (mPAP) above 25 mmHg, measured at rest by right heart catheterization. The exact global prevalence of PH is difficult to estimate, mainly due to the complex aetiology, and its spread may be underestimated. To date, numerous studies on the aetiology and pathophysiology of PH at molecular level were conducted. Simultaneously, some clinical studies have shown potential usefulness of well-known and widely recognized cardiovascular biomarkers, but their potential clinical usefulness in diagnosis and management of PH is poor due to their low specificity accompanied with numerous other cardiovascular comorbidities of PH subjects. On the other hand, a large body of basic research-based studies provides us with novel molecular pathomechanisms, biomarkers, and drug targets, according to the evidence-based medicine principles. Unfortunately, the simple implementation of these results to clinical practice is impossible due to a large heterogeneity of the PH pathophysiology, where the clinical symptoms constitute only a common denominator and a final result of numerous crosstalking metabolic pathways. Therefore, future studies, based mostly on translational medicine, are needed in order to both organize better the pathophysiological classification of various forms of PH and define precisely the optimal diagnostic markers and therapeutic targets in particular forms of PH. This review paper summarizes the current state of the art regarding the molecular background of PH with respect to its current classification. Novel therapeutic strategies and potential biomarkers are discussed with respect to their limitations in use in common clinical practice.
Collapse
|
122
|
Luo L, Xiao L, Lian G, Wang H, Xie L. miR-125a-5p inhibits glycolysis by targeting hexokinase-II to improve pulmonary arterial hypertension. Aging (Albany NY) 2020; 12:9014-9030. [PMID: 32427576 PMCID: PMC7288947 DOI: 10.18632/aging.103163] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Purpose: The aim of this study was to investigate the effect of microRNAs on the proliferation of pulmonary arterial smooth muscle cells (PASMCs) as a result of targeting hexokinase-II (HK-II) and its mechanism of action. Results: Differences in metabolic patterns were found between the normal group and monocrotaline-induced pulmonary arterial hypertension (MCT-PH) group. miR-125a-5p decreased glycolysis levels of monocrotaline (MCT)-induced PASMCs by targeting HK-II and inhibiting its proliferation. In vivo experiments found that miR-125a-5p agomir upregulated HK-II expression in the MCT-PH. Right ventricular hypertrophy was reversed and cardiac function improved as a result of decreased mean pulmonary artery pressure (mPAP). Conclusion: In vitro and in vivo experiments both confirmed that miR-125a-5p could inhibit cell glycolysis and PASMC proliferation to improve PAH by targeting HK-II. Methods: HK-II overexpression was constructed, and differentially expressed microRNAs were screened for using microarrays. Serum metabolites were detected using Nuclear Magnetic Resonance (NMR). Through screening for characteristic metabolites in rat body fluids and by analyzing biological functions, disordered metabolic pathways were identified. Activity of the miR-125a-5p target HK-II was measured using a luciferase reporter assay. Expression of downstream molecules was measured by RT–qPCR and/or western blot. Glucose consumption and lactic acid production were analyzed and used as a reflection of glycolysis.
Collapse
Affiliation(s)
- Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | | | - Guili Lian
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Huajun Wang
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Department of General Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
123
|
Benabid A, Peduto L. Mesenchymal perivascular cells in immunity and disease. Curr Opin Immunol 2020; 64:50-55. [PMID: 32387900 PMCID: PMC7597593 DOI: 10.1016/j.coi.2020.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 12/13/2022]
Abstract
The mesenchymal microenvironment is increasingly recognized as a major player in immunity. Here we focus on mesenchymal cells located within or in proximity to the blood vessels wall, which include pericytes, adventitial fibroblasts and mesenchymal stromal cells. We discuss recent evidence that these cells play a role in tissue homeostasis, immunity and inflammatory pathologies by multiple mechanisms, including vascular modulation, leucocyte migration, activation or survival in the perivascular space and differentiation into specialized 'effector' mesenchymal cells essential for tissue repair and immunity, such as myofibroblasts and lymphoid stromal cells. When dysregulated, these responses contribute to inflammatory and fibrotic diseases.
Collapse
Affiliation(s)
- Adam Benabid
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Inserm U1224, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Lucie Peduto
- Stroma, Inflammation & Tissue Repair Unit, Institut Pasteur, Inserm U1224, Paris, France.
| |
Collapse
|
124
|
Frid MG, Thurman JM, Hansen KC, Maron BA, Stenmark KR. Inflammation, immunity, and vascular remodeling in pulmonary hypertension; Evidence for complement involvement? Glob Cardiol Sci Pract 2020; 2020:e202001. [PMID: 32478115 PMCID: PMC7232865 DOI: 10.21542/gcsp.2020.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 03/28/2020] [Indexed: 12/15/2022] Open
Abstract
Pulmonary (arterial) hypertension (PH/PAH) is a life-threatening cardiopulmonary disorder. Experimental evidence suggests involvement of inflammatory and autoimmune processes in pathogenesis of PH/PAH, however the triggering and disease-promoting mechanisms remain unknown. The complement system is a key arm of innate immunity implicated in various pro-inflammatory and autoimmune diseases, yet, surprisingly little is known about the role of complement in PH/PAH pathogenesis. The preponderance of the existing data associates complement with PH/PAH via analysis of plasma and does not study the lung directly. Therefore, we aimed to resolve this by analyzing both the mechanisms of local lung-specific complement activation and the correlation of dysregulated plasma complement to clinical outcome in PAH patients. In our recent studies, reviewed herein, we show, for the first time, that immunoglobulin-driven activation of the complement cascade, specifically its alternative pathway, in the pulmonary perivascular areas, is a key mechanism initiating pro-inflammatory processes in the early stage of experimental hypoxic PH (a form of "sterile inflammation"). In human patients with end-stage PAH, we have demonstrated that perivascular deposition of immunoglobulin G (IgG) and activation of the complement cascade are "longitudinally" persistent in the disease. We also showed, using unbiased network analysis, that plasma complement signaling, including again the Alternative pathway, is a prognostic factor of survival in patients with idiopathic PAH (IPAH). Based on these initial findings, we suggest that vascular-specific, immunoglobulin-driven dysregulated complement signaling triggers and maintains pulmonary vascular remodeling and PH. Future experiments in this area would facilitate discoveries on whether complement signaling can serve both as a biomarker and therapeutic target in PH/PAH.
