1
|
Crnkovic S, Thekkekara Puthenparampil H, Mulch S, Biasin V, Radic N, Wilhelm J, Bartkuhn M, Bonyadi Rad E, Wawrzen A, Matzer I, Mitra A, Leib RD, Nagy BM, Sahu-Osen A, Valzano F, Bordag N, Evermann M, Hoetzenecker K, Olschewski A, Ljubojevic-Holzer S, Wygrecka M, Stenmark K, Marsh LM, de Jesus Perez V, Kwapiszewska G. Adventitial fibroblasts direct smooth muscle cell-state transition in pulmonary vascular disease. eLife 2025; 13:RP98558. [PMID: 40208251 PMCID: PMC11984959 DOI: 10.7554/elife.98558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Background Pulmonary vascular remodeling is a progressive pathological process characterized by functional alterations within pulmonary artery smooth muscle cells (PASMCs) and adventitial fibroblasts (PAAFs). Mechanisms driving the transition to a diseased phenotype remain elusive. Methods We combined transcriptomic and proteomic profiling with phenotypic characterization of source-matched cells from healthy controls and individuals with idiopathic pulmonary arterial hypertension (IPAH). Bidirectional cellular crosstalk was examined using direct and indirect co-culture models, and phenotypic responses were assessed via transcriptome analysis. Results PASMC and PAAF undergo distinct phenotypic shifts during pulmonary vascular remodeling, with limited shared features, such as reduced mitochondrial content and hyperpolarization. IPAH-PASMC exhibit increased glycosaminoglycan production and downregulation of contractile machinery, while IPAH-PAAF display a hyperproliferative phenotype. We identified alterations in extracellular matrix components, including laminin and collagen, alongside pentraxin-3 and hepatocyte growth factor, as potential regulators of PASMC phenotypic transitions mediated by PAAF. Conclusions While PASMCs and PAAFs retain their core cellular identities, they acquire distinct disease-associated states. These findings provide new insights into the dynamic interplay of pulmonary vascular mesenchymal cells in disease pathogenesis. Funding This work was supported by Cardio-Pulmonary Institute EXC 2026 390649896 (GK) and Austrian Science Fund (FWF) grant I 4651-B (SC).
Collapse
Affiliation(s)
- Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | | | - Shirin Mulch
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Valentina Biasin
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Nemanja Radic
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Jochen Wilhelm
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Marek Bartkuhn
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | | | - Alicja Wawrzen
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Ingrid Matzer
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Ankita Mitra
- Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Ryan D Leib
- Mass Spectrometry Laboratory, Stanford University School of MedicineStanfordUnited States
| | | | - Anita Sahu-Osen
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
| | | | - Natalie Bordag
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | | | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | - Malgorzata Wygrecka
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Kurt Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of ColoradoAuroraUnited States
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| |
Collapse
|
2
|
Dey T, Zhyvylo I, Jiang L, Olapoju SO, Pena A, Avolio T, Lin D, Goncharov D, Greenland JR, Wolters PJ, DeLisser H, Pullamsetti SS, Kudryashova TV, Goncharova EA. Non-canonical HIPPO-MST1/2 promotes hyper-proliferation of pulmonary vascular cells through CDC20. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.646845. [PMID: 40236147 PMCID: PMC11996459 DOI: 10.1101/2025.04.03.646845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
HIPPO components mammalian Ste20-like protein kinases 1 and 2 (MST1/2) are well described growth suppressors. However, in pulmonary arterial hypertension (PAH), MST1/2 switch their roles and become pro-proliferative and pro-survival molecules, supporting hyper-proliferation of pulmonary artery (PA) smooth muscle cells (PASMCs) and adventitial fibroblasts (PAAFs), remodeling of small PAs, and pulmonary hypertension. Here, we report that MST1/2 promotes hyper-proliferation and apoptosis resistance of human PAH PASMCs and PAAFs by up-regulating cell division cycle protein 20 (CDC20), establishing novel link between HIPPO-MST1/2 and cell cycle regulation in PAH. Authors Contributions conception and design of the work (EAG, SSP, TVK); acquisition, analysis, and interpretation of data (TD, IZ, LJ, SOO, AP, TA, DL, DG, JRG, PJW, HD, TK); drafting and editing the manuscript (EAG, SSP, TVK, JRG, PJW).
Collapse
|
3
|
Peng TY, Lu JM, Zheng XL, Zeng C, He YH. The role of lactate metabolism and lactylation in pulmonary arterial hypertension. Respir Res 2025; 26:99. [PMID: 40075458 PMCID: PMC11905457 DOI: 10.1186/s12931-025-03163-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary artery pressure and vascular remodeling. Recent studies have underscored the pivotal role of metabolic dysregulation and epigenetic modifications in the pathogenesis of PAH. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that links cellular metabolism with activity regulation. Recent findings indicate that, in addition to altered glycolytic activity and dysregulated. Lactate homeostasis and lactylation-a novel epigenetic modification-also play a significant role in the development of PAH. This review synthesizes current knowledge regarding the relationship between altered glycolytic activity and PAH, with a particular focus on the cumulative effects of lactate in pulmonary vascular cells. Furthermore, lactylation, an emerging epigenetic modification, is discussed in the context of PAH. By elucidating the complex interplay between lactate metabolism and lactylation in PAH, this review aims to provide insights into potential therapeutic targets. Understanding these metabolic pathways may lead to innovative strategies for managing PAH and improving patient outcomes. Future research should focus on the underlying mechanisms through which lactylation influences the pathophysiology of PAH, thereby aiding in the development of targeted interventions.
Collapse
Affiliation(s)
- Tong-Yu Peng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jun-Mi Lu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xia-Lei Zheng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu-Hu He
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
4
|
Pullamsetti SS, Savai R. Decoding lysosome communication. Science 2025; 387:359-361. [PMID: 39847645 DOI: 10.1126/science.adv1201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Lysosome interaction with other organelles may be linked to pulmonary hypertension.
