1
|
Mutgan AC, Radic N, Valzano F, Crnkovic S, El-Merhie N, Evermann M, Hoetzenecker K, Foris V, Brcic L, Marsh LM, Tran-Lundmark K, Jandl K, Kwapiszewska G. A comprehensive map of proteoglycan expression and deposition in the pulmonary arterial wall in health and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L173-L188. [PMID: 38771138 DOI: 10.1152/ajplung.00022.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Changes in the extracellular matrix of pulmonary arteries (PAs) are a key aspect of vascular remodeling in pulmonary hypertension (PH). Yet, our understanding of the alterations affecting the proteoglycan (PG) family remains limited. We sought to investigate the expression and spatial distribution of major vascular PGs in PAs from healthy individuals and various PH groups (chronic obstructive pulmonary disease: PH-COPD, pulmonary fibrosis: PH-PF, idiopathic: IPAH). PG regulation, deposition, and synthesis were notably heightened in IPAH, followed by PH-PF, with minor alterations in PH-COPD. Single-cell analysis unveiled cell-type and disease-specific PG regulation. Agrin expression, a basement membrane PG, was increased in IPAH, with PA endothelial cells (PAECs) identified as a major source. PA smooth muscle cells (PASMCs) mainly produced large-PGs, aggrecan and versican, and small-leucine-like proteoglycan (SLRP) biglycan, whereas the major PGs produced by adventitial fibroblasts were SLRP decorin and lumican. In IPAH and PF-PH, the neointima-forming PASMC population increased the expression of all investigated large-PGs and SLRPs, except fibroblast-predominant decorin (DCN). Expression of lumican, versican, and biglycan also positively correlated with collagen 1α1/1α2 expression in PASMCs in patients with IPAH and PH-PF. We demonstrated that transforming growth factor-beta (TGF-β) regulates versican and biglycan expression, indicating their contribution to vessel fibrosis in IPAH and PF-PH. We furthermore show that certain circulating PG levels display a disease-dependent pattern, with increased decorin and lumican across all patient groups, while versican was elevated in PH-COPD and IPAH and biglycan reduced in IPAH. These findings suggest unique compartment-specific PG regulation in different forms of PH, indicating distinct pathological processes.NEW & NOTEWORTHY Idiopathic pulmonary arterial hypertension (IPAH) pulmonary arteries (PAs) displayed the greatest proteoglycan (PG) changes, with PH associated with pulmonary fibrosis (PH-PF) and PH associated with chronic obstructive pulmonary disease (PH-COPD) following. Agrin, an endothelial cell-specific PG, was solely upregulated in IPAH. Among all cells, neo-intima-forming smooth muscle cells (SMCs) displayed the most significant PG increase. Increased levels of circulating decorin, lumican, and versican, mainly derived from SMCs, and adventitial fibroblasts, may serve as systemic indicators of pulmonary remodeling, reflecting perivascular fibrosis and neointima formation.
Collapse
MESH Headings
- Humans
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Proteoglycans/metabolism
- Male
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Female
- Middle Aged
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Vascular Remodeling
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/pathology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Aged
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Biglycan/metabolism
- Decorin/metabolism
- Adult
- Fibroblasts/metabolism
- Fibroblasts/pathology
- Pulmonary Fibrosis/metabolism
- Pulmonary Fibrosis/pathology
- Lumican/metabolism
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Nemanja Radic
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Francesco Valzano
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Slaven Crnkovic
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Lung Center (DZL), Giessen, Germany
| | - Natalia El-Merhie
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Lung Center (DZL), Giessen, Germany
| | - Matthias Evermann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Vasile Foris
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Leigh M Marsh
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Karin Tran-Lundmark
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- The Pediatric Heart Center, Skåne University Hospital, Lund, Sweden
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
2
|
Liu Y, Lin Y, Huang X, Li Y, Liu Y, Shi L. Association of serum transforming growth factor β 1 with left ventricular hypertrophy in children with primary hypertension. Eur J Pediatr 2023; 182:5439-5446. [PMID: 37755472 PMCID: PMC10746758 DOI: 10.1007/s00431-023-05219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/31/2023] [Accepted: 09/16/2023] [Indexed: 09/28/2023]
Abstract
The current study was designed to assess the association of serum transforming growth factor β1 (TGF-β1) with left ventricular hypertrophy (LVH) in children with primary hypertension. The present single-center prospective trial examined 182 patients diagnosed with primary hypertension in Children's Hospital, Capital Institute of Pediatrics, between January 2021 and September 2022. Clinical data were analyzed, and ambulatory blood pressure was assessed for 24 h. LVH, the commonest subclinical cardiac feature of hypertension, was assessed by echocardiography. According to left ventricular geometry, cases were assigned to the LVH (n = 44) and normal geometry (n = 138) groups. Serum TGF-β1 amounts were quantitated by enzyme-linked immunosorbent assay (ELISA). Receiver operating characteristic (ROC) curves were established to analyze various variables for their predictive values in LVH. Among 182 children with primary hypertension, the concentrations of serum TGF-β1 were higher in stage 2 hypertension than in stage 1 (47.3 (38.8, 52.5) vs. 46.0 (38.6, 48.2) ng/L, Z = - 2.376; P = 0.018). Additionally, serum TGF-β1 content showed a positive correlation with BP levels (P < 0.05). TGF-β1 amounts were significantly elevated in the LVH group compared with the normal geometry group (51.7 (46.1, 54.9) vs. 46.1 (38.7, 48.1) ng/L, Z = - 4.324; P = 0.0000). Serum TGF-β1 content was positively associated with LVH (r = 0.321, P = 0.0000). Multivariable logistic regression analysis showed BMI (OR = 1.188, 95% CI 1.082-1.305; P = 0.0000) and elevated serum TGF-β1 content (OR = 1.063, 95% CI 1.016-1.113; P = 0.009) independently predicted LVH. A multivariable logistic regression model considering BMI and TGF-β1 content in LVH prediction was 0.771, with sensitivity and specificity of 72.7% and 70.3%, respectively. CONCLUSION These data revealed an association of serum TGF-β1 with BP in children with primary hypertension. Serum TGF-β1 concentration was positively correlated with hypertensive cardiac damage. Serum TGF-β1 might constitute a valuable molecular marker for the prediction of LVH in children with primary hypertension. The combination of BMI and TGF-β1 has a certain diagnostic and predictive value for LVH in children with primary hypertension, which may provide a new reference index for early clinical identification of hypertensive cardiac damage. WHAT IS KNOWN • Experimental and clinical data indicated TGF-β1 is involved in BP elevation. • TGF-β1 is positively correlated with LVMI and hypertrophy in adults. WHAT IS NEW • Our current study reveals an association of serum TGF-β1 with BP in children with primary hypertension. • Elevated serum TGF-β1 level is positively associated with LVH in children with primary hypertension. • The combination of BMI and TGF-β1 has a certain diagnostic and predictive value for LVH in children with primary hypertension.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cardiology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Yao Lin
- Department of Cardiology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Xiaolan Huang
- Central Laboratory, Capital Institute of Pediatrics, Beijing, China
| | - Yaqi Li
- Department of Cardiology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Yanyan Liu
- Department of Cardiology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Lin Shi
- Department of Cardiology, Children's Hospital, Capital Institute of Pediatrics, Beijing, China.