Collapse
Affiliation(s)
- Maria G. Frid
- University of Colorado, Denver, Anschutz Medical Campus, USA
| | | | - Kirk C. Hansen
- University of Colorado, Denver, Anschutz Medical Campus, USA
| | | | | |
Collapse
|
125
|
Harbaum L, Rhodes CJ, Otero-Núñez P, Wharton J, Wilkins MR. The application of 'omics' to pulmonary arterial hypertension. Br J Pharmacol 2020; 178:108-120. [PMID: 32201940 DOI: 10.1111/bph.15056] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 01/03/2020] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
Recent genome-wide analyses of rare and common sequence variations have brought greater clarity to the genetic architecture of pulmonary arterial hypertension and implicated novel genes in disease development. Transcriptional signatures have been reported in whole lung tissue, pulmonary vascular cells and peripheral circulating cells. High-throughput platforms for plasma proteomics and metabolomics have identified novel biomarkers associated with clinical outcomes and provided molecular instruments for risk assessment. There are methodological challenges to integrating these datasets, coupled to statistical power limitations inherent to the study of a rare disease, but the expectation is that this approach will reveal novel druggable targets and biomarkers that will open the way to personalized medicine. Here, we review the current state-of-the-art and future promise of 'omics' in the field of translational medicine in pulmonary arterial hypertension. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.1/issuetoc.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Pablo Otero-Núñez
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
126
|
Le S, Zhang H, Huang X, Chen S, Wu J, Chen S, Ding X, Chen S, Zhao J, Xu H, Cui J, Zou Y, Yu J, Jiang L, Wu J, Ye P, Xia J. PKM2 Activator TEPP-46 Attenuates Thoracic Aortic Aneurysm and Dissection by Inhibiting NLRP3 Inflammasome-Mediated IL-1β Secretion. J Cardiovasc Pharmacol Ther 2020; 25:364-376. [PMID: 32323562 DOI: 10.1177/1074248420919966] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND The development of thoracic aortic aneurysm and dissection (TAAD) is mediated by inflammasome activation, which exacerbates the secretion of pro-inflammatory cytokines, chemokines, matrix metalloproteinases (MMPs), and reactive oxygen species (ROS). The glycolytic enzyme pyruvate kinase M2 (PKM2) has shown a protective role against various disorders with an inflammatory basis, such as sepsis, tumorigenesis, and diabetic nephropathy. However, its potential role in TAAD has not been investigated so far. APPROACH AND RESULTS We analyzed aortic tissues from TAAD patients and the β-aminopropionitrile fumarate (BAPN)-induced mouse model of TAAD and observed elevated levels of PKM2 in the aortic lesions of both. Treatment with the PKM2 activator TEPP-46 markedly attenuated the progression of TAAD in the mouse model as demonstrated by decreased morbidity and luminal diameter of the aorta. In addition, the thoracic aortas of the BAPN-induced mice showed reduced monocytes and macrophages infiltration and lower levels of IL-1β, MMPs, and ROS when treated with TEPP-46. Furthermore, TEPP-46 treatment also suppressed the activation of the NOD-like receptor (NLR) family and pyrin domain-containing protein 3 (NLRP3) inflammasome by downregulating p-STAT3 and HIF1-α. CONCLUSION Pyruvate kinase M2 plays a protective role in TAAD development, and its activation is a promising therapeutic strategy against the progression of TAAD.
Collapse
Affiliation(s)
- Sheng Le
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The authers Sheng Le and Hao Zhang contributed equally to this article as first authors
| | - Hao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The authers Sheng Le and Hao Zhang contributed equally to this article as first authors
| | - Xiaofan Huang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Wu
- Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Chen
- Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - XiangChao Ding
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jikai Cui
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqiang Zou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lang Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Ye
- Department of Cardiology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
127
|
Joshi SR, Kitagawa A, Jacob C, Hashimoto R, Dhagia V, Ramesh A, Zheng C, Zhang H, Jordan A, Waddell I, Leopold J, Hu CJ, McMurtry IF, D'Alessandro A, Stenmark KR, Gupte SA. Hypoxic activation of glucose-6-phosphate dehydrogenase controls the expression of genes involved in the pathogenesis of pulmonary hypertension through the regulation of DNA methylation. Am J Physiol Lung Cell Mol Physiol 2020; 318:L773-L786. [PMID: 32159369 PMCID: PMC7191486 DOI: 10.1152/ajplung.00001.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is considered important in the pathogenesis of the occlusive vasculopathy observed in pulmonary hypertension (PH). However, the mechanisms that link reprogrammed metabolism to aberrant expression of genes, which modulate functional phenotypes of cells in PH, remain enigmatic. Herein, we demonstrate that, in mice, hypoxia-induced PH was prevented by glucose-6-phosphate dehydrogenase deficiency (G6PDDef), and further show that established severe PH in Cyp2c44-/- mice was attenuated by knockdown with G6PD shRNA or by G6PD inhibition with an inhibitor (N-ethyl-N'-[(3β,5α)-17-oxoandrostan-3-yl]urea, NEOU). Mechanistically, G6PDDef, knockdown and inhibition in lungs: 1) reduced hypoxia-induced changes in cytoplasmic and mitochondrial metabolism, 2) increased expression of Tet methylcytosine dioxygenase 2 (Tet2) gene, and 3) upregulated expression of the coding genes and long noncoding (lnc) RNA Pint, which inhibits cell growth, by hypomethylating the promoter flanking region downstream of the transcription start site. These results suggest functional TET2 is required for G6PD inhibition to increase gene expression and to reverse hypoxia-induced PH in mice. Furthermore, the inhibitor of G6PD activity (NEOU) decreased metabolic reprogramming, upregulated TET2 and lncPINT, and inhibited growth of control and diseased smooth muscle cells isolated from pulmonary arteries of normal individuals and idiopathic-PAH patients, respectively. Collectively, these findings demonstrate a previously unrecognized function for G6PD as a regulator of DNA methylation. These findings further suggest that G6PD acts as a link between reprogrammed metabolism and aberrant gene regulation and plays a crucial role in regulating the phenotype of cells implicated in the pathogenesis of PH, a debilitating disorder with a high mortality rate.