Collapse
Affiliation(s)
- Soni Savai Pullamsetti
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Department of Internal Medicine, Justus Liebig University, Giessen, Germany
| |
Collapse
|
5
|
Grobs Y, Romanet C, Lemay SE, Bourgeois A, Voisine P, Theberge C, Sauvaget M, Breuils-Bonnet S, Martineau S, El Kabbout R, Valasarajan C, Chelladurai P, Pelletier A, Mougin M, Dumais E, Perron J, Flamand N, Potus F, Provencher S, Pullamsetti SS, Boucherat O, Bonnet S. ATP citrate lyase drives vascular remodeling in systemic and pulmonary vascular diseases through metabolic and epigenetic changes. Sci Transl Med 2024; 16:eado7824. [PMID: 39661707 DOI: 10.1126/scitranslmed.ado7824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/04/2024] [Accepted: 11/19/2024] [Indexed: 12/13/2024]
Abstract
ATP citrate lyase (ACLY), a crucial enzyme in de novo lipid synthesis and histone acetylation, plays a key role in regulating vascular smooth muscle cell (VSMC) proliferation and survival. We found that human coronary and pulmonary artery tissues had up-regulated ACLY expression during vascular remodeling in coronary artery disease and pulmonary arterial hypertension. Pharmacological and genetic inhibition of ACLY in human primary cultured VSMCs isolated from the coronary arteries of patients with coronary artery diseases and from the distal pulmonary arteries of patients with pulmonary arterial hypertension resulted in reduced cellular proliferation and migration and increased susceptibility to apoptosis. These cellular changes were linked to diminished glycolysis, reduced lipid synthesis, impairment in general control nonrepressed protein 5 (GCN5)-dependent histone acetylation and suppression of the transcription factor FOXM1. In vivo studies using a pharmacological inhibitor and VSMC-specific Acly knockout mice showed that ACLY inhibition alleviated vascular remodeling. ACLY inhibition alleviated remodeling in carotid injury and ligation models in rodents and attenuated pulmonary arterial hypertension in Sugen/hypoxia rat and mouse models. Moreover, ACLY inhibition showed improvements in vascular remodeling in human ex vivo models, which included cultured human coronary artery and saphenous vein rings as well as precision-cut lung slices. Our results propose ACLY as a novel therapeutic target for treating complex vascular diseases, offering promising avenues for future clinical intervention.
Collapse
Affiliation(s)
- Yann Grobs
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Charlotte Romanet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Sarah-Eve Lemay
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Alice Bourgeois
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Pierre Voisine
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Charlie Theberge
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Melanie Sauvaget
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Sandra Breuils-Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Sandra Martineau
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Reem El Kabbout
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Chanil Valasarajan
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
| | - Prakash Chelladurai
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
| | - Andreanne Pelletier
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Manon Mougin
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Elizabeth Dumais
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Québec Heart and Lung Institute Research Centre (G1V 4G5), Department of Medicine, Faculty of Medicine, Québec City, QC G1V 0A6, Canada
| | - Jean Perron
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Nicolas Flamand
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Québec Heart and Lung Institute Research Centre (G1V 4G5), Department of Medicine, Faculty of Medicine, Québec City, QC G1V 0A6, Canada
| | - François Potus
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Steeve Provencher
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Soni Savai Pullamsetti
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany
| | - Olivier Boucherat
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Québec Heart and Lung Institute Research Centre, Québec City, QC G1V 4G5, Canada
| |
Collapse
|
6
|
Dou L, You W, Chai Y, Shi H, Liu Q, Jiang Q, Li H. LncRNA H19 Promotes Angiogenesis in Mouse Pulmonary Artery Endothelial Cells by Regulating the HIF-1α/VEGF Signaling Pathway. Biochem Genet 2024:10.1007/s10528-024-10983-3. [PMID: 39633221 DOI: 10.1007/s10528-024-10983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024]
Abstract
Persistent pulmonary hypertension of the newborn (PPHN) is a syndrome of acute respiratory failure characterized by systemic hypoxemia and elevated pulmonary arterial pressure, which leads to pathological changes in pulmonary vascular remodeling and endothelial cell function. Long non-coding RNA (lncRNA) H19 has been shown to be involved in the regulation of arterial endothelial cell function, but its regulatory role in PPHN is not fully understood. In the present study, mouse pulmonary artery endothelial cells (MPAECs) were cultured in a hypoxic conditions. Subsequently, the regulatory function of lncRNA H19 on MPAECs was explored by constructing adenoviruses knocking down and overexpressing lncRNA H19. The results revealed that the hypoxic conditions could induce the proliferation and migration of MPAECs, as well as the high expression of lncRNA H19 in MPAECs. Knockdown of lncRNA H19 expression in MPAECs reversed hypoxic environment-induced functional changes in endothelial cells, whereas overexpression of lncRNA H19 further enhanced the proliferation and migration of MPAECs. In addition, lncRNA H19 upregulated the hypoxia-inducible factor-1α (HIF-1α)/vascular endothelial growth factor (VEGF) pathway through sponge of miNA-20a-5p, which in turn promoted changes in endothelial cell function. LncRNA H19 may interfere with vascular remodeling in hypoxia-induced pulmonary hypertension by upregulating the expression of HIF-1α and VEGF in vascular endothelial cells.
Collapse
Affiliation(s)
- Lei Dou
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China.
| | - Wei You
- Orthopedics Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen, China
| | - Yannan Chai
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Huiju Shi
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Qing Liu
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Qiaoli Jiang
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Huiling Li
- Department of Neonatology, Southern University of Science and Technology Hospital, Shenzhen, China
| |
Collapse
|
7
|
Klibaner-Schiff E, Simonin EM, Akdis CA, Cheong A, Johnson MM, Karagas MR, Kirsh S, Kline O, Mazumdar M, Oken E, Sampath V, Vogler N, Wang X, Nadeau KC. Environmental exposures influence multigenerational epigenetic transmission. Clin Epigenetics 2024; 16:145. [PMID: 39420431 PMCID: PMC11487774 DOI: 10.1186/s13148-024-01762-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Epigenetic modifications control gene expression and are essential for turning genes on and off to regulate and maintain differentiated cell types. Epigenetics are also modified by a multitude of environmental exposures, including diet and pollutants, allowing an individual's environment to influence gene expression and resultant phenotypes and clinical outcomes. These epigenetic modifications due to gene-environment interactions can also be transmitted across generations, raising the possibility that environmental influences that occurred in one generation may be transmitted beyond the second generation, exerting a long-lasting effect. In this review, we cover the known mechanisms of epigenetic modification acquisition, reprogramming and persistence, animal models and human studies used to understand multigenerational epigenetic transmission, and examples of environmentally induced epigenetic change and its transmission across generations. We highlight the importance of environmental health not only on the current population but also on future generations that will experience health outcomes transmitted through epigenetic inheritance.
Collapse
Affiliation(s)
- Eleanor Klibaner-Schiff
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Elisabeth M Simonin
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ana Cheong
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Mary M Johnson
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03756, USA
| | - Sarah Kirsh
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Olivia Kline
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Maitreyi Mazumdar
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Emily Oken
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA, USA
| | - Vanitha Sampath
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Nicholas Vogler
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health, Center On the Early Life Origins of Disease, Johns Hopkins Bloomberg School of Public Health, Baltimore, MA, USA
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kari C Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA.
| |
Collapse
|
8
|
Guignabert C, Aman J, Bonnet S, Dorfmüller P, Olschewski AJ, Pullamsetti S, Rabinovitch M, Schermuly RT, Humbert M, Stenmark KR. Pathology and pathobiology of pulmonary hypertension: current insights and future directions. Eur Respir J 2024; 64:2401095. [PMID: 39209474 PMCID: PMC11533988 DOI: 10.1183/13993003.01095-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
In recent years, major advances have been made in the understanding of the cellular and molecular mechanisms driving pulmonary vascular remodelling in various forms of pulmonary hypertension, including pulmonary arterial hypertension, pulmonary hypertension associated with left heart disease, pulmonary hypertension associated with chronic lung disease and hypoxia, and chronic thromboembolic pulmonary hypertension. However, the survival rates for these different forms of pulmonary hypertension remain unsatisfactory, underscoring the crucial need to more effectively translate innovative scientific knowledge into healthcare interventions. In these proceedings of the 7th World Symposium on Pulmonary Hypertension, we delve into recent developments in the field of pathology and pathophysiology, prioritising them while questioning their relevance to different subsets of pulmonary hypertension. In addition, we explore how the latest omics and other technological advances can help us better and more rapidly understand the myriad basic mechanisms contributing to the initiation and progression of pulmonary vascular remodelling. Finally, we discuss strategies aimed at improving patient care, optimising drug development, and providing essential support to advance research in this field.