| |
Collapse
|
3
|
Villanueva B, Cerdà P, Torres-Iglesias R, Rocamora JL, Figueras A, Viñals F, Riera-Mestre A. Potential angiogenic biomarkers in hereditary hemorrhagic telangiectasia and other vascular diseases. Eur J Intern Med 2023; 115:10-17. [PMID: 37225595 DOI: 10.1016/j.ejim.2023.05.020] [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: 03/12/2023] [Revised: 05/01/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Biomarkers are new tools framed in precision and personalized medicine. Hereditary hemorrhagic telangiectasia (HHT) is a rare genetic vascular disease with disturbances in the angiogenic pathways. Descriptive evidence supports that some angiogenesis-related molecules are differently detected in HHT patients compared to healthy subjects. These molecules are also related to diagnosis, prognosis, complications and therapy monitoring in other common vascular diseases. Despite the need for improving knowledge before applying them in daily clinical practice, there are good candidates to be considered as potential biomarkers in HHT and other vascular diseases. In the present review, the authors aim to summarize and discuss current evidence regarding the main putative angiogenic biomarkers by describing the biological role of each biomarker, the evidence related to HHT and their potential use in this and other common vascular diseases from a clinical point-of-view.
Collapse
Affiliation(s)
- B Villanueva
- HHT Unit. Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - P Cerdà
- HHT Unit. Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - R Torres-Iglesias
- HHT Unit. Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - J L Rocamora
- HHT Unit. Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - A Figueras
- Program Against Cancer Therapeutic Resistance, Institut Catala d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain; Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - F Viñals
- Program Against Cancer Therapeutic Resistance, Institut Catala d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain; Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - A Riera-Mestre
- HHT Unit. Internal Medicine Department, Hospital Universitari Bellvitge, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
4
|
Jandl K, Radic N, Zeder K, Kovacs G, Kwapiszewska G. Pulmonary vascular fibrosis in pulmonary hypertension - The role of the extracellular matrix as a therapeutic target. Pharmacol Ther 2023; 247:108438. [PMID: 37210005 DOI: 10.1016/j.pharmthera.2023.108438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Pulmonary hypertension (PH) is a condition characterized by changes in the extracellular matrix (ECM) deposition and vascular remodeling of distal pulmonary arteries. These changes result in increased vessel wall thickness and lumen occlusion, leading to a loss of elasticity and vessel stiffening. Clinically, the mechanobiology of the pulmonary vasculature is becoming increasingly recognized for its prognostic and diagnostic value in PH. Specifically, the increased vascular fibrosis and stiffening resulting from ECM accumulation and crosslinking may be a promising target for the development of anti- or reverse-remodeling therapies. Indeed, there is a huge potential in therapeutic interference with mechano-associated pathways in vascular fibrosis and stiffening. The most direct approach is aiming to restore extracellular matrix homeostasis, by interference with its production, deposition, modification and turnover. Besides structural cells, immune cells contribute to the level of ECM maturation and degradation by direct cell-cell contact or the release of mediators and proteases, thereby opening a huge avenue to target vascular fibrosis via immunomodulation approaches. Indirectly, intracellular pathways associated with altered mechanobiology, ECM production, and fibrosis, offer a third option for therapeutic intervention. In PH, a vicious cycle of persistent activation of mechanosensing pathways such as YAP/TAZ initiates and perpetuates vascular stiffening, and is linked to key pathways disturbed in PH, such as TGF-beta/BMPR2/STAT. Together, this complexity of the regulation of vascular fibrosis and stiffening in PH allows the exploration of numerous potential therapeutic interventions. This review discusses connections and turning points of several of these interventions in detail.
Collapse
Affiliation(s)
- Katharina Jandl
- Division of Pharmacology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria.
| | - Nemanja Radic
- Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria
| | - Katarina Zeder
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
5
|
Yi X, Tang X, Li T, Chen L, He H, Wu X, Xiang C, Cao M, Wang Z, Wang Y, Wang Y, Huang X. Therapeutic potential of the sphingosine kinase 1 inhibitor, PF-543. Biomed Pharmacother 2023; 163:114401. [PMID: 37167721 DOI: 10.1016/j.biopha.2023.114401] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 05/13/2023] Open
Abstract
PF-543 is a sphingosine kinase 1(SPHK1)inhibitor developed by Pfizer and is currently considered the most potent selective SPHK1 inhibitor. SPHK1 catalyses the production of sphingosine 1-phosphate (S1P) from sphingosine. It is the rate-limiting enzyme of S1P production, and there is substantial evidence to support a very important role for sphingosine kinase in health and disease. This review is the first to summarize the role and mechanisms of PF-543 as an SPHK1 inhibitor in anticancer, antifibrotic, and anti-inflammatory processes, providing new therapeutic leads and ideas for future research and clinical trials.