Collapse
Affiliation(s)
| | - Atsushi Kitagawa
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Christina Jacob
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Ryota Hashimoto
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Amrit Ramesh
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Connie Zheng
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Hui Zhang
- Division of Pediatric Critical Care Medicine, Cardiovascular Pulmonary Research and Developmental Lung Biology Laboratories, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Allan Jordan
- Drug Discovery Unit, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Ian Waddell
- Drug Discovery Unit, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Jane Leopold
- Department of Medicine, Division of Cardiology, Brigham Women and Children's Hospital, Harvard School of Medicine, Boston, Massachusetts
| | - Cheng-Jun Hu
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ivan F McMurtry
- Departments of Pharmacology and Internal Medicine and Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Division of Pediatric Critical Care Medicine, Cardiovascular Pulmonary Research and Developmental Lung Biology Laboratories, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sachin A Gupte
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
128
|
Tian L, Wu D, Dasgupta A, Chen KH, Mewburn J, Potus F, Lima PDA, Hong Z, Zhao YY, Hindmarch CCT, Kutty S, Provencher S, Bonnet S, Sutendra G, Archer SL. Epigenetic Metabolic Reprogramming of Right Ventricular Fibroblasts in Pulmonary Arterial Hypertension: A Pyruvate Dehydrogenase Kinase-Dependent Shift in Mitochondrial Metabolism Promotes Right Ventricular Fibrosis. Circ Res 2020; 126:1723-1745. [PMID: 32216531 DOI: 10.1161/circresaha.120.316443] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Right ventricular (RV) fibrosis in pulmonary arterial hypertension contributes to RV failure. While RV fibrosis reflects changes in the function of resident RV fibroblasts (RVfib), these cells are understudied. OBJECTIVE Examine the role of mitochondrial metabolism of RVfib in RV fibrosis in human and experimental pulmonary arterial hypertension. METHODS AND RESULTS Male Sprague-Dawley rats received monocrotaline (MCT; 60 mg/kg) or saline. Drinking water containing no supplement or the PDK (pyruvate dehydrogenase kinase) inhibitor dichloroacetate was started 7 days post-MCT. At week 4, treadmill testing, echocardiography, and right heart catheterization were performed. The effects of PDK activation on mitochondrial dynamics and metabolism, RVfib proliferation, and collagen production were studied in RVfib in cell culture. Epigenetic mechanisms for persistence of the profibrotic RVfib phenotype in culture were evaluated. PDK expression was also studied in the RVfib of patients with decompensated RV failure (n=11) versus control (n=7). MCT rats developed pulmonary arterial hypertension, RV fibrosis, and RV failure. MCT-RVfib (but not left ventricular fibroblasts) displayed excess mitochondrial fission and had increased expression of PDK isoforms 1 and 3 that persisted for >5 passages in culture. PDK-mediated decreases in pyruvate dehydrogenase activity and oxygen consumption rate were reversed by dichloroacetate (in RVfib and in vivo) or siRNA targeting PDK 1 and 3 (in RVfib). These interventions restored mitochondrial superoxide and hydrogen peroxide production and inactivated HIF (hypoxia-inducible factor)-1α, which was pathologically activated in normoxic MCT-RVfib. Redox-mediated HIF-1α inactivation also decreased the expression of TGF-β1 (transforming growth factor-beta-1) and CTGF (connective tissue growth factor), reduced fibroblast proliferation, and decreased collagen production. HIF-1α activation in MCT-RVfib reflected increased DNMT (DNA methyltransferase) 1 expression, which was associated with a decrease in its regulatory microRNA, miR-148b-3p. In MCT rats, dichloroacetate, at therapeutic levels in the RV, reduced phospho-pyruvate dehydrogenase expression, RV fibrosis, and hypertrophy and improved RV function. In patients with pulmonary arterial hypertension and RV failure, RVfib had increased PDK1 expression. CONCLUSIONS MCT-RVfib manifest a DNMT1-HIF-1α-PDK-mediated, chamber-specific, metabolic memory that promotes collagen production and RV fibrosis. This epigenetic mitochondrial-metabolic pathway is a potential antifibrotic therapeutic target.
Collapse
Affiliation(s)
- Lian Tian
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Danchen Wu
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Asish Dasgupta
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Kuang-Hueih Chen
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Jeffrey Mewburn
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Francois Potus
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Patricia D A Lima
- Queen'ps CardioPulmonary Unit, Department of Medicine, Translational Institute of Medicine (P.D.A.L., C.C.T.H., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Zhigang Hong
- Department of Pharmacology, University of Illinois at Chicago (Z.H.)
| | - Yuan-Yuan Zhao
- Department of Agricultural, Food and Nutritional Science (Y.-Y.Z.), University of Alberta, Edmonton, Canada
| | - Charles C T Hindmarch
- Queen'ps CardioPulmonary Unit, Department of Medicine, Translational Institute of Medicine (P.D.A.L., C.C.T.H., S.L.A.), Queen's University, Kingston, Ontario, Canada
| | - Shelby Kutty
- Department of Medicine, John Hopkins University, Baltimore, MD (S.K.)
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Department of Medicine, Heart and Lung Institute of Quebec, Laval University, Canada (S.P., S.B.)
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Department of Medicine, Heart and Lung Institute of Quebec, Laval University, Canada (S.P., S.B.)
| | - Gopinath Sutendra
- Department of Medicine (G.S.), University of Alberta, Edmonton, Canada
| | - Stephen L Archer
- From the Department of Medicine (L.T., D.W., A.D., K.-H.C., J.M., F.P., S.L.A.), Queen's University, Kingston, Ontario, Canada.,Queen'ps CardioPulmonary Unit, Department of Medicine, Translational Institute of Medicine (P.D.A.L., C.C.T.H., S.L.A.), Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
129
|
Tuder RM, Stenmark KR. Perspective: pathobiological paradigms in pulmonary hypertension, time for reappraisal. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1131-L1137. [PMID: 32186206 DOI: 10.1152/ajplung.00067.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
For the past 120 years, there has been a progressive evolution of the pathobiological concepts underlying pulmonary hypertension. Conceptual frameworks, build around the paradigms of excessive vasoconstriction (vs. vasodilation) and, more recently, of the cancer-like hypothesis of pulmonary hypertension, have served to consolidate key discoveries; moreover, they have and continue contributing to innovative advances that have been translated into either successful or potential new therapies. However, those frameworks do not fully address the complexity and challenges facing pulmonary hypertension, particularly those involving the marked heterogeneity of disease presentation and the dynamic changes occurring over time in affected tissues and cells. This is particularly relevant in regards to the molecular pathways of pulmonary hypertension; the ever growing understanding of molecular and cellular pathways requires clarification if they drive distinctive pulmonary vascular lesions in a given lung and disease patients with the same group pulmonary hypertension. Novel methodologies and approaches can start dissecting this key challenge in the field as it is critical to address the key angle of heterogeneity of the disease and reappraisal of disease-modifying therapies.