Collapse
Affiliation(s)
- Christophe Guignabert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Sébastien Bonnet
- Pulmonary Hypertension research group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Dorfmüller
- Department of Pathology, University Hospital Giessen/Marburg, Giessen, Germany
| | - Andrea J Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research Bad Nauheim, Bad Nauheim, Germany
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
- Universities of Giessen and Marburg Lung Centre, Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Marlene Rabinovitch
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ralph T Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
| | - Marc Humbert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver, CO, USA
| |
Collapse
|
9
|
Guo YZ, Cui HY, Cai MY, Wang D, Deng WP, Hu CP. SOX9 promotes hypoxic pulmonary hypertension through stabilization of DPP4 in pulmonary artery smooth muscle cells. Exp Cell Res 2024; 442:114254. [PMID: 39276964 DOI: 10.1016/j.yexcr.2024.114254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 09/17/2024]
Abstract
Pulmonary hypertension (PH) is a progressive cardiopulmonary disorder characterized by pulmonary vascular remodeling (PVR), primarily due to the excessive proliferation of pulmonary artery smooth muscle cells (PASMCs). This study aimed to investigate the role and molecular mechanism of SOX9 in hypoxic PH in rats. The findings revealed that SOX9 was upregulated in the pulmonary arteries and PASMCs of hypoxia-exposed rats. SOX9 knockdown inhibited hypoxia-induced proliferation and migration of PASMCs, reduced PVR, and subsequently alleviated hypoxia-induced PH in rats, suggesting that SOX9 plays a critical role in PH. Further investigation demonstrated that SOX9 interacted with DPP4, preventing its ubiquitin degradation in hypoxia-exposed PASMCs. DPP4 knockdown inhibited hypoxia-induced PASMC proliferation and migration, and administration of the DPP4 inhibitor sitagliptin (5 mg/kg) significantly reduced PVR and alleviated hypoxia-induced PH in rats, indicating that SOX9 contributes to PH by stabilizing DPP4. The results also showed that hypoxia induced YAP1 expression and dephosphorylation, leading to YAP1 nuclear localization. YAP1 knockdown promoted the degradation of HIF-1α in hypoxia-exposed PASMCs and inhibited hypoxia-induced proliferation and migration of PASMCs. Additionally, HIF-1α, as a transcription factor, promoted SOX9 expression by binding to the SOX9 promoter in hypoxia-exposed PASMCs. In conclusion, hypoxia promotes the proliferation and migration of PASMCs through the regulation of the YAP1/HIF-1α/SOX9/DPP4 signaling pathway, leading to PH in rats. These findings suggest that SOX9 may serve as a potential prognostic marker and therapeutic target for PH.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Cell Hypoxia
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Dipeptidyl Peptidase 4/metabolism
- Dipeptidyl Peptidase 4/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Signal Transduction
- SOX9 Transcription Factor/metabolism
- SOX9 Transcription Factor/genetics
- Vascular Remodeling
- YAP-Signaling Proteins/metabolism
Collapse
Affiliation(s)
- Yan-Zi Guo
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Han-Yu Cui
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Ming-Yuan Cai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Di Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Wei-Ping Deng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, 410078, China.
| |
Collapse
|
10
|
Tuder RM, Gandjeva A, Williams S, Kumar S, Kheyfets VO, Hatton-Jones KM, Starr JR, Yun J, Hong J, West NP, Stenmark KR. Digital Spatial Profiling Identifies Distinct Molecular Signatures of Vascular Lesions in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2024; 210:329-342. [PMID: 38568479 PMCID: PMC11348978 DOI: 10.1164/rccm.202307-1310oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/03/2024] [Indexed: 06/09/2024] Open
Abstract
Rationale: Idiopathic pulmonary arterial hypertension (IPAH) is characterized by extensive pulmonary vascular remodeling caused by plexiform and obliterative lesions, media hypertrophy, inflammatory cell infiltration, and alterations of the adventitia. Objective: We sought to test the hypothesis that microscopic IPAH vascular lesions express unique molecular profiles, which collectively are different from control pulmonary arteries. Methods: We used digital spatial transcriptomics to profile the genomewide differential transcriptomic signature of key pathological lesions (plexiform, obliterative, intima+media hypertrophy, and adventitia) in IPAH lungs (n = 11) and compared these data with the intima+media hypertrophy and adventitia of control pulmonary artery (n = 5). Measurements and Main Results: We detected 8,273 transcripts in the IPAH lesions and control lung pulmonary arteries. Plexiform lesions and IPAH adventitia exhibited the greatest number of differentially expressed genes when compared with intima+media hypertrophy and obliterative lesions. Plexiform lesions in IPAH showed enrichment for 1) genes associated with transforming growth factor β signaling and 2) mutated genes affecting the extracellular matrix and endothelial-mesenchymal transformation. Plexiform lesions and IPAH adventitia showed upregulation of genes involved in immune and IFN signaling, coagulation, and complement pathways. Cellular deconvolution indicated variability in the number of vascular and inflammatory cells between IPAH lesions, which underlies the differential transcript profiling. Conclusions: IPAH lesions express unique molecular transcript profiles enriched for pathways involving pathogenetic pathways, including genetic disease drivers, innate and acquired immunity, hypoxia sensing, and angiogenesis signaling. These data provide a rich molecular-structural framework in IPAH vascular lesions that inform novel biomarkers and therapeutic targets in this highly morbid disease.