Collapse
Affiliation(s)
- Xueliang Yi
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China
| | - Xuemei Tang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tianlong Li
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Chen
- University of Electronic Science and Technology of China, China
| | - Hongli He
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China
| | - Xiaoxiao Wu
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunlin Xiang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Cao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zixiang Wang
- University of Electronic Science and Technology of China, China
| | - Yi Wang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| | - Yiping Wang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| | - Xiaobo Huang
- Department of ICU, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; University of Electronic Science and Technology of China, China.
| |
Collapse
|
6
|
Gallardo-Vara E, Ntokou A, Dave JM, Jovin DG, Saddouk FZ, Greif DM. Vascular pathobiology of pulmonary hypertension. J Heart Lung Transplant 2023; 42:544-552. [PMID: 36604291 PMCID: PMC10121751 DOI: 10.1016/j.healun.2022.12.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/31/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Pulmonary hypertension (PH), increased blood pressure in the pulmonary arteries, is a morbid and lethal disease. PH is classified into several groups based on etiology, but pathological remodeling of the pulmonary vasculature is a common feature. Endothelial cell dysfunction and excess smooth muscle cell proliferation and migration are central to the vascular pathogenesis. In addition, other cell types, including fibroblasts, pericytes, inflammatory cells and platelets contribute as well. Herein, we briefly note most of the main cell types active in PH and for each cell type, highlight select signaling pathway(s) highly implicated in that cell type in this disease. Among others, the role of hypoxia-inducible factors, growth factors (e.g., vascular endothelial growth factor, platelet-derived growth factor, transforming growth factor-β and bone morphogenetic protein), vasoactive molecules, NOTCH3, Kruppel-like factor 4 and forkhead box proteins are discussed. Additionally, deregulated processes of endothelial-to-mesenchymal transition, extracellular matrix remodeling and intercellular crosstalk are noted. This brief review touches upon select critical facets of PH pathobiology and aims to incite further investigation that will result in discoveries with much-needed clinical impact for this devastating disease.
Collapse
Affiliation(s)
- Eunate Gallardo-Vara
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut
| | - Aglaia Ntokou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut
| | - Jui M Dave
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut
| | - Daniel G Jovin
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut
| | - Fatima Z Saddouk
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut
| | - Daniel M Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, New Haven, Connecticut; Department of Genetics, Yale University, New Haven, Connecticut.
| |
Collapse
|
7
|
Bouchet C, Cardouat G, Douard M, Coste F, Robillard P, Delcambre F, Ducret T, Quignard JF, Vacher P, Baudrimont I, Marthan R, Berger P, Guibert C, Freund-Michel V. Inflammation and Oxidative Stress Induce NGF Secretion by Pulmonary Arterial Cells through a TGF-β1-Dependent Mechanism. Cells 2022; 11:cells11182795. [PMID: 36139373 PMCID: PMC9496672 DOI: 10.3390/cells11182795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Expression of the nerve growth factor NGF is increased in pulmonary hypertension (PH). We have here studied whether oxidative stress and inflammation, two pathological conditions associated with transforming growth factor-β1 (TGF-β1) in PH, may trigger NGF secretion by pulmonary arterial (PA) cells. Effects of hydrogen peroxide (H2O2) and interleukin-1β (IL-1β) were investigated ex vivo on rat pulmonary arteries, as well as in vitro on human PA smooth muscle (hPASMC) or endothelial cells (hPAEC). TβRI expression was assessed by Western blotting. NGF PA secretion was assessed by ELISA after TGF-β1 blockade (anti-TGF-β1 siRNA, TGF-β1 blocking antibodies, TβRI kinase, p38 or Smad3 inhibitors). TβRI PA expression was evidenced by Western blotting both ex vivo and in vitro. H2O2 or IL-1β significantly increased NGF secretion by hPASMC and hPAEC, and this effect was significantly reduced when blocking TGF-β1 expression, binding to TβRI, TβRI activity, or signaling pathways. In conclusion, oxidative stress and inflammation may trigger TGF-β1 secretion by hPASMC and hPAEC. TGF-β1 may then act as an autocrine factor on these cells, increasing NGF secretion via TβRI activation. Since NGF and TGF-β1 are relevant growth factors involved in PA remodeling, such mechanisms may therefore be relevant to PH pathophysiology.
Collapse
Affiliation(s)
- Clément Bouchet
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Guillaume Cardouat
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Matthieu Douard
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
- IHU Institut de Rythmologie et Modélisation Cardiaque (LIRYC), 33600 Pessac, France
| | - Florence Coste
- Laboratoire de Pharm-Écologie Cardiovasculaire (LaPEC-EA 4278), Université d’Avignon et des Pays du Vaucluse, 84000 Avignon, France
| | - Paul Robillard
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | | | - Thomas Ducret
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Jean-François Quignard
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Pierre Vacher
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Isabelle Baudrimont
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Roger Marthan
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
- CHU de Bordeaux, 33000 Bordeaux, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
- CHU de Bordeaux, 33000 Bordeaux, France
| | - Christelle Guibert
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
| | - Véronique Freund-Michel
- Centre de Recherche Cardio-Thoracique de Bordeaux, University Bordeaux, U1045, 33600 Pessac, France
- INSERM (Institut National de la Santé Et de la Recherche Médicale), Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, 33600 Pessac, France
- Correspondence:
| |
Collapse
|
8
|
Andre P, Joshi SR, Briscoe SD, Alexander MJ, Li G, Kumar R. Therapeutic Approaches for Treating Pulmonary Arterial Hypertension by Correcting Imbalanced TGF-β Superfamily Signaling. Front Med (Lausanne) 2022; 8:814222. [PMID: 35141256 PMCID: PMC8818880 DOI: 10.3389/fmed.2021.814222] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterized by high blood pressure in the pulmonary circulation driven by pathological remodeling of distal pulmonary arteries, leading typically to death by right ventricular failure. Available treatments improve physical activity and slow disease progression, but they act primarily as vasodilators and have limited effects on the biological cause of the disease—the uncontrolled proliferation of vascular endothelial and smooth muscle cells. Imbalanced signaling by the transforming growth factor-β (TGF-β) superfamily contributes extensively to dysregulated vascular cell proliferation in PAH, with overactive pro-proliferative SMAD2/3 signaling occurring alongside deficient anti-proliferative SMAD1/5/8 signaling. We review the TGF-β superfamily mechanisms underlying PAH pathogenesis, superfamily interactions with inflammation and mechanobiological forces, and therapeutic strategies under development that aim to restore SMAD signaling balance in the diseased pulmonary arterial vessels. These strategies could potentially reverse pulmonary arterial remodeling in PAH by targeting causative mechanisms and therefore hold significant promise for the PAH patient population.