Collapse
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, Aurora, Colorado
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Division of Critical Care Medicine, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
130
|
Dasgupta A, Wu D, Tian L, Xiong PY, Dunham-Snary KJ, Chen KH, Alizadeh E, Motamed M, Potus F, Hindmarch CCT, Archer SL. Mitochondria in the Pulmonary Vasculature in Health and Disease: Oxygen-Sensing, Metabolism, and Dynamics. Compr Physiol 2020; 10:713-765. [PMID: 32163206 DOI: 10.1002/cphy.c190027] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In lung vascular cells, mitochondria serve a canonical metabolic role, governing energy homeostasis. In addition, mitochondria exist in dynamic networks, which serve noncanonical functions, including regulation of redox signaling, cell cycle, apoptosis, and mitochondrial quality control. Mitochondria in pulmonary artery smooth muscle cells (PASMC) are oxygen sensors and initiate hypoxic pulmonary vasoconstriction. Acquired dysfunction of mitochondrial metabolism and dynamics contribute to a cancer-like phenotype in pulmonary arterial hypertension (PAH). Acquired mitochondrial abnormalities, such as increased pyruvate dehydrogenase kinase (PDK) and pyruvate kinase muscle isoform 2 (PKM2) expression, which increase uncoupled glycolysis (the Warburg phenomenon), are implicated in PAH. Warburg metabolism sustains energy homeostasis by the inhibition of oxidative metabolism that reduces mitochondrial apoptosis, allowing unchecked cell accumulation. Warburg metabolism is initiated by the induction of a pseudohypoxic state, in which DNA methyltransferase (DNMT)-mediated changes in redox signaling cause normoxic activation of HIF-1α and increase PDK expression. Furthermore, mitochondrial division is coordinated with nuclear division through a process called mitotic fission. Increased mitotic fission in PAH, driven by increased fission and reduced fusion favors rapid cell cycle progression and apoptosis resistance. Downregulation of the mitochondrial calcium uniporter complex (MCUC) occurs in PAH and is one potential unifying mechanism linking Warburg metabolism and mitochondrial fission. Mitochondrial metabolic and dynamic disorders combine to promote the hyperproliferative, apoptosis-resistant, phenotype in PAH PASMC, endothelial cells, and fibroblasts. Understanding the molecular mechanism regulating mitochondrial metabolism and dynamics has permitted identification of new biomarkers, nuclear and CT imaging modalities, and new therapeutic targets for PAH. © 2020 American Physiological Society. Compr Physiol 10:713-765, 2020.
Collapse
Affiliation(s)
- Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Lian Tian
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Ping Yu Xiong
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | | | - Kuang-Hueih Chen
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Elahe Alizadeh
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Mehras Motamed
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - François Potus
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Department of Medicine, Queen's Cardiopulmonary Unit (QCPU), Translational Institute of Medicine (TIME), Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.,Kingston Health Sciences Centre, Kingston, Ontario, Canada.,Providence Care Hospital, Kingston, Ontario, Canada
| |
Collapse
|
131
|
Saygin D, Tabib T, Bittar HET, Valenzi E, Sembrat J, Chan SY, Rojas M, Lafyatis R. Transcriptional profiling of lung cell populations in idiopathic pulmonary arterial hypertension. Pulm Circ 2020; 10:10.1177_2045894020908782. [PMID: 32166015 PMCID: PMC7052475 DOI: 10.1177/2045894020908782] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Despite recent improvements in management of idiopathic pulmonary arterial
hypertension, mortality remains high. Understanding the alterations in the
transcriptome–phenotype of the key lung cells involved could provide insight
into the drivers of pathogenesis. In this study, we examined differential gene
expression of cell types implicated in idiopathic pulmonary arterial
hypertension from lung explants of patients with idiopathic pulmonary arterial
hypertension compared to control lungs. After tissue digestion, we analyzed all
cells from three idiopathic pulmonary arterial hypertension and six control
lungs using droplet-based single cell RNA-sequencing. After dimensional
reduction by t-stochastic neighbor embedding, we compared the transcriptomes of
endothelial cells, pericyte/smooth muscle cells, fibroblasts, and macrophage
clusters, examining differential gene expression and pathways implicated by
analysis of Gene Ontology Enrichment. We found that endothelial cells and
pericyte/smooth muscle cells had the most differentially expressed gene profile
compared to other cell types. Top differentially upregulated genes in
endothelial cells included novel genes: ROBO4, APCDD1, NDST1, MMRN2,
NOTCH4, and DOCK6, as well as previously reported
genes: ENG, ORAI2, TFDP1, KDR, AMOTL2, PDGFB, FGFR1, EDN1, and
NOTCH1. Several transcription factors were also found to be
upregulated in idiopathic pulmonary arterial hypertension endothelial cells
including SOX18, STRA13, LYL1, and ELK, which
have known roles in regulating endothelial cell phenotype. In particular,
SOX18 was implicated through bioinformatics analyses in
regulating the idiopathic pulmonary arterial hypertension endothelial cell
transcriptome. Furthermore, idiopathic pulmonary arterial hypertension
endothelial cells upregulated expression of FAM60A and
HDAC7, potentially affecting epigenetic changes in
idiopathic pulmonary arterial hypertension endothelial cells. Pericyte/smooth
muscle cells expressed genes implicated in regulation of cellular apoptosis and
extracellular matrix organization, and several ligands for genes showing
increased expression in endothelial cells. In conclusion, our study represents
the first detailed look at the transcriptomic landscape across idiopathic
pulmonary arterial hypertension lung cells and provides robust insight into
alterations that occur in vivo in idiopathic pulmonary arterial hypertension
lungs.
Collapse
Affiliation(s)
- Didem Saygin
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Tracy Tabib
- Division of Rheumatology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Humberto E T Bittar
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Eleanor Valenzi
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Stephen Y Chan
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
132
|
Stable isotope metabolomics of pulmonary artery smooth muscle and endothelial cells in pulmonary hypertension and with TGF-beta treatment. Sci Rep 2020; 10:413. [PMID: 31942023 PMCID: PMC6962446 DOI: 10.1038/s41598-019-57200-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
Altered metabolism in pulmonary artery smooth muscle cells (PASMCs) and endothelial cells (PAECs) contributes to the pathology of pulmonary hypertension (PH), but changes in substrate uptake and how substrates are utilized have not been fully characterized. We hypothesized stable isotope metabolomics would identify increased glucose, glutamine and fatty acid uptake and utilization in human PASMCs and PAECs from PH versus control specimens, and that TGF-β treatment would phenocopy these metabolic changes. We used 13C-labeled glucose, glutamine or a long-chain fatty acid mixture added to cell culture media, and mass spectrometry-based metabolomics to detect and quantify 13C-labeled metabolites. We found PH PASMCs had increased glucose uptake and utilization by glycolysis and the pentose shunt, but no changes in glutamine or fatty acid uptake or utilization. Diseased PAECs had increased proximate glycolysis pathway intermediates, less pentose shunt flux, increased anaplerosis from glutamine, and decreased fatty acid β-oxidation. TGF-β treatment increased glycolysis in PASMCs, but did not recapitulate the PAEC disease phenotype. In TGF-β-treated PASMCs, glucose, glutamine and fatty acids all contributed carbons to the TCA cycle. In conclusion, PASMCs and PAECs collected from PH subjects have significant changes in metabolite uptake and utilization, partially recapitulated by TGF-β treatment.