Collapse
Affiliation(s)
- Rubin M. Tuder
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine
- Program in Translational Lung Research, Division of Pulmonary and Critical Care Sciences, Department of Medicine
| | - Aneta Gandjeva
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine
- Program in Translational Lung Research, Division of Pulmonary and Critical Care Sciences, Department of Medicine
| | - Sarah Williams
- Queensland Cyber Infrastructure Foundation, St. Lucia, Queensland, Australia
- Griffith Institute for Drug Discovery
| | - Sushil Kumar
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine
| | - Vitaly O. Kheyfets
- Program in Translational Lung Research, Division of Pulmonary and Critical Care Sciences, Department of Medicine
- Division of Pediatric Critical Care Medicine and Cardiovascular Pulmonary Research Laboratory, and
- Department of Biomedical Informatics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado
| | | | - Jacqueline R. Starr
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | - Jeong Yun
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | - Jason Hong
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California
| | - Nicholas P. West
- Menzies Health Institute, and
- School of Pharmacy and Medical Science, Griffith University, Nathan, Queensland, Australia
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine
- Division of Pediatric Critical Care Medicine and Cardiovascular Pulmonary Research Laboratory, and
| |
Collapse
|
11
|
Kong D, Liu J, Lu J, Zeng C, Chen H, Duan Z, Yu K, Zheng X, Zou P, Zhou L, Lv Y, Zeng Q, Lu L, Li J, He Y. HMGB2 Release Promotes Pulmonary Hypertension and Predicts Severity and Mortality of Patients With Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:e172-e195. [PMID: 38572649 DOI: 10.1161/atvbaha.123.319916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.
Collapse
MESH Headings
- Animals
- HMGB2 Protein/genetics
- HMGB2 Protein/metabolism
- Humans
- Vascular Remodeling
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Pulmonary Artery/pathology
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Rats
- Mice, Inbred C57BL
- Mice
- Cell Proliferation
- Severity of Illness Index
- Signal Transduction
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Rats, Sprague-Dawley
- Female
- Cells, Cultured
- Middle Aged
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
Collapse
Affiliation(s)
- Deping Kong
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Jing Liu
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junmi Lu
- Pathology (J. Lu), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cheng Zeng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hao Chen
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhenzhen Duan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Ke Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangdong, China (K.Y.)
| | - Xialei Zheng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pu Zou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liufang Zhou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Cardiovascular Medicine, The Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China (L.Z.)
| | - Yicheng Lv
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Qingye Zeng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital (L.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Jiang Li
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuhu He
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Zhang H, Li M, Hu CJ, Stenmark KR. Fibroblasts in Pulmonary Hypertension: Roles and Molecular Mechanisms. Cells 2024; 13:914. [PMID: 38891046 PMCID: PMC11171669 DOI: 10.3390/cells13110914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Fibroblasts, among the most prevalent and widely distributed cell types in the human body, play a crucial role in defining tissue structure. They do this by depositing and remodeling extracellular matrixes and organizing functional tissue networks, which are essential for tissue homeostasis and various human diseases. Pulmonary hypertension (PH) is a devastating syndrome with high mortality, characterized by remodeling of the pulmonary vasculature and significant cellular and structural changes within the intima, media, and adventitia layers. Most research on PH has focused on alterations in the intima (endothelial cells) and media (smooth muscle cells). However, research over the past decade has provided strong evidence of the critical role played by pulmonary artery adventitial fibroblasts in PH. These fibroblasts exhibit the earliest, most dramatic, and most sustained proliferative, apoptosis-resistant, and inflammatory responses to vascular stress. This review examines the aberrant phenotypes of PH fibroblasts and their role in the pathogenesis of PH, discusses potential molecular signaling pathways underlying these activated phenotypes, and highlights areas of research that merit further study to identify promising targets for the prevention and treatment of PH.
Collapse
Affiliation(s)
- Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Min Li
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cheng-Jun Hu
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
13
|
Chu X, Kheirollahi V, Lingampally A, Chelladurai P, Valasarajan C, Vazquez-Armendariz AI, Hadzic S, Khadim A, Pak O, Rivetti S, Wilhelm J, Bartkuhn M, Crnkovic S, Moiseenko A, Heiner M, Kraut S, Atefi LS, Koepke J, Valente G, Ruppert C, Braun T, Samakovlis C, Alexopoulos I, Looso M, Chao CM, Herold S, Seeger W, Kwapiszewska G, Huang X, Zhang JS, Pullamsetti SS, Weissmann N, Li X, El Agha E, Bellusci S. GLI1+ Cells Contribute to Vascular Remodeling in Pulmonary Hypertension. Circ Res 2024; 134:e133-e149. [PMID: 38639105 DOI: 10.1161/circresaha.123.323736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The precise origin of newly formed ACTA2+ (alpha smooth muscle actin-positive) cells appearing in nonmuscularized vessels in the context of pulmonary hypertension is still debatable although it is believed that they predominantly derive from preexisting vascular smooth muscle cells (VSMCs). METHODS Gli1Cre-ERT2; tdTomatoflox mice were used to lineage trace GLI1+ (glioma-associated oncogene homolog 1-positive) cells in the context of pulmonary hypertension using 2 independent models of vascular remodeling and reverse remodeling: hypoxia and cigarette smoke exposure. Hemodynamic measurements, right ventricular hypertrophy assessment, flow cytometry, and histological analysis of thick lung sections followed by state-of-the-art 3-dimensional reconstruction and quantification using Imaris software were used to investigate the contribution of GLI1+ cells to neomuscularization of the pulmonary vasculature. RESULTS The data show that GLI1+ cells are abundant around distal, nonmuscularized vessels during steady state, and this lineage contributes to around 50% of newly formed ACTA2+ cells around these normally nonmuscularized vessels. During reverse remodeling, cells derived from the GLI1+ lineage are largely cleared in parallel to the reversal of muscularization. Partial ablation of GLI1+ cells greatly prevented vascular remodeling in response to hypoxia and attenuated the increase in right ventricular systolic pressure and right heart hypertrophy. Single-cell RNA sequencing on sorted lineage-labeled GLI1+ cells revealed an Acta2high fraction of cells with pathways in cancer and MAPK (mitogen-activated protein kinase) signaling as potential players in reprogramming these cells during vascular remodeling. Analysis of human lung-derived material suggests that GLI1 signaling is overactivated in both group 1 and group 3 pulmonary hypertension and can promote proliferation and myogenic differentiation. CONCLUSIONS Our data highlight GLI1+ cells as an alternative cellular source of VSMCs in pulmonary hypertension and suggest that these cells and the associated signaling pathways represent an important therapeutic target for further studies.