Collapse
|
9
|
Yan Y, Jiang R, Yuan P, Wen L, Pang X, Jing Z, He Y, Han Z. Implication of proliferation gene biomarkers in pulmonary hypertension. Animal Model Exp Med 2021; 4:369-380. [PMID: 34977488 PMCID: PMC8690983 DOI: 10.1002/ame2.12191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022] Open
Abstract
Objective/Background Proliferation is a widely recognized trigger for pulmonary hypertension (PH), a life-threatening, progressive disorder of pulmonary blood vessels. This study was aimed to identify some proliferation associated genes/targets for better comprehension of PH pathogenesis. Methods Human pulmonary arterial smooth muscle cells (hPASMCs) were cultured in the presence or absence of human recombinant platelet derived growth factor (rhPDGF)-BB. Cells were collected for metabolomics or transcriptomics study. Gene profiling of lungs of PH rats after hypoxia exposure or of PH patients were retrieved from GEO database. Results 90 metabolites (VIP score >1, fold change >2 or <0.5 and p < .05) and 2701 unique metabolism associated genes (MAGs) were identified in rhPDGF-BB treated hPASMCs compared to control cells. In addition, 1151 differentially expressed genes (313 upregulated and 838 downregulated) were identified in rhPDGF-BB treated hPASMCs compared to control cells (fold change >2 or <0.5 and p < .05). 152 differentially expressed MAGs were then determined, out of which 9 hub genes (IL6, CXCL8, CCL2, CXCR4, CCND1, PLAUR, PLAU, HBEGF and F3) were defined as core proliferation associated hub genes in protein proten interaction analysis. In addition, the hub gene-based LASSO model can predict the occurrence of PH (AUC = 0.88). The expression of CXCR4, as one of the hub genes, was positively correlated to immune cell infiltrates. Conclusion Our findings revealed some key proliferation associated genes in PH, which provide the crucial information concerning complex metabolic reprogramming and inflammatory modulation in response to proliferation signals and might offer therapeutic gains for PH.
Collapse
Affiliation(s)
- Yi Yan
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐University MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research)partner site Munich Heart AllianceMunichGermany
| | - Rong Jiang
- Department of Cardio‐Pulmonary CirculationShanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Ping Yuan
- Department of Cardio‐Pulmonary CirculationShanghai Pulmonary Hospital, Tongji University School of MedicineShanghaiChina
| | - Li Wen
- Department of CardiologyThe First Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | | | - Zhi‐Cheng Jing
- State Key Laboratory of Complex, Severe, and Rare Diseases, and Department of CardiologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | | | - Zhi‐Yan Han
- State Key Laboratory of Cardiovascular Disease and FuWai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
10
|
Huang N, Zhu TT, Liu T, Ge XY, Wang D, Liu H, Zhu GX, Zhang Z, Hu CP. Aspirin ameliorates pulmonary vascular remodeling in pulmonary hypertension by dampening endothelial-to-mesenchymal transition. Eur J Pharmacol 2021; 908:174307. [PMID: 34245748 DOI: 10.1016/j.ejphar.2021.174307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Pulmonary vascular remodeling (PVR) is the pathological basis of pulmonary hypertension (PH). Incomplete understanding of PVR etiology has hindered drug development for this devastating disease, which exhibits poor prognosis despite the currently available therapies. Endothelial-to-mesenchymal transition (EndMT), a process of cell transdifferentiation, has been recently implicated in cardiovascular diseases, including PH. But the questions of how EndMT occurs and how to pharmacologically target EndMT in vivo have yet to be further answered. Herein, by performing hematoxylin-eosin and immunofluorescence staining, transmission electron microscopy and Western blotting, we found that EndMT plays a key role in the pathogenesis of PH, and importantly that aspirin, a FDA-approved widely used drug, was capable of ameliorating PVR in a preclinical rat model of hypoxia-induced PH. Moreover, aspirin exerted its inhibitory effects on EndMT in vitro and in vivo by suppressing HIF-1α/TGF-β1/Smads/Snail signaling pathway. Our data suggest that EndMT represents an intriguing drug target for the prevention and treatment of hypoxic PH and that aspirin may be repurposed to meet the urgent therapeutic needs of hypoxic PH patients.
Collapse
Affiliation(s)
- Ning Huang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Tian-Tian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, 453000, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, Henan, 453000, China
| | - Ting Liu
- Department of Pharmacy, Hangzhou First Peoples Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Xiao-Yue Ge
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Di Wang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Hong Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Guang-Xuan Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China.
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan, 410078, China.
| |
Collapse
|
11
|
Ji Y, Lisabeth EM, Neubig RR. Transforming Growth Factor β1 Increases Expression of Contractile Genes in Human Pulmonary Arterial Smooth Muscle Cells by Potentiating Sphingosine-1-Phosphate Signaling. Mol Pharmacol 2021; 100:53-60. [PMID: 34031187 DOI: 10.1124/molpharm.120.000019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/30/2021] [Indexed: 11/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by elevated pulmonary arterial pressure and carries a very poor prognosis. Understanding of PAH pathogenesis is needed to support the development of new therapeutic strategies. Transforming growth factor β (TGF-β) drives vascular remodeling and increases vascular resistance by regulating differentiation and proliferation of smooth muscle cells (SMCs). Also, sphingosine-1-phosphate (S1P) has been implicated in PAH, but the relation between these two signaling mechanisms is not well understood. Here, we characterize the signaling networks downstream of TGF-β in human pulmonary arterial smooth muscle cells (HPASMCs), which involves mothers against decapentaplegic homolog (SMAD) signaling as well as Rho GTPases. Activation of Rho GTPases regulates myocardin-related transcription factor (MRTF) and serum response factor (SRF) transcription activity and results in upregulation of contractile gene expression. Our genetic and pharmacologic data show that in HPASMCs upregulation of α smooth muscle actin (αSMA) and calponin by TGF-β is dependent on both SMAD and Rho/MRTF-A/SRF transcriptional mechanisms.The kinetics of TGF-β-induced myosin light chain (MLC) 2 phosphorylation, a measure of RhoA activation, are slow, as is regulation of the Rho/MRTF/SRF-induced αSMA expression. These results suggest that TGF-β1 activates Rho/phosphorylated MLC2 through an indirect mechanism, which was confirmed by sensitivity to cycloheximide treatment. As a potential mechanism for this indirect action, TGF-β1 upregulates mRNA for sphingosine kinase (SphK1), the enzyme that produces S1P, an upstream Rho activator, as well as mRNA levels of the S1P receptor (S1PR) 3. SphK1 inhibitor and S1PR3 inhibitors (PF543 and TY52156/VPC23019) reduce TGF-β1-induced αSMA upregulation. Overall, we propose a model in which TGF-β1 activates Rho/MRTF-A/SRF by potentiating an autocrine/paracrine S1P signaling mechanism through SphK1 and S1PR3. SIGNIFICANCE STATEMENT: In human pulmonary arterial smooth muscle cells, transforming growth factor β depends on sphingosine-1-phosphate signaling to bridge the interaction between mothers against decapentaplegic homolog and Rho/myocardin-related transcription factor (MRTF) signaling in regulating α smooth muscle actin (αSMA) expression. The Rho/MRTF pathway is a signaling node in the αSMA regulatory network and is a potential therapeutic target for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Yajing Ji
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| | - Erika M Lisabeth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| |