Collapse
|
133
|
Napoli C, Benincasa G, Loscalzo J. Epigenetic Inheritance Underlying Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2020; 39:653-664. [PMID: 30727752 DOI: 10.1161/atvbaha.118.312262] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In pulmonary arterial hypertension (PAH), the Warburg effect (glycolytic shift) and mitochondrial fission are determinants of phenotype alterations characteristic of the disease, such as proliferation, apoptosis resistance, migration, endothelial-mesenchymal transition, and extracellular matrix stiffness. Current therapies, focusing largely on vasodilation and antithrombotic protection, do not restore these aberrant phenotypes suggesting that additional pathways need be targeted. The multifactorial nature of PAH suggests epigenetic changes as potential determinants of vascular remodeling. Transgenerational epigenetic changes induced by hypoxia can result in permanent changes early in fetal development increasing PAH risk in adulthood. Unlike genetic mutations, epigenetic changes are pharmacologically reversible, making them an attractive target as therapeutic strategies for PAH. This review offers a landscape of the most current clinical, epigenetic-sensitive changes contributing to PAH vascular remodeling both in early and later life, with a focus on a network medicine strategy. Furthermore, we discuss the importance of the application (from morphogenesis to disease onset) of molecular network-based algorithms to dissect PAH molecular pathobiology. Additionally, we suggest an integrated network-based program for clinical disease gene discovery that may reveal novel biomarkers and novel disease targets, thus offering a truly innovative path toward redefining and treating PAH, as well as facilitating the trajectory of a comprehensive precision medicine approach to PAH.
Collapse
Affiliation(s)
- Claudio Napoli
- From the Department of Medical, Surgical, Neurological, Metabolic, and Geriatric Sciences (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units AOU (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giuditta Benincasa
- From the Department of Medical, Surgical, Neurological, Metabolic, and Geriatric Sciences (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units AOU (C.N., G.B.), University of Campania Luigi Vanvitelli, Naples, Italy
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.)
| |
Collapse
|
134
|
Prins KW, Thenappan T, Weir EK, Kalra R, Pritzker M, Archer SL. Repurposing Medications for Treatment of Pulmonary Arterial Hypertension: What's Old Is New Again. J Am Heart Assoc 2020; 8:e011343. [PMID: 30590974 PMCID: PMC6405714 DOI: 10.1161/jaha.118.011343] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kurt W Prins
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Thenappan Thenappan
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - E Kenneth Weir
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Rajat Kalra
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | - Marc Pritzker
- 1 Cardiovascular Division University of Minnesota Medical School Minneapolis MN
| | | |
Collapse
|
135
|
Bisserier M, Janostiak R, Lezoualc’h F, Hadri L. Targeting epigenetic mechanisms as an emerging therapeutic strategy in pulmonary hypertension disease. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2020; 2:R17-R34. [PMID: 32161845 PMCID: PMC7065685 DOI: 10.1530/vb-19-0030] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial cardiopulmonary disease characterized by an elevation of pulmonary artery pressure (PAP) and pulmonary vascular resistance (PVR), which can lead to right ventricular (RV) failure, multi-organ dysfunction, and ultimately to premature death. Despite the advances in molecular biology, the mechanisms underlying pulmonary hypertension (PH) remain unclear. Nowadays, there is no curative treatment for treating PH. Therefore, it is crucial to identify novel, specific therapeutic targets and to offer more effective treatments against the progression of PH. Increasing amounts of evidence suggest that epigenetic modification may play a critical role in the pathogenesis of PAH. In the presented paper, we provide an overview of the epigenetic mechanisms specifically, DNA methylation, histone acetylation, histone methylation, and ncRNAs. As the recent identification of new pharmacological drugs targeting these epigenetic mechanisms has opened new therapeutic avenues, we also discuss the importance of epigenetic-based therapies in the context of PH.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Frank Lezoualc’h
- Inserm, UMR-1048, Institut des Maladies Métaboliques et Cardiovasculaires, University of Toulouse, Toulouse Cedex 4, France
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
136
|
Bhedi CD, Nasirova S, Toksoz D, Warburton RR, Morine KJ, Kapur NK, Galper JB, Preston IR, Hill NS, Fanburg BL, Penumatsa KC. Glycolysis regulated transglutaminase 2 activation in cardiopulmonary fibrogenic remodeling. FASEB J 2020; 34:930-944. [PMID: 31914588 PMCID: PMC6956703 DOI: 10.1096/fj.201902155r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/22/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022]
Abstract
The pathophysiology of pulmonary hypertension (PH) and heart failure (HF) includes fibrogenic remodeling associated with the loss of pulmonary arterial (PA) and cardiac compliance. We and others have previously identified transglutaminase 2 (TG2) as a participant in adverse fibrogenic remodeling. However, little is known about the biologic mechanisms that regulate TG2 function. We examined physiological mouse models of experimental PH, HF, and type 1 diabetes that are associated with altered glucose metabolism/glycolysis and report here that TG2 expression and activity are elevated in pulmonary and cardiac tissues under all these conditions. We additionally used PA adventitial fibroblasts to test the hypothesis that TG2 is an intermediary between enhanced tissue glycolysis and fibrogenesis. Our in vitro results show that glycolytic enzymes and TG2 are upregulated in fibroblasts exposed to high glucose, which stimulates cellular glycolysis as measured by Seahorse analysis. We examined the relationship of TG2 to a terminal glycolytic enzyme, pyruvate kinase M2 (PKM2), and found that PKM2 regulates glucose-induced TG2 expression and activity as well as fibrogenesis. Our studies further show that TG2 inhibition blocks glucose-induced fibrogenesis and cell proliferation. Our findings support a novel role for glycolysis-mediated TG2 induction and tissue fibrosis associated with experimental PH, HF, and hyperglycemia.