Collapse
MESH Headings
- Animals
- Zinc Finger Protein GLI1/metabolism
- Zinc Finger Protein GLI1/genetics
- Mice
- Vascular Remodeling
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Inbred C57BL
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Mice, Transgenic
- Male
- Humans
- Hypoxia/metabolism
- Hypoxia/physiopathology
Collapse
Affiliation(s)
- Xuran Chu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) (X.C., S.B.), Wenzhou Medical University, China
- School of Pharmaceutical Sciences (X.C., X.L.), Wenzhou Medical University, China
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Vahid Kheirollahi
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Arun Lingampally
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Prakash Chelladurai
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Chanil Valasarajan
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Ana Ivonne Vazquez-Armendariz
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Stefan Hadzic
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Ali Khadim
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Oleg Pak
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Stefano Rivetti
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Jochen Wilhelm
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Medical University Graz, Austria (S.C., G.K.)
| | - Alena Moiseenko
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Monika Heiner
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Simone Kraut
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | | | - Janine Koepke
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Guilherme Valente
- Max Planck Institute for Lung and Heart, Bad Nauheim, Germany (G.V., T.B., M.L., W.S.)
| | - Clemens Ruppert
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Thomas Braun
- Max Planck Institute for Lung and Heart, Bad Nauheim, Germany (G.V., T.B., M.L., W.S.)
| | - Christos Samakovlis
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Ioannis Alexopoulos
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Mario Looso
- Max Planck Institute for Lung and Heart, Bad Nauheim, Germany (G.V., T.B., M.L., W.S.)
| | - Cho-Ming Chao
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Pediatrics, HELIOS University Medical Center, Witten/Herdecke University, Wuppertal, Germany (C.-M.C.)
| | - Susanne Herold
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Werner Seeger
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
- Max Planck Institute for Lung and Heart, Bad Nauheim, Germany (G.V., T.B., M.L., W.S.)
| | - Grazyna Kwapiszewska
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
- Ludwig Boltzmann Institute for Lung Vascular Research, Medical University Graz, Austria (S.C., G.K.)
| | - Xiaoying Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, China (X.H., J.-S.Z.)
| | - Jin-San Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, China (X.H., J.-S.Z.)
| | - Soni Savai Pullamsetti
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Norbert Weissmann
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
| | - Xiaokun Li
- School of Pharmaceutical Sciences (X.C., X.L.), Wenzhou Medical University, China
| | - Elie El Agha
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control (A.L., A.I.V.-A., A.K., M.H., I.A., S. Herold, E.E.A.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| | - Saverio Bellusci
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) (X.C., S.B.), Wenzhou Medical University, China
- Cardio-Pulmonary Institute (X.C., V.K., A.L., P.C., C.V., A.I.V.-A., S. Hadzic, A.K., O.P., S.R., J.W., M.B., A.M., M.H., S.K., L.S., J.K., C.R., C.S., I.A., C.-M.C., S. Herold, W.S., G.K., S.S.P., N.W., E.E.A., S.B.), Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Justus Liebig University Giessen, Germany
- Institute for Lung Health, Giessen, Germany (P.C., C.V., A.I.V.-A., A.K., J.W., M.B., J.K., C.S., I.A., S. Herold, W.S., G.K., S.S.P., E.E.A., S.B.)
| |
Collapse
|
14
|
Ranasinghe ADCU, Tennakoon TMPB, Schwarz MA. Emerging Epigenetic Targets and Their Molecular Impact on Vascular Remodeling in Pulmonary Hypertension. Cells 2024; 13:244. [PMID: 38334636 PMCID: PMC10854593 DOI: 10.3390/cells13030244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Pulmonary Hypertension (PH) is a terminal disease characterized by severe pulmonary vascular remodeling. Unfortunately, targeted therapy to prevent disease progression is limited. Here, the vascular cell populations that contribute to the molecular and morphological changes of PH in conjunction with current animal models for studying vascular remodeling in PH will be examined. The status quo of epigenetic targeting for treating vascular remodeling in different PH subtypes will be dissected, while parallel epigenetic threads between pulmonary hypertension and pathogenic cancer provide insight into future therapeutic PH opportunities.
Collapse
Affiliation(s)
| | | | - Margaret A. Schwarz
- Department of Pediatrics, Indiana University School of Medicine, 1234 Notre Dame Ave, South Bend, IN 46617, USA
| |
Collapse
|
15
|
Jankowski K, Jagana V, Bisserier M, Hadri L. Switch-Independent 3A: An Epigenetic Regulator in Cancer with New Implications for Pulmonary Arterial Hypertension. Biomedicines 2023; 12:10. [PMID: 38275371 PMCID: PMC10813728 DOI: 10.3390/biomedicines12010010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Epigenetic mechanisms, including DNA methylation, histone modifications, and non-coding RNA, play a crucial role in the regulation of gene expression and are pivotal in biological processes like apoptosis, cell proliferation, and differentiation. SIN3a serves as a scaffold protein and facilitates interactions with transcriptional epigenetic partners and specific DNA-binding transcription factors to modulate gene expression by adding or removing epigenetic marks. However, the activation or repression of gene expression depends on the factors that interact with SIN3a, as it can recruit both transcriptional activators and repressors. The role of SIN3a has been extensively investigated in the context of cancer, including melanoma, lung, and breast cancer. Our group is interested in defining the roles of SIN3a and its partners in pulmonary vascular disease. Pulmonary arterial hypertension (PAH) is a multifactorial disease often described as a cancer-like disease and characterized by disrupted cellular metabolism, sustained vascular cell proliferation, and resistance to apoptosis. Molecularly, PAH shares many common signaling pathways with cancer cells, offering the opportunity to further consider therapeutic strategies used for cancer. As a result, many signaling pathways observed in cancer were studied in PAH and have encouraged new research studying SIN3a's role in PAH due to its impact on cancer growth. This comparison offers new therapeutic options. In this review, we delineate the SIN3a-associated epigenetic mechanisms in cancer and PAH cells and highlight their impact on cell survival and proliferation. Furthermore, we explore in detail the role of SIN3a in cancer to provide new insights into its emerging role in PAH pathogenesis.
Collapse
Affiliation(s)
- Katherine Jankowski
- Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy & Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY 10595, USA; (V.J.); (M.B.)
| | - Malik Bisserier
- Department of Cell Biology and Anatomy & Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY 10595, USA; (V.J.); (M.B.)
| | - Lahouaria Hadri
- Center for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
16
|
Ljubojevic-Holzer S, Crnkovic S. Boosting the Exhausted Vasculature-SIRT3 (to the) Rescue. Am J Respir Cell Mol Biol 2023; 69:497-499. [PMID: 37586074 PMCID: PMC10633846 DOI: 10.1165/rcmb.2023-0199ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Affiliation(s)
- Senka Ljubojevic-Holzer
- Division of Cardiology and Division of Molecular Biology Medical University of Graz Graz, Austria
| | - Slaven Crnkovic
- Division of Physiology Medical University of Graz Graz, Austria
- Institute for Lung Health Giessen, Germany
- Cardiopulmonary Institute Giessen, Germany
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Austria
| |
Collapse
|
17
|
Eichstaedt CA, Bikou O, Sommer N, Schermuly RT, Pullamsetti SS, Weissmann N, Harbaum L, Tabeling C, Wißmüller M, Foris V, Kuebler WM, Hinderhofer K, Olschewski A, Kwapiszewska G. [Genetic diagnostics and molecular approaches in pulmonary arterial hypertension]. Pneumologie 2023; 77:862-870. [PMID: 37963476 DOI: 10.1055/a-2145-4663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
The recently published new European guidelines for diagnosis and treatment of pulmonary hypertension now offer the so far most extensive description of genetic testing and counselling for pulmonary arterial hypertension patients. In addition, the importance of a clinical screening of healthy mutation carriers is highlighted as well as the genetic testing of patients with a suspicion of pulmonary veno-occlusive disease. We frame the respective parts of the guidelines on genetic testing and counselling in the context of recent data and provide comments. Finally, we give an outlook on novel molecular approaches starting from Sotatercept, addressing ion channels and novel therapeutic developments.