Collapse
|
12
|
Yung LM, Yang P, Joshi S, Augur ZM, Kim SSJ, Bocobo GA, Dinter T, Troncone L, Chen PS, McNeil ME, Southwood M, Poli de Frias S, Knopf J, Rosas IO, Sako D, Pearsall RS, Quisel JD, Li G, Kumar R, Yu PB. ACTRIIA-Fc rebalances activin/GDF versus BMP signaling in pulmonary hypertension. Sci Transl Med 2021; 12:12/543/eaaz5660. [PMID: 32404506 DOI: 10.1126/scitranslmed.aaz5660] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 01/22/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022]
Abstract
Human genetics, biomarker, and animal studies implicate loss of function in bone morphogenetic protein (BMP) signaling and maladaptive transforming growth factor-β (TGFβ) signaling as drivers of pulmonary arterial hypertension (PAH). Although sharing common receptors and effectors with BMP/TGFβ, the function of activin and growth and differentiation factor (GDF) ligands in PAH are less well defined. Increased expression of GDF8, GDF11, and activin A was detected in lung lesions from humans with PAH and experimental rodent models of pulmonary hypertension (PH). ACTRIIA-Fc, a potent GDF8/11 and activin ligand trap, was used to test the roles of these ligands in animal and cellular models of PH. By blocking GDF8/11- and activin-mediated SMAD2/3 activation in vascular cells, ACTRIIA-Fc attenuated proliferation of pulmonary arterial smooth muscle cells and pulmonary microvascular endothelial cells. In several experimental models of PH, prophylactic administration of ACTRIIA-Fc markedly improved hemodynamics, right ventricular (RV) hypertrophy, RV function, and arteriolar remodeling. When administered after the establishment of hemodynamically severe PH in a vasculoproliferative model, ACTRIIA-Fc was more effective than vasodilator in attenuating PH and arteriolar remodeling. Potent antiremodeling effects of ACTRIIA-Fc were associated with inhibition of SMAD2/3 activation and downstream transcriptional activity, inhibition of proliferation, and enhancement of apoptosis in the vascular wall. ACTRIIA-Fc reveals an unexpectedly prominent role of GDF8, GDF11, and activin as drivers of pulmonary vascular disease and represents a therapeutic strategy for restoring the balance between SMAD1/5/9 and SMAD2/3 signaling in PAH.
Collapse
Affiliation(s)
- Lai-Ming Yung
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peiran Yang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Zachary M Augur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie S J Kim
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Geoffrey A Bocobo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Teresa Dinter
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Luca Troncone
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Po-Sheng Chen
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan City 704, Taiwan
| | - Megan E McNeil
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Southwood
- Department of Pathology, Royal Papworth Hospital, Cambridge CB2 0AY, UK
| | - Sergio Poli de Frias
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - John Knopf
- Acceleron Pharma Inc., Cambridge, MA 02139, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dianne Sako
- Acceleron Pharma Inc., Cambridge, MA 02139, USA
| | | | | | - Gang Li
- Acceleron Pharma Inc., Cambridge, MA 02139, USA
| | | | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Guignabert C, Humbert M. Targeting transforming growth factor-β receptors in pulmonary hypertension. Eur Respir J 2021; 57:2002341. [PMID: 32817256 DOI: 10.1183/13993003.02341-2020] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
Abstract
The transforming growth factor-β (TGF-β) superfamily includes several groups of multifunctional proteins that form two major branches, namely the TGF-β-activin-nodal branch and the bone morphogenetic protein (BMP)-growth differentiation factor (GDF) branch. The response to the activation of these two branches, acting through canonical (small mothers against decapentaplegic (Smad) 2/3 and Smad 1/5/8, respectively) and noncanonical signalling pathways, are diverse and vary for different environmental conditions and cell types. An extensive body of data gathered in recent years has demonstrated a central role for the cross-talk between these two branches in a number of cellular processes, which include the regulation of cell proliferation and differentiation, as well as the transduction of signalling cascades for the development and maintenance of different tissues and organs. Importantly, alterations in these pathways, which include heterozygous germline mutations and/or alterations in the expression of several constitutive members, have been identified in patients with familial/heritable pulmonary arterial hypertension (PAH) or idiopathic PAH (IPAH). Consequently, loss or dysfunction in the delicate, finely-tuned balance between the TGF-β-activin-nodal branch and the BMP-GDF branch are currently viewed as the major molecular defect playing a critical role in PAH predisposition and disease progression. Here we review the role of the TGF-β-activin-nodal branch in PAH and illustrate how this knowledge has not only provided insight into understanding its pathogenesis, but has also paved the way for possible novel therapeutic approaches.
Collapse
Affiliation(s)
- Christophe Guignabert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Marc Humbert
- Faculty of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 (Pulmonary Hypertension: Pathophysiology and Novel Therapies), Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Dept of Respiratory and Intensive Care Medicine, French Pulmonary Hypertension Reference Center, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin-Bicêtre, France
| |
Collapse
|
14
|
Xu J, Yang Y, Yang Y, Xiong C. Identification of Potential Risk Genes and the Immune Landscape of Idiopathic Pulmonary Arterial Hypertension via Microarray Gene Expression Dataset Reanalysis. Genes (Basel) 2021; 12:125. [PMID: 33478117 PMCID: PMC7835985 DOI: 10.3390/genes12010125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
Gene dysfunction and immune cell infiltration play an essential role in the pathogenesis of idiopathic pulmonary arterial hypertension (IPAH). We aimed to investigate the immune landscape and novel differentially expressed genes (DEGs) of IPAH. In addition, potential druggable molecular targets for IPAH were also explored. In this study, the GSE117261 dataset was reanalyzed to explore the immune landscape and hub DEGs of IPAH. Lasso Cox regression analysis and receiver operating characteristic curve analysis were performed to detect the predictive value of IPAH. Additionally, the underlying drug targets for IPAH treatment were determined by drug-gene analysis. IPAH was significantly associated with the transforming growth factor-β (TGF-β) signaling pathway and Wnt signaling pathway as well as energetic metabolism dysfunction. We identified 31 upregulated and 39 downregulated DEGs in IPAH patients. Six hub genes, namely, SAA1, CCL5, CXCR1, CXCR2, CCR1, and ADORA3, were related to IPAH pathogenesis regardless of sex differences. Prediction model analysis showed that the area under the curve values of the hub DEGs except CXCR2 were all above 0.9 for distinguishing IPAH patients. In addition, the relative proportions of 5 subtypes of immune cells, namely, CD8+ T cells, CD4+ memory resting T cells, γ delta T cells, M1 macrophages, and resting mast cells, were significantly upregulated in the IPAH samples, while 6 subtypes of immune cells, namely, CD4+ naive T cells, resting NK cells, monocytes, M0 macrophages, activated mast cells, and neutrophils, were downregulated. Additionally, a total of 17 intersecting drugs targeting 5 genes, CCL5, CXCR1, CXCR2, CCR1, and ADORA3, were generated as potential druggable molecular targets for IPAH. Our study revealed the underlying correlations between genes and immune cells in IPAH and demonstrated for the first time that SAA1, CCL5, CXCR1, CCR1, and ADORA3 may be novel genetic targets for IPAH.