Collapse
Affiliation(s)
- Chinmayee D. Bhedi
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Sabina Nasirova
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Deniz Toksoz
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Rod R. Warburton
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Kevin J. Morine
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Navin K. Kapur
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Jonas B. Galper
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Ioana R. Preston
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Nicholas S. Hill
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Barry L. Fanburg
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Krishna C. Penumatsa
- Pulmonary Division, Department of Medicine, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
137
|
Novoyatleva T, Rai N, Weissmann N, Grimminger F, Ghofrani HA, Seeger W, Gall H, Schermuly RT. Is PKM2 Phosphorylation a Prerequisite for Oligomer Disassembly in Pulmonary Arterial Hypertension? Am J Respir Crit Care Med 2019; 200:1550-1554. [PMID: 31486671 DOI: 10.1164/rccm.201904-0782le] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | - Nabham Rai
- Justus Liebig University GiessenGiessen, Germanyand
| | | | | | | | - Werner Seeger
- Justus Liebig University GiessenGiessen, Germanyand
- Max Planck Institute for Heart and Lung ResearchBad Nauheim, Germany
| | - Henning Gall
- Justus Liebig University GiessenGiessen, Germanyand
| | | |
Collapse
|
138
|
Rihan M, Nalla LV, Dharavath A, Shard A, Kalia K, Khairnar A. Pyruvate Kinase M2: a Metabolic Bug in Re-Wiring the Tumor Microenvironment. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2019; 12:149-167. [PMID: 31183810 PMCID: PMC6937361 DOI: 10.1007/s12307-019-00226-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/17/2019] [Indexed: 12/16/2022]
Abstract
Metabolic reprogramming is a newly emerged hallmark of cancer attaining a recent consideration as an essential factor for the progression and endurance of cancer cells. A prime event of this altered metabolism is increased glucose uptake and discharge of lactate into the cells surrounding constructing a favorable tumor niche. Several oncogenic factors help in promoting this consequence including, pyruvate kinase M2 (PKM2) a rate-limiting enzyme of glycolysis in tumor metabolism via exhibiting its low pyruvate kinase activity and nuclear moon-lightening functions to increase the synthesis of lactate and macromolecules for tumor proliferation. Not only its role in cancer cells but also its role in the tumor microenvironment cells has to be understood for developing the small molecules against it which is lacking with the literature till date. Therefore, in this present review, the role of PKM2 with respect to various tumor niche cells will be clarified. Further, it highlights the updated list of therapeutics targeting PKM2 pre-clinically and clinically with their added limitations. This upgraded understanding of PKM2 may provide a pace for the reader in developing chemotherapeutic strategies for better clinical survival with limited resistance.
Collapse
Affiliation(s)
- Mohd Rihan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Lakshmi Vineela Nalla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Anil Dharavath
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India.
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gujarat, India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Palaj, Gandhinagar, Gujarat, -382355, India.
| |
Collapse
|
139
|
Su W, Shi J, Zhao Y, Yan F, Lei L, Li H. Porphyromonas gingivalis triggers inflammatory responses in periodontal ligament cells by succinate-succinate dehydrogenase-HIF-1α axis. Biochem Biophys Res Commun 2019; 522:184-190. [PMID: 31757417 DOI: 10.1016/j.bbrc.2019.11.074] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023]
Abstract
Metabolic reprogramming from oxidative phosphorylation to glycolysis have been implicated in the pathogenesis of inflammatory diseases, such as pulmonary hypertension, rheumatoid arthritis and sepsis. Whether metabolic reprogramming participates in the progression of bacteriogenic periodontitis has never been reported. In the present study, we explored metabolic changes in periodontal ligament cells (PDLSCs) in response to Porphyromonas gingivalis. (P. gingivalis)-infected PDLSCs showed distinct metabolomics with metabolic reprogramming from oxidative phosphorylation to glycolysis. In addition, bacteria invasion triggered fundamental changes in glycolysis and tricarboxylate acid (TCA) cycle-related genes, such as the hexokinase (HK), isocitrate dehydrogenase (IDH) and succinate dehydrogenase (SDH). Moreover, P. gingivalis-infected PDLSCs showed accumulation of succinate, elevation in succinate dehydrogenase activity, pileup of reactive oxygen species and activation of hypoxia inducible factor-1α (HIF-1α) pathway. HIF-1α and succinate inhibitors, as well as SDH knockdown alleviated proinflammatory cytokine expression in P. gingivalis-infected PDLSCs. Therefore, targeting metabolic reprogramming by regulating the succinate-SDH-HIF-1α axis may facilitate host modulation therapy of chronic periodontitis.
Collapse
Affiliation(s)
- Wenqi Su
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China; Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jiahong Shi
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yunhe Zhao
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lang Lei
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Houxuan Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
140
|
Role of polypyrimidine tract-binding protein 1/yin yang 2 signaling in regulating vascular smooth muscle cell proliferation and neointima hyperplasia. Toxicol Appl Pharmacol 2019; 383:114747. [DOI: 10.1016/j.taap.2019.114747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/22/2019] [Accepted: 09/05/2019] [Indexed: 11/21/2022]
|
141
|
Cheng X, Wang Y, Du L. Epigenetic Modulation in the Initiation and Progression of Pulmonary Hypertension. Hypertension 2019; 74:733-739. [PMID: 31476913 DOI: 10.1161/hypertensionaha.119.13458] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pulmonary hypertension (PH) is a severe disease with multiple etiologies. In addition to genetics, recent studies have revealed the epigenetic modulation in the initiation and progression of PH. In this review, we summarize the epigenetic mechanisms in the pathogenesis of PH, specifically, DNA methylation, histone modifications, and microRNAs. We further emphasize the diagnostic and therapeutic potential of these epigenetic hallmarks in PH. Finally, we highlight the developmental reprogramming in adult-onset PH because of adverse perinatal exposures such as intrauterine growth restriction and extrauterine growth restriction. Therefore, epigenetic modifications provide promise for the therapy and prevention of PH.