Collapse
Affiliation(s)
- Christina A Eichstaedt
- Thoraxklinik Heidelberg gGmbH am Universitätsklinikum Heidelberg und TLRC am Deutschen Zentrum für Lungenforschung (DZL), Heidelberg, Deutschland
- Institut für Humangenetik, Universität Heidelberg, Heidelberg, Deutschland
| | - Olympia Bikou
- Medizinische Klinik und Poliklinik I, LMU Klinikum, LMU München, Deutschland
| | - Natascha Sommer
- Pneumologie und Intensivmedizin, Medizinische Klinik II, Universitätsklinikum Gießen und Marburg und UGMLC am Deutschen Zentrum für Lungenforschung (DZL), Gießen, Deutschland
| | - Ralph T Schermuly
- Zentrum für Innere Medizin, Justus-Liebig-Universität, Gießen, UGMLC Deutsches Zentrum für Lungenforschung (DZL), Gießen, Deutschland
| | - Soni S Pullamsetti
- Medizinische Klinik II, Cardio-Pulmonary Institute (CPI), UGMLC Deutsches Zentrum für Lungenforschung (DZL), Justus-Liebig-Universität, Gießen, Deutschland
- Max-Planck-Institut für Herz- und Lungenforschung und UGMLC am Deutschen Zentrum für Lungenforschung (DZL), Bad Nauheim, Deutschland
| | - Norbert Weissmann
- Medizinische Klinik II, Cardio-Pulmonary Institute (CPI), UGMLC Deutsches Zentrum für Lungenforschung (DZL), Justus-Liebig-Universität, Gießen, Deutschland
| | - Lars Harbaum
- Abteilung für Pneumologie, II. Medizinische Klinik und Poliklinik, zzt. Klinik für Intensivmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
| | - Christoph Tabeling
- Fächerverbund Infektiologie, Pneumologie und Intensivmedizin, Klinik für Pneumologie, Beatmungsmedizin und Intensivmedizin mit dem Arbeitsbereich Schlafmedizin, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Max Wißmüller
- Klinik III für Innere Medizin, Herzzentrum der Universität zu Köln und Cologne Cardiovascular Research Center (CCRC), Universität zu Köln, Köln, Deutschland
| | - Vasile Foris
- Universitätsklinik für Innere Medizin, Klinische Abteilung für Pneumologie, Medizinische Universität Graz, Graz, Österreich
- Ludwig Boltzmann Institut für Lungengefäßforschung, Graz, Österreich
| | - Wolfgang M Kuebler
- Institut für Physiologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - Katrin Hinderhofer
- Institut für Humangenetik, Universität Heidelberg, Heidelberg, Deutschland
| | - Andrea Olschewski
- Ludwig Boltzmann Institut für Lungengefäßforschung, Graz, Österreich
- Experimentelle Anästhesiologie, Universitätsklinik für Anästhesiologie und Intensivmedizin, Medizinische Universität Graz, Graz, Österreich
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institut für Lungengefäßforschung, Graz, Österreich
- Otto Loewi Research Center, Medizinische Universität Graz, Graz, Österreich
- Institute for Lung Health, Giessen, Germany
| |
Collapse
|
18
|
Chen CN, Hajji N, Yeh FC, Rahman S, Ali S, Wharton J, Baxan N, Zhao L, Xie CY, Chen YG, Frid MG, Chelladurai P, Pullamsetti SS, Stenmark KR, Wilkins MR, Zhao L. Restoration of Foxp3 + Regulatory T Cells by HDAC-Dependent Epigenetic Modulation Plays a Pivotal Role in Resolving Pulmonary Arterial Hypertension Pathology. Am J Respir Crit Care Med 2023; 208:879-895. [PMID: 37676930 DOI: 10.1164/rccm.202301-0181oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023] Open
Abstract
Rationale: Immune dysregulation is a common feature of pulmonary arterial hypertension (PAH). Histone deacetylase (HDAC)-dependent transcriptional reprogramming epigenetically modulates immune homeostasis and is a novel disease-oriented approach in modern times. Objectives: To identify a novel functional link between HDAC and regulatory T cells (Tregs) in PAH, aiming to establish disease-modified biomarkers and therapeutic targets. Methods: Peripheral blood mononuclear cells were isolated from patients with idiopathic PAH (IPAH) and rodent models of pulmonary hypertension (PH): monocrotaline rats, Sugen5416-hypoxia rats, and Treg-depleted mice. HDAC inhibitor vorinostat (suberoylanilide hydroxamic acid, SAHA) was used to examine the immune modulatory effects in vivo, ex vivo, and in vitro. Measurements and Main Results: Increased HDAC expression was associated with reduced Foxp3+ Tregs and increased PD-1 (programmed cell death-1) signaling in peripheral blood mononuclear cells from patients with IPAH. SAHA differentially modified a cluster of epigenetic-sensitive genes and induced Foxp3+ Treg conversion in IPAH T cells. Rodent models recapitulated these epigenetic aberrations and T-cell dysfunction. SAHA attenuated PH phenotypes and restored FOXP3 transcription and Tregs in PH rats; interestingly, the effects were more profound in female rats. Selective depletion of CD25+ Tregs in Sugen5416-hypoxia mice neutralized the effects of SAHA. Furthermore, SAHA inhibited endothelial cytokine/chemokine release upon stimulation and subsequent immune chemotaxis. Conclusions: Our results indicated HDAC aberration was associated with Foxp3+ Treg deficiency and demonstrated an epigenetic-mediated mechanism underlying immune dysfunction in PAH. Restoration of Foxp3+ Tregs by HDAC inhibitors is a promising approach to resolve pulmonary vascular pathology, highlighting the potential benefit of developing epigenetic therapies for PAH.