Collapse
Affiliation(s)
- Jing Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China;
| | - Yicheng Yang
- Pulmonary Vascular Disease Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China;
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China;
| | - Changming Xiong
- Pulmonary Vascular Disease Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China;
| |
Collapse
|
15
|
Dunmore BJ, Jones RJ, Toshner MR, Upton PD, Morrell NW. Approaches to treat pulmonary arterial hypertension by targeting bmpr2 - from cell membrane to nucleus. Cardiovasc Res 2021; 117:2309-2325. [PMID: 33399862 DOI: 10.1093/cvr/cvaa350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/06/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is estimated to affect between 10-50 people per million worldwide. The lack of cure and devastating nature of the disease means that treatment is crucial to arrest rapid clinical worsening. Current therapies are limited by their focus on inhibiting residual vasoconstriction rather than targeting key regulators of the cellular pathology. Potential disease-modifying therapies may come from research directed towards causal pathways involved in the cellular and molecular mechanisms of disease. It is widely acknowledged, that targeting reduced expression of the critical bone morphogenetic protein type-2 receptor (BMPR2) and its associated signalling pathways is a compelling therapeutic avenue to explore. In this review we highlight the advances that have been made in understanding this pathway and the therapeutics that are being tested in clinical trials and the clinic to treat PAH.
Collapse
Affiliation(s)
- Benjamin J Dunmore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Rowena J Jones
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Mark R Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Paul D Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| |
Collapse
|
16
|
Csósza G, Karlócai K, Losonczy G, Müller V, Lázár Z. Growth factors in pulmonary arterial hypertension: Focus on preserving right ventricular function. Physiol Int 2020; 107:177-194. [PMID: 32692713 DOI: 10.1556/2060.2020.00021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disease, characterized by increased vascular resistance leading to right ventricle (RV) failure. The extent of right ventricular dysfunction crucially influences disease prognosis; however, currently no therapies have specific cardioprotective effects. Besides discussing the pathophysiology of right ventricular adaptation in PAH, this review focuses on the roles of growth factors (GFs) in disease pathomechanism. We also summarize the involvement of GFs in the preservation of cardiomyocyte function, to evaluate their potential as cardioprotective biomarkers and novel therapeutic targets in PAH.
Collapse
Affiliation(s)
- G Csósza
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - K Karlócai
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - G Losonczy
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - V Müller
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Z Lázár
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
17
|
Bochenek ML, Leidinger C, Rosinus NS, Gogiraju R, Guth S, Hobohm L, Jurk K, Mayer E, Münzel T, Lankeit M, Bosmann M, Konstantinides S, Schäfer K. Activated Endothelial TGFβ1 Signaling Promotes Venous Thrombus Nonresolution in Mice Via Endothelin-1: Potential Role for Chronic Thromboembolic Pulmonary Hypertension. Circ Res 2019; 126:162-181. [PMID: 31747868 DOI: 10.1161/circresaha.119.315259] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Chronic thromboembolic pulmonary hypertension (CTEPH) is characterized by defective thrombus resolution, pulmonary artery obstruction, and vasculopathy. TGFβ (transforming growth factor-β) signaling mutations have been implicated in pulmonary arterial hypertension, whereas the role of TGFβ in the pathophysiology of CTEPH is unknown. OBJECTIVE To determine whether defective TGFβ signaling in endothelial cells contributes to thrombus nonresolution and fibrosis. METHODS AND RESULTS Venous thrombosis was induced by inferior vena cava ligation in mice with genetic deletion of TGFβ1 in platelets (Plt.TGFβ-KO) or TGFβ type II receptors in endothelial cells (End.TGFβRII-KO). Pulmonary endarterectomy specimens from CTEPH patients were analyzed using immunohistochemistry. Primary human and mouse endothelial cells were studied using confocal microscopy, quantitative polymerase chain reaction, and Western blot. Absence of TGFβ1 in platelets did not alter platelet number or function but was associated with faster venous thrombus resolution, whereas endothelial TGFβRII deletion resulted in larger, more fibrotic and higher vascularized venous thrombi. Increased circulating active TGFβ1 levels, endothelial TGFβRI/ALK1 (activin receptor-like kinase), and TGFβRI/ALK5 expression were detected in End.TGFβRII-KO mice, and activated TGFβ signaling was present in vessel-rich areas of CTEPH specimens. CTEPH-endothelial cells and murine endothelial cells lacking TGFβRII simultaneously expressed endothelial and mesenchymal markers and transcription factors regulating endothelial-to-mesenchymal transition, similar to TGFβ1-stimulated endothelial cells. Mechanistically, increased endothelin-1 levels were detected in TGFβRII-KO endothelial cells, murine venous thrombi, or endarterectomy specimens and plasma of CTEPH patients, and endothelin-1 overexpression was prevented by inhibition of ALK5, and to a lesser extent of ALK1. ALK5 inhibition and endothelin receptor antagonization inhibited mesenchymal lineage conversion in TGFβ1-exposed human and murine endothelial cells and improved venous thrombus resolution and pulmonary vaso-occlusions in End.TGFβRII-KO mice. CONCLUSIONS Endothelial TGFβ1 signaling via type I receptors and endothelin-1 contribute to mesenchymal lineage transition and thrombofibrosis, which were prevented by blocking endothelin receptors. Our findings may have relevant implications for the prevention and management of CTEPH.
Collapse
Affiliation(s)
- Magdalena L Bochenek
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Christiane Leidinger
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany
| | - Nico S Rosinus
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Rajinikanth Gogiraju
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Stefan Guth
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.)
| | - Lukas Hobohm
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Kerstin Jurk
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany
| | - Eckhard Mayer
- Thoracic Surgery, Kerckhoff Clinic, Bad Nauheim, Germany (S.G., E.M.).,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Thomas Münzel
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| | - Mareike Lankeit
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Internal Medicine and Cardiology, Campus Virchow Klinikum, Charité -University Medicine, Berlin, Germany (M.L.)