Collapse
Affiliation(s)
- Xinyu Cheng
- From the Department of Pediatrics, (X.C., Y.W.) Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Wang
- From the Department of Pediatrics, (X.C., Y.W.) Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | |
Collapse
|
142
|
Spiekerkoetter E, Goncharova EA, Guignabert C, Stenmark K, Kwapiszewska G, Rabinovitch M, Voelkel N, Bogaard HJ, Graham B, Pullamsetti SS, Kuebler WM. Hot topics in the mechanisms of pulmonary arterial hypertension disease: cancer-like pathobiology, the role of the adventitia, systemic involvement, and right ventricular failure. Pulm Circ 2019; 9:2045894019889775. [PMID: 31798835 PMCID: PMC6868582 DOI: 10.1177/2045894019889775] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/29/2019] [Indexed: 02/06/2023] Open
Abstract
In order to intervene appropriately and develop disease-modifying therapeutics for pulmonary arterial hypertension, it is crucial to understand the mechanisms of disease pathogenesis and progression. We herein discuss four topics of disease mechanisms that are currently highly debated, yet still unsolved, in the field of pulmonary arterial hypertension. Is pulmonary arterial hypertension a cancer-like disease? Does the adventitia play an important role in the initiation of pulmonary vascular remodeling? Is pulmonary arterial hypertension a systemic disease? Does capillary loss drive right ventricular failure? While pulmonary arterial hypertension does not replicate all features of cancer, anti-proliferative cancer therapeutics might still be beneficial in pulmonary arterial hypertension if monitored for safety and tolerability. It was recognized that the adventitia as a cell-rich compartment is important in the disease pathogenesis of pulmonary arterial hypertension and should be a therapeutic target, albeit the data are inconclusive as to whether the adventitia is involved in the initiation of neointima formation. There was agreement that systemic diseases can lead to pulmonary arterial hypertension and that pulmonary arterial hypertension can have systemic effects related to the advanced lung pathology, yet there was less agreement on whether idiopathic pulmonary arterial hypertension is a systemic disease per se. Despite acknowledging the limitations of exactly assessing vascular density in the right ventricle, it was recognized that the failing right ventricle may show inadequate vascular adaptation resulting in inadequate delivery of oxygen and other metabolites. Although the debate was not meant to result in a definite resolution of the specific arguments, it sparked ideas about how we might resolve the discrepancies by improving our disease modeling (rodent models, large-animal studies, studies of human cells, tissues, and organs) as well as standardization of the models. Novel experimental approaches, such as lineage tracing and better three-dimensional imaging of experimental as well as human lung and heart tissues, might unravel how different cells contribute to the disease pathology.
Collapse
Affiliation(s)
- Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Elena A. Goncharova
- Pittsburgh Heart, Blood and Vascular Medicine Institute, Pulmonary, Allergy & Critical Care Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christophe Guignabert
- INSERM UMR_S 999, Université Paris-Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Kurt Stenmark
- Department of Pediatrics, School of Medicine, University of Colorado, Denver, CO, USA
- Cardio Vascular Pulmonary Research Lab, University of Colorado, Denver, CO, USA
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute, Lung Vascular Research, Medical University of Graz, Graz, Austria
| | - Marlene Rabinovitch
- Division of Pediatric Cardiology, Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Norbert Voelkel
- Department of Pulmonary Medicine, Vrije Universiteit MC, Amsterdam, The Netherlands
| | - Harm J. Bogaard
- Department of Pulmonary Medicine, Vrije Universiteit MC, Amsterdam, The Netherlands
| | - Brian Graham
- Pulmonary Sciences and Critical Care, School of Medicine, University of Colorado, Denver, CO, USA
| | - Soni S. Pullamsetti
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité – Universitaetsmedizin Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
143
|
Chen Z, Wang Z, Jin T, Shen G, Wang Y, Tan X, Gan Y, Yang F, Liu Y, Huang C, Zhang Y, Fu X, Shi C. Fibrogenic fibroblast-selective near-infrared phototherapy to control scarring. Theranostics 2019; 9:6797-6808. [PMID: 31660069 PMCID: PMC6815952 DOI: 10.7150/thno.36375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
Rationale: Fibroblasts, the predominant cell type responsible for tissue fibrosis, are heterogeneous, and the targeting of unique fibrogenic population of fibroblasts is highly expected. Very recently, elevated glycolysis is demonstrated to play a pivotal role in the determination of fibrogenic phenotype of fibroblasts. However, it is lack of specific strategies for targeting and elimination of such fibrogenic populations. In this study, a novel strategy to use the a near-infrared (NIR) dye IR-780 for the targeting and elimination of a fibrogenic population of glycolytic fibroblasts to control the cutaneous scarring is developed. Methods: The identification and cell properties test of fibrogenic fibroblasts with IR-780 were conducted by using fluorescence activated cell sorting, transplantation experiments, in vivo imaging, RNA sequencing in human cell experiments and mouse and rat wound models. The uptake of IR-780 in fibroblasts mediated by HIF-1α/SLCO2A1 and the metabolic properties of IR-780H fibroblasts were investigated using RNA interference or signaling inhibitors. The fibrogenic fibroblast-selective near-infrared phototherapy of IR-780 were evaluated in human cell experiments and mouse wound models. Results: IR-780 is demonstrated to recognize a unique glycolytic fibroblast lineage, which is responsible for the bulk of connective tissue deposition during cutaneous wound healing and cancer stroma formation. Further results identified that SLCO2A1 is involved in the preferential uptake of IR-780 in fibrogenic fibroblasts, which is regulated by HIF-1α. Moreover, with intrinsic dual phototherapeutic activities, IR-780 significantly diminishes cutaneous scarring through the targeted ablation of the fibrogenic population by photothermal and photodynamic effects. Conclusion: This work provides a unique strategy for the targeted control of tissue scarring by fibrogenic fibroblast-selective near-infrared phototherapy. It is proposed that IR-780 based theranostic methodology holds promise for translational medicine aimed at regulation of fibrogenic behavior.