Collapse
Affiliation(s)
- Chien-Nien Chen
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Nabil Hajji
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Fu-Chiang Yeh
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sunniyat Rahman
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
- Department of Haematology, University College London Cancer Institute, University College London, London, United Kingdom
| | - Souad Ali
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Nicoleta Baxan
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Lin Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Chong-Yang Xie
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Yi-Guan Chen
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Maria G Frid
- Division of Critical Care Medicine and Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado, Denver, Colorado
| | - Prakash Chelladurai
- Max-Planck Institute for Heart and Lung Research, Member of German Center for Lung Research, Giessen, Germany; and
| | - Soni Savai Pullamsetti
- Max-Planck Institute for Heart and Lung Research, Member of German Center for Lung Research, Giessen, Germany; and
- Institute of Molecular Biology and Tumor Research, Marburg, Germany
| | - Kurt R Stenmark
- Division of Critical Care Medicine and Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado, Denver, Colorado
| | - Martin R Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Lan Zhao
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
19
|
Li L, Xue Q, Zhang M, Yang Z, Wang D, Yan G, Qiao Y, Tang C, Zhang R. Upregulation of the key biomarker kinesin family member 20A (KIF20A) is associated with pulmonary artery hypertension. Genomics 2023; 115:110705. [PMID: 37703933 DOI: 10.1016/j.ygeno.2023.110705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/21/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
OBJECTIVE Pulmonary artery hypertension (PAH) is a complex, fatal disease with limited treatments. This study aimed to investigate possible key targets in PAH through bioinformatics. METHODS GSE144274 were obtained from Gene Expression Omnibus (GEO) database. Then, differentially expressed genes (DEGs) between idiopathic pulmonary hypertension (IPAH) and healthy subjects were identified and analyzed. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) were analyzed, and a protein-protein interaction (PPI) network was constructed using STRING. The hub genes were identified by MCODE method. The expression levels of hub genes were validated in vitro and in vivo models. Finally, the ROC analysis was performed based on the level of hub genes in clinical plasma samples. RESULTS A total of 363 DEGs were identified. GO analysis on these DEGs were mainly enriched in cell division, inflammatory response, among others. In the KEGG pathways analysis, DEGs mainly involved in cytokine-cytokine receptor interaction, rheumatoid arthritis, and IL-17 signaling pathways were enriched. The DEGs were analyzed with the STRING for PPI network analysis, and 62 hub genes were identified by MCODE. Finally, 6 central genes, KIF18B, SPC25, DLGAP5, KIF20A, CEP55 and ANLN, were screened out due to their novelty role in PAH. The expression of KIF20A was validated to be significantly upregulated both in the lung tissue of hypoxia-induced pulmonary hypertension (HPH) mice and proliferative PASMCs. Additionally, KIF20A levels is evelated in PAH plasma and the area under the curve (AUC) to identify PAH was 0.8591 for KIF20A. CONCLUSION The level of KIF20A elevates during the progression of PAH, which suggestes it could be a potential diagnostic and therapeutic target for the PAH.
Collapse
Affiliation(s)
- Linqing Li
- Zhongda Hospital, Southeast University, Department of Cardiology, China; Linyi People's Hospital, 210009 Nanjing, China
| | - Qi Xue
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Minhao Zhang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Zhanneng Yang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Dong Wang
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Gaoliang Yan
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Yong Qiao
- Zhongda Hospital, Southeast University, Department of Cardiology, China
| | - Chengchun Tang
- Zhongda Hospital, Southeast University, Department of Cardiology, China.
| | - Rui Zhang
- Zhongda Hospital, Southeast University, Department of Cardiology, China.
| |
Collapse
|
20
|
Johnson S, Sommer N, Cox-Flaherty K, Weissmann N, Ventetuolo CE, Maron BA. Pulmonary Hypertension: A Contemporary Review. Am J Respir Crit Care Med 2023; 208:528-548. [PMID: 37450768 PMCID: PMC10492255 DOI: 10.1164/rccm.202302-0327so] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
Major advances in pulmonary arterial hypertension, pulmonary hypertension (PH) associated with lung disease, and chronic thromboembolic PH cast new light on the pathogenetic mechanisms, epidemiology, diagnostic approach, and therapeutic armamentarium for pulmonary vascular disease. Here, we summarize key basic, translational, and clinical PH reports, emphasizing findings that build on current state-of-the-art research. This review includes cutting-edge progress in translational pulmonary vascular biology, with a guide to the diagnosis of patients in clinical practice, incorporating recent PH definition revisions that continue emphasis on early detection of disease. PH management is reviewed including an overview of the evolving considerations for the approach to treatment of PH in patients with cardiopulmonary comorbidities, as well as a discussion of the groundbreaking sotatercept data for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Shelsey Johnson
- The Pulmonary Center, Division of Pulmonary, Allergy, Sleep and Critical Care, Boston University School of Medicine, Boston, Massachusetts
- Department of Pulmonary and Critical Care Medicine and
| | - Natascha Sommer
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | | | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | - Corey E. Ventetuolo
- Department of Medicine and
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| | - Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts
- Department of Cardiology and Department of Pulmonary, Allergy, Sleep, and Critical Care Medicine, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; and
- The University of Maryland-Institute for Health Computing, Bethesda, Maryland
| |
Collapse
|
21
|
Dave J, Jagana V, Janostiak R, Bisserier M. Unraveling the epigenetic landscape of pulmonary arterial hypertension: implications for personalized medicine development. J Transl Med 2023; 21:477. [PMID: 37461108 DOI: 10.1186/s12967-023-04339-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease associated with the remodeling of pulmonary blood vessels. If left unaddressed, PAH can lead to right heart failure and even death. Multiple biological processes, such as smooth muscle proliferation, endothelial dysfunction, inflammation, and resistance to apoptosis, are associated with PAH. Increasing evidence suggests that epigenetic factors play an important role in PAH by regulating the chromatin structure and altering the expression of critical genes. For example, aberrant DNA methylation and histone modifications such as histone acetylation and methylation have been observed in patients with PAH and are linked to vascular remodeling and pulmonary vascular dysfunction. In this review article, we provide a comprehensive overview of the role of key epigenetic targets in PAH pathogenesis, including DNA methyltransferase (DNMT), ten-eleven translocation enzymes (TET), switch-independent 3A (SIN3A), enhancer of zeste homolog 2 (EZH2), histone deacetylase (HDAC), and bromodomain-containing protein 4 (BRD4). Finally, we discuss the potential of multi-omics integration to better understand the molecular signature and profile of PAH patients and how this approach can help identify personalized treatment approaches.
Collapse
Affiliation(s)
- Jaydev Dave
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Radoslav Janostiak
- First Faculty of Medicine, BIOCEV, Charles University, Vestec, 25250, Czech Republic
| | - Malik Bisserier
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
| |
Collapse
|
22
|
Benincasa G, Napoli C, Loscalzo J, Maron BA. Pursuing functional biomarkers in complex disease: Focus on pulmonary arterial hypertension. Am Heart J 2023; 258:96-113. [PMID: 36565787 DOI: 10.1016/j.ahj.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 05/11/2023]
Abstract
A major gap in diagnosis, classification, risk stratification, and prediction of therapeutic response exists in pulmonary arterial hypertension (PAH), driven in part by a lack of functional biomarkers that are also disease-specific. In this regard, leveraging big data-omics analyses using innovative approaches that integrate network medicine and machine learning correlated with clinically useful indices or risk stratification scores is an approach well-positioned to advance PAH precision medicine. For example, machine learning applied to a panel of 48 cytokines, chemokines, and growth factors could prognosticate PAH patients with immune-dominant subphenotypes at elevated or low-risk for mortality. Here, we discuss strengths and weaknesses of the most current studies evaluating omics-derived biomarkers in PAH. Progress in this field is offset by studies with small sample size, pervasive limitations in bioinformatics, and lack of standardized methods for data processing and interpretation. Future success in this field, in turn, is likely to hinge on mechanistic validation of data outputs in order to couple functional biomarker data with target-specific therapeutics in clinical practice.