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Medicine, Boston University School of Medicine, MA (M.B.)
| | - Stavros Konstantinides
- Center for Thrombosis and Hemostasis (M.L.B., L.H., K.J., M.L., M.B., S.K.), University Medical Center Mainz, Germany.,Department of Cardiology, Democritus University of Thrace, Alexandroupolis, Greece (S.K.)
| | - Katrin Schäfer
- From the Center for Cardiology, Cardiology I (M.L.B., C.L., N.S.R., R.G., L.H., T.M., K.S.), University Medical Center Mainz, Germany.,German Center for Cardiovascular Research (DZHK e.V.; RheinMain) (M.L.B., N.S.R., R.G., E.M., T.M., K.S.)
| |
Collapse
|
18
|
Li Q, Zhou X, Zhou X. Downregulation of miR‑98 contributes to hypoxic pulmonary hypertension by targeting ALK1. Mol Med Rep 2019; 20:2167-2176. [PMID: 31322216 PMCID: PMC6691262 DOI: 10.3892/mmr.2019.10482] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
Chronic hypoxia is one of the most common causes of secondary pulmonary hypertension, the mechanisms of which remain unclear. MicroRNAs (miRNAs) are small, noncoding RNAs that inhibit the translation or accelerate the degradation of mRNA. Previous studies have demonstrated that deregulated miRNA expression contributes to various cellular processes including cell apoptosis and proliferation, which are mediated by hypoxia. In the present study, the expression of miR‑98 was identified to be decreased in the lung tissue of a hypoxic pulmonary hypertension (HPH) rat model and pulmonary artery (PA) smooth muscle cells (PASMCs), which was induced by hypoxia. By transfecting miR‑98 mimics into PASMCs, the high expression of miR‑98 inhibited cell proliferation, but upregulated hypoxia‑induced PASMCs apoptosis. However, these effects of miR‑98 mimics on PASMCs were reversed by ALK1 (activin receptor‑like kinase‑1) overexpression. ALK1 was identified as a candidate target of miR‑98. In addition, overexpressing miR‑98 markedly decreased the pulmonary artery wall thickness and the right ventricular systolic pressure in rats induced by hypoxia. These results provided clear evidence that miR‑98 was a direct regulator of ALK1, and that the downregulation of miR‑98 contributed to the pathogenesis of HPH. These results provide a novel potential therapeutic strategy for the treatment of HPH.
Collapse
Affiliation(s)
- Qingling Li
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xincan Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xianghui Zhou
- Department of Respiratory Medicine, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
19
|
Zhou Y, Shao L, Ruan W, Jin J, Xu H, Ying K, Wu X. Pulmonary vascular involvement of IgG4-related disease: Case series with a PRISMA-compliant systemic review. Medicine (Baltimore) 2019; 98:e14437. [PMID: 30732204 PMCID: PMC6380675 DOI: 10.1097/md.0000000000014437] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 12/29/2018] [Accepted: 01/16/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Immunoglobulin G4-related disease (IgG4-RD) is a recently recognized, immune-mediated chronic fibrotic inflammation that can involve almost all organs, causing tumefaction and dysfunction. Its presence in pulmonary circulation is underestimated and has not yet been investigated. OBJECTIVES We describe a representative IgG4-RD patient with pulmonary artery stenosis and pulmonary embolism, leading to reversible pulmonary hypertension. Literature review of IgG4-RD with pulmonary circulation involvement was conducted. DATA SOURCES References for this review were identified through searches via PubMed, EBSCO, and Web of Science for published articles before November 2016. RESULTS There were 15 published cases of IgG4-RD with pulmonary vascular involvement, 3 with pulmonary arteritis, 2 with pulmonary artery aneurysm, 3 with pulmonary artery stenosis, 1 with obliterative phlebitis, and 1 with pulmonary embolism. Possible immunity and inflammation mechanisms were summarized. CONCLUSIONS IgG4-RD with pulmonary vascular involvement is rare. Echocardiogram and contrast-enhanced chest CT are helpful to screen the disease. Clinical manifestations were found from asymptomatic to dyspnea or even syncope. And nearly all cases had more than 1 organ affected, with significantly increased serum IgG4 levels. PET/CT aided in identifying affected organs and determining candidate biopsy sites. More awareness is urged to evaluate the pulmonary vascular manifestations of this disease.
Collapse
Affiliation(s)
- Yong Zhou
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, CA
| | - Lingyan Shao
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenjing Ruan
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Joy Jin
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, CA
| | - Hangdi Xu
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kejing Ying
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohong Wu
- Respiratory and Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
20
|
Cong LH, Du SY, Wu YN, Liu Y, Li T, Wang H, Li G, Duan J. Upregulation of Klotho potentially inhibits pulmonary vascular remodeling by blocking the activation of the Wnt signaling pathway in rats with PM2.5-induced pulmonary arterial hypertension. J Cell Biochem 2018; 119:5581-5597. [PMID: 29380911 DOI: 10.1002/jcb.26729] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
We evaluated the effects of Klotho on pulmonary vascular remodeling and cell proliferation and apoptosis in rat models with PM2.5-induced pulmonary arterial hypertension (PAH) via the Wnt signaling pathway. After establishing rat models of PM2.5-induced PAH, these Sprague-Dawley male rats were randomized into control and model groups. Cells extracted from the model rats were sub-categorized into different groups. Activation of Wnt/β-catenin signaling transcription factor was detected by a TOPFlash/FOPFlash assay. A serial of experiment was conducted to identify the mechanism of Klotho on PHA via the Wnt signaling pathway. VEGF levels and PaCO2 content were higher in the model group, while PaO2, NO2- /NO3- content and Klotho level was lower compared to the control group. In comparison to the control group, the model group had decreased Klotho and Bax levels, and elevated Wnt-1, β-catenin, bcl-2, survivin, and PCNA expression, VEGF, IL-6, TNF-α, TNF-β1, and bFGF levels, as well as the percentage of pulmonary artery ring contraction. The Klotho vector, DKK-1 and DKK-1 + Klotho vector groups exhibited reduced cell proliferation, luciferase activity, and the expression of Wnt-1, β-catenin, bcl-2, survivin, and PCNA, as well as shortened S phase compared with the blank and NC groups. Compared with the Klotho vector and DKK-1 groups, the DKK-1 + Klotho vector groups had reduced cell proliferation, luciferase activity, and the expression of Wnt-1, β-catenin, bcl-2, survivin, and PCNA, as well as a shortened S phase. Conclusively, Klotho inhibits pulmonary vascular remodeling by inactivation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Lu-Hong Cong