Collapse
Affiliation(s)
- Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Taotao Jin
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Gufang Shen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yibo Gan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Fan Yang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yunsheng Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Chunji Huang
- College of Basic Medical Sciences, Army Medical University, 400038 Chongqing, China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital Shanghai Jiao Tong University, School of Medicine, Shanghai, 200011, China
| | - Xiaobing Fu
- Institute of Basic Medical Sciences, Chinese PLA General Hospital, Beijing, 100853, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
144
|
Bonnet S, Boucherat O, Paulin R, Wu D, Hindmarch CCT, Archer SL, Song R, Moore JB, Provencher S, Zhang L, Uchida S. Clinical value of non-coding RNAs in cardiovascular, pulmonary, and muscle diseases. Am J Physiol Cell Physiol 2019; 318:C1-C28. [PMID: 31483703 DOI: 10.1152/ajpcell.00078.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although a majority of the mammalian genome is transcribed to RNA, mounting evidence indicates that only a minor proportion of these transcriptional products are actually translated into proteins. Since the discovery of the first non-coding RNA (ncRNA) in the 1980s, the field has gone on to recognize ncRNAs as important molecular regulators of RNA activity and protein function, knowledge of which has stimulated the expansion of a scientific field that quests to understand the role of ncRNAs in cellular physiology, tissue homeostasis, and human disease. Although our knowledge of these molecules has significantly improved over the years, we have limited understanding of their precise functions, protein interacting partners, and tissue-specific activities. Adding to this complexity, it remains unknown exactly how many ncRNAs there are in existence. The increased use of high-throughput transcriptomics techniques has rapidly expanded the list of ncRNAs, which now includes classical ncRNAs (e.g., ribosomal RNAs and transfer RNAs), microRNAs, and long ncRNAs. In addition, splicing by-products of protein-coding genes and ncRNAs, so-called circular RNAs, are now being investigated. Because there is substantial heterogeneity in the functions of ncRNAs, we have summarized the present state of knowledge regarding the functions of ncRNAs in heart, lungs, and skeletal muscle. This review highlights the pathophysiologic relevance of these ncRNAs in the context of human cardiovascular, pulmonary, and muscle diseases.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Olivier Boucherat
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Roxane Paulin
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Danchen Wu
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Charles C T Hindmarch
- Queen's Cardiopulmonary Unit, Translational Institute of Medicine, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Joseph B Moore
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Steeve Provencher
- Pulmonary Hypertension and Vascular Biology Research Group, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval, Quebec City, Quebec, Canada.,Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Shizuka Uchida
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky.,The Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, Kentucky.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky
| |
Collapse
|
145
|
Brittain EL, Thennapan T, Maron BA, Chan SY, Austin ED, Spiekerkoetter E, Bogaard HJ, Guignabert C, Paulin R, Machado RF, Yu PB. Update in Pulmonary Vascular Disease 2016 and 2017. Am J Respir Crit Care Med 2019. [PMID: 29533671 DOI: 10.1164/rccm.201801-0062up] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Evan L Brittain
- 1 Division of Cardiovascular Medicine, Department of Medicine.,2 Vanderbilt Translational and Clinical Cardiovascular Research Center.,3 Pulmonary Vascular Center, Department of Medicine, and
| | | | - Bradley A Maron
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,6 Department of Cardiology, Boston VA Healthcare System, Boston, Massachusetts
| | - Stephen Y Chan
- 7 Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Eric D Austin
- 3 Pulmonary Vascular Center, Department of Medicine, and.,8 Pediatric Pulmonary Hypertension Program, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Edda Spiekerkoetter
- 9 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and.,10 Vera Moulton Wall Center for Pulmonary Vascular Disease, Cardiovascular Institute, Stanford University, Stanford, California
| | - Harm J Bogaard
- 11 Pulmonary Hypertension Expert Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Christophe Guignabert
- 12 INSERM UMR-S 999, Le Plessis-Robinson, France.,13 Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Roxane Paulin
- 14 Quebec Heart and Lung Institute, Laval University, Quebec, Quebec, Canada; and
| | - Roberto F Machado
- 15 Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - Paul B Yu
- 5 Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
146
|
Pang H, Jia W, Hu Z. Emerging Applications of Metabolomics in Clinical Pharmacology. Clin Pharmacol Ther 2019; 106:544-556. [DOI: 10.1002/cpt.1538] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/18/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Huanhuan Pang
- School of Pharmaceutical Sciences Tsinghua University Beijing China
| | - Wei Jia
- Cancer Biology Program University of Hawaii Cancer Center Honolulu Hawaii USA
| | - Zeping Hu
- School of Pharmaceutical Sciences Tsinghua University Beijing China
- Tsinghua‐Peking Joint Center for Life Sciences Tsinghua University Beijing China
- Beijing Frontier Research Center for Biological Structure Tsinghua University Beijing China
| |
Collapse
|
147
|
Jiang D, Zhang Y, Yang L, Lu W, Mai L, Guo H, Liu X. Retracted
: Long noncoding RNA HCG22 suppresses proliferation and metastasis of bladder cancer cells by regulation of PTBP1. J Cell Physiol 2019; 235:1711-1722. [DOI: 10.1002/jcp.29090] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/27/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Dong Jiang
- Department of Urology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Yongyu Zhang
- Department of Interventional Radiology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Lewei Yang
- Department of Radiotherapy for Abdominal Neoplasms The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Wuzhu Lu
- Department of Ultrasound The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Lei Mai
- Department of Gastroenterology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Huixue Guo
- Department of Gastroenterology The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| | - Xialei Liu
- Department of Hepatobiliary Surgery The Fifth Affiliated Hospital of Sun Yat‐sen University Zhuhai China
| |
Collapse
|
148
|
Kurakula K, Sun XQ, Happé C, da Silva Goncalves Bos D, Szulcek R, Schalij I, Wiesmeijer KC, Lodder K, Tu L, Guignabert C, de Vries CJ, de Man FS, Vonk Noordegraaf A, ten Dijke P, Goumans MJ, Bogaard HJ. Prevention of progression of pulmonary hypertension by the Nur77 agonist 6-mercaptopurine: role of BMP signalling. Eur Respir J 2019; 54:13993003.02400-2018. [DOI: 10.1183/13993003.02400-2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/19/2019] [Indexed: 01/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive fatal disease characterised by abnormal remodelling of pulmonary vessels, leading to increased vascular resistance and right ventricle failure. This abnormal vascular remodelling is associated with endothelial cell dysfunction, increased proliferation of smooth muscle cells, inflammation and impaired bone morphogenetic protein (BMP) signalling. Orphan nuclear receptor Nur77 is a key regulator of proliferation and inflammation in vascular cells, but its role in impaired BMP signalling and vascular remodelling in PAH is unknown.We hypothesised that activation of Nur77 by 6-mercaptopurine (6-MP) would improve PAH by inhibiting endothelial cell dysfunction and vascular remodelling.Nur77 expression is decreased in cultured pulmonary microvascular endothelial cells (MVECs) and lungs of PAH patients. Nur77 significantly increased BMP signalling and strongly decreased proliferation and inflammation in MVECs. In addition, conditioned medium from PAH MVECs overexpressing Nur77 inhibited the growth of healthy smooth muscle cells. Pharmacological activation of Nur77 by 6-MP markedly restored MVEC function by normalising proliferation, inflammation and BMP signalling. Finally, 6-MP prevented and reversed abnormal vascular remodelling and right ventricle hypertrophy in the Sugen/hypoxia rat model of severe angioproliferative PAH.Our data demonstrate that Nur77 is a critical modulator in PAH by inhibiting vascular remodelling and increasing BMP signalling, and activation of Nur77 could be a promising option for the treatment of PAH.
Collapse
|
149
|
Wang F, Yao X, Zhang Y, Tang J. Synthesis, biological function and evaluation of Shikonin in cancer therapy. Fitoterapia 2019; 134:329-339. [DOI: 10.1016/j.fitote.2019.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022]
|
150
|
Archer SL. Pyruvate Kinase and Warburg Metabolism in Pulmonary Arterial Hypertension: Uncoupled Glycolysis and the Cancer-Like Phenotype of Pulmonary Arterial Hypertension. Circulation 2019; 136:2486-2490. [PMID: 29255124 DOI: 10.1161/circulationaha.117.031655] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|