Collapse
Affiliation(s)
- Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA.
| |
Collapse
|
23
|
Karolak JA, Welch CL, Mosimann C, Bzdęga K, West JD, Montani D, Eyries M, Mullen MP, Abman SH, Prapa M, Gräf S, Morrell NW, Hemnes AR, Perros F, Hamid R, Logan MPO, Whitsett J, Galambos C, Stankiewicz P, Chung WK, Austin ED. Molecular Function and Contribution of TBX4 in Development and Disease. Am J Respir Crit Care Med 2023; 207:855-864. [PMID: 36367783 PMCID: PMC10111992 DOI: 10.1164/rccm.202206-1039tr] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past decade, recognition of the profound impact of the TBX4 (T-box 4) gene, which encodes a member of the evolutionarily conserved family of T-box-containing transcription factors, on respiratory diseases has emerged. The developmental importance of TBX4 is emphasized by the association of TBX4 variants with congenital disorders involving respiratory and skeletal structures; however, the exact role of TBX4 in human development remains incompletely understood. Here, we discuss the developmental, tissue-specific, and pathological TBX4 functions identified through human and animal studies and review the published TBX4 variants resulting in variable disease phenotypes. We also outline future research directions to fill the gaps in our understanding of TBX4 function and of how TBX4 disruption affects development.
Collapse
Affiliation(s)
- Justyna A. Karolak
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Katarzyna Bzdęga
- Chair and Department of Genetics and Pharmaceutical Microbiology, Poznan University of Medical Sciences, Poznan, Poland
| | - James D. West
- Division of Allergy, Pulmonary and Critical Care Medicine, and
| | - David Montani
- Université Paris-Saclay, Assistance Publique–Hôpitaux de Paris, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Mélanie Eyries
- Sorbonne Université, AP-HP, Département de Génétique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Mary P. Mullen
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | | | - Matina Prapa
- St. George’s University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Stefan Gräf
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, United Kingdom
| | - Nicholas W. Morrell
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Heart and Lung Research Institute, Cambridge, United Kingdom
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, and
| | - Frédéric Perros
- Université Paris-Saclay, Assistance Publique–Hôpitaux de Paris, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital de Bicêtre, DMU 5 Thorinno, Inserm UMR_S999, Le Kremlin-Bicêtre, France
| | - Rizwan Hamid
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Malcolm P. O. Logan
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - Jeffrey Whitsett
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Perinatal Institute, Cincinnati, Ohio
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Csaba Galambos
- Department of Pathology, University of Colorado School of Medicine, and Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Wendy K. Chung
- Department of Pediatrics and
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Eric D. Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
24
|
Wang Y, Su L, Wang W, Zhao J, Wang Y, Li S, Liu Y, Chai R, Li X, Teng Z, Liu C, Hu B, Ji F, Jiao J. Endothelial Arid1a deletion disrupts the balance among angiogenesis, neurogenesis and gliogenesis in the developing brain. Cell Prolif 2023; 56:e13447. [PMID: 36916004 DOI: 10.1111/cpr.13447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/22/2023] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
The vascular system and the neural system processes occur simultaneously, the interaction among them is fundamental to the normal development of the central nervous system. Arid1a (AT-rich interaction domain 1A), which encodes an epigenetic subunit of the SWI/SNF chromatin-remodelling complex, is associated with promoter-mediated gene regulation and histone modification. However, the molecular mechanism of the interaction between cerebrovascular and neural progenitor cells (NPCs) remains unclear. To generate Arid1acKO-Tie2 mice, Arid1afl/fl mice were hybridized with Tie2-Cre mice. The Angiogenesis, neurogenesis and gliogenesis were studied by immunofluorescence staining and Western blotting. RNA-seq, RT-PCR, Western blotting, CO-IP and rescue experiments were performed to dissect the molecular mechanisms of Arid1a regulates fate determination of NPCs. We found that the absence of Arid1a results in increased the density of blood vessels, delayed neurogenesis and decreased gliogenesis, even after birth. Mechanistically, the deletion of Arid1a in endothelial cells causes a significant increase in H3k27ac and the secretion of maternal protein 2 (MATN2). In addition, matn2 alters the AKT/SMAD4 signalling pathway through its interaction with the NPCs receptor EGFR, leading to the decrease of SMAD4. SMAD complex further mediates the expression of downstream targets, thereby promoting neurogenesis and inhibiting gliogenesis. This study suggests that endothelial Arid1a tightly controls fate determination of NPCs by regulating the AKT-SMAD signalling pathway.
Collapse
Affiliation(s)
- Yuanyuan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Libo Su
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Wenwen Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jinyue Zhao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yanyan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sihan Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Renjie Chai
- Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Xin Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Zhaoqian Teng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Changmei Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Fen Ji
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Innovation Academy for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
25
|
Yang L, Tian J, Wang J, Zeng J, Wang T, Lin B, Linneman J, Li L, Niu Y, Gou D, Zhang Y. The protective role of EP300 in monocrotaline-induced pulmonary hypertension. Front Cardiovasc Med 2023; 10:1037217. [PMID: 36910531 PMCID: PMC9992637 DOI: 10.3389/fcvm.2023.1037217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Background Pulmonary hypertension (PH) is a lethal disease characterized by pulmonary vascular remodeling, which is mediated by the abnormal proliferation/migration of pulmonary arterial smooth muscle cells (PASMCs). Recent reports suggest the involvement of histone acetylation in PAH development and that histone deacetylase (HDAC) inhibitors have therapeutic potential for the treatment of PAH. EP300 is an acetyltransferase that plays diverse roles in cell proliferation, differentiation, and apoptosis. However, the functions of EP3000 in PH are rarely studied. Results In this work, we found that the expression of EP300 was increased in the pulmonary arteries of monocrotaline (MCT)-induced PH rats. Knockdown of EP300 by AAV-mediated shRNA exacerbated the PH, with a higher right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), and wall thickness in the pulmonary artery of MCT-induced PH rat. On the cellular level, the proliferation of PASMCs was promoted by EP300 knockdown. In addition, the expression of EP300 was increased in PASMCs by the overexpression of EGR1, while the deletion of EGR1 binding sites in the EP300 promoter region decreased the activity of EP300 promoter. Moreover, deleting the EP300 promoter region containing EGR1 binding sites using CRISPR/Cas9 abolished the upregulation of EP300 in MCT-induced rats and exacerbated MCT-induced PH. To summarize, our data indicate that EP300 upregulation mediated by EGR1 has a protective effect on MCT-induced PH. Conclusion These findings showed EP300 expression was increased in the MCT-induced PH model in rats, which could be mediated by EGR1; the EP300 also displayed the potential to provide protection from PH.
Collapse
Affiliation(s)
- Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jinglin Tian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jie Zeng
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ting Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Boya Lin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - John Linneman
- School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|