- Department of Emergency, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Shi-Yu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Yi-Na Wu
- Surgical Intensive Care Unit, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Ying Liu
- Department of Geriatric, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Tao Li
- Surgical Intensive Care Unit, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Hui Wang
- Surgical Intensive Care Unit, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Gang Li
- Surgical Intensive Care Unit, China-Japan Friendship Hospital, Beijing, P. R. China
| | - Jun Duan
- Surgical Intensive Care Unit, China-Japan Friendship Hospital, Beijing, P. R. China
| |
Collapse
|
21
|
Usefulness of Preoperative Transforming Growth Factor-Beta to Predict New Onset Atrial Fibrillation After Surgical Ventricular Septal Myectomy in Patients With Obstructive Hypertrophic Cardiomyopathy. Am J Cardiol 2017; 120:118-123. [PMID: 28483207 DOI: 10.1016/j.amjcard.2017.03.252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 12/19/2022]
Abstract
Postoperative atrial fibrillation (AF) occurs frequently after cardiac surgery and contributes significantly to mortality. Transforming growth factor-beta (TGF-β) is associated with postoperative AF after coronary artery bypass grafting and valve surgery. We performed a prospective study to evaluate the role of TGF-β as a predictor of AF after myectomy. A total of 109 consecutive obstructive hypertrophic cardiomyopathy patients without previous AF who underwent myectomy were identified. We measured plasma TGF-β levels before surgery, monitored heart rhythm until discharge, and followed patients for a mean of 36 ± 10 months. AF was documented in 19 patients (17%). AF patients were older (50 ± 10 vs 43 ± 15 years, p = 0.037). Patients who developed AF had higher plasma TGF-β levels (1,695 ± 2,011 vs 1,099 ± 2,494 pg/ml, p = 0.011), more major adverse cardiac events (32% vs 7%, p = 0.006), and more strokes (16% vs 0%, p = 0.005) than patients who did not. TGF-β level ≥358 pg/ml predicted AF with sensitivity and specificity of 58% and 77% (p = 0.011), respectively. Higher TGF-β levels were associated with pulmonary hypertension (25% vs 8%, p = 0.033). In multivariable regression analysis, age (odds ratio 1.05, 95% confidence interval 1.00 to 1.11, p = 0.041) and TGF-β levels (odds ratio 2.42, 95% confidence interval 1.30 to 4.50, p = 0.005) predicted AF independently. In conclusion, elevated preoperative TGF-β value is an independent predictor of postoperative AF in hypertrophic cardiomyopathy patients after surgical ventricular septal myectomy.
Collapse
|
22
|
Endothelial-to-mesenchymal transition: A novel therapeutic target for cardiovascular diseases. Trends Cardiovasc Med 2017; 27:383-393. [PMID: 28438397 DOI: 10.1016/j.tcm.2017.03.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 02/17/2017] [Accepted: 03/08/2017] [Indexed: 01/14/2023]
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a complex biological process in which endothelial cells lose their specific markers and acquire a mesenchymal or myofibroblastic phenotype. Similar to epithelial-to-mesenchymal transition (EMT), EndMT can be induced by multiple stimulants such as cytokines and metabolic factors that play crucial roles in the development of the cardiovascular system. Recent studies have demonstrated that EndMT may play a significant role in the pathogenesis of cardiovascular diseases (CVDs), and may represent a novel therapeutic target for cardiovascular remodeling and fibrotic disorders. The exact molecular mechanisms involved in cardiovascular pathogenesis that occur as a result of EndMT, however, are not fully explained. In this review, we reveal the multiple intercellular mechanisms of EndMT including stimulants, signaling pathways, and seek to explore the relationship between this biological process, cardiovascular system development, and CVDs that may lead to new therapeutic strategies for the treatment of CVDs.
Collapse
|
23
|
Decreased Endoglin expression in the pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia rat model. Pediatr Surg Int 2017; 33:263-268. [PMID: 27822781 DOI: 10.1007/s00383-016-4004-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
Abstract
AIM OF THE STUDY Pulmonary hypertension (PH) remains a therapeutical challenge in neonates born with congenital diaphragmatic hernia (CDH). Endoglin (Eng), an auxiliary receptor component of the transforming growth factor β (TGFβ) signalling pathway, is expressed mainly by endothelial cells and has been found to be involved in angiogenesis and vascular remodelling. Genetic studies have linked TGFβ and Eng mutations to human arterial PH and other cardiovascular syndromes. Eng interacts with the TGFβ receptors 1 and 2 (Tgfβr1, Tgfβr2). We designed this study to investigate the hypothesis that Eng is altered in the pulmonary vasculature of rats with nitrofen-induced CDH subjected to its interdependency with Tgfβr1 and Tgfβr2. METHODS After ethical approval (Rec 913b), time-pregnant Sprague-Dawley rats received either nitrofen or olive oil on gestational day (D9). The foetuses (n = 22) were sacrificed and divided into CDH and control group on D21. Gene and protein expressions of Eng, Tgfβr1 and Tgfβr2 were assessed via qRT-PCR and western blotting. Immunofluorescence staining for Eng was combined with CD34 to evaluate Eng expression in the pulmonary vasculature. MAIN RESULTS Relative mRNA levels of Eng, Tgfβr1 and Tgfβr2 were significantly downregulated in CDH lungs compared to controls (Eng CDH 0.341 ± 0.022, Eng Ctrl 0.471 ± 0.031, p = 0.0015; Tgfβr1 CDH 0.161 ± 0.008, Tgfβr1 Ctrl 0.194 ± 0.01, p = 0.0114; Tgfβr2 CDH 0.896 ± 0.099, Tgfβr2 Ctrl 1.379 ± 0.081, p = 0.0006) Western blotting confirmed the reduced pulmonary protein expression of these three proteins in the CDH lungs. A markedly diminished endothelial expression of Eng in the pulmonary vasculature of nitrofen-exposed foetuses compared to controls was seen in laser scanning confocal-microscopy. CONCLUSION This study demonstrates for the first time a reduced expression of Endoglin in the pulmonary vasculature of nitrofen-induced CDH. Abnormal Eng/Tgfβr1/Tgfβr2 signalling may contribute to impaired vascular remodelling and development of PH in this CDH animal model.
Collapse
|