1
|
Zhang X, Fu Z, Wang H, Sheng L. Metabolic pathways, genomic alterations, and post-translational modifications in pulmonary hypertension and cancer as therapeutic targets and biomarkers. Front Pharmacol 2024; 15:1490892. [PMID: 39635438 PMCID: PMC11614602 DOI: 10.3389/fphar.2024.1490892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Background Pulmonary hypertension (PH) can lead to right ventricular hypertrophy, significantly increasing mortality rates. This study aims to clarify PH-specific metabolites and their impact on genomic and post-translational modifications (PTMs) in cancer, evaluating DHA and EPA's therapeutic potential to mitigate oxidative stress and inflammation. Methods Data from 289,365 individuals were analyzed using Mendelian randomization to examine 1,400 metabolites' causal roles in PH. Anti-inflammatory and antioxidative effects of DHA and EPA were tested in RAW 264.7 macrophages and cancer cell lines (A549, HCT116, HepG2, LNCaP). Genomic features like CNVs, DNA methylation, tumor mutation burden (TMB), and PTMs were analyzed. DHA and EPA's effects on ROS production and cancer cell proliferation were assessed. Results We identified 57 metabolites associated with PH risk and examined key tumor-related pathways through promoter methylation analysis. DHA and EPA significantly reduced ROS levels and inflammatory markers in macrophages, inhibited the proliferation of various cancer cell lines, and decreased nuclear translocation of SUMOylated proteins during oxidative stress and inflammatory responses. These findings suggest a potential anticancer role through the modulation of stress-related nuclear signaling, as well as a regulatory function on cellular PTMs. Conclusion This study elucidates metabolic and PTM changes in PH and cancer, indicating DHA and EPA's role in reducing oxidative stress and inflammation. These findings support targeting these pathways for early biomarkers and therapies, potentially improving disease management and patient outcomes.
Collapse
Affiliation(s)
- Xiujin Zhang
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | | | | | - Li Sheng
- Department of Cardiology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Chakraborty A, Nathan A, Orcholski M, Agarwal S, Shamskhou EA, Auer N, Mitra A, Guardado ES, Swaminathan G, Condon DF, Yu J, McCarra M, Juul NH, Mallory A, Guzman-Hernandez RA, Yuan K, Rojas V, Crossno JT, Yung LM, Yu PB, Spencer T, Winn RA, Frump A, Karoor V, Lahm T, Hedlin H, Fineman JR, Lafyatis R, Knutsen CNF, Alvira CM, Cornfield DN, de Jesus Perez VA. Wnt7a deficit is associated with dysfunctional angiogenesis in pulmonary arterial hypertension. Eur Respir J 2023; 61:2201625. [PMID: 37024132 PMCID: PMC10259331 DOI: 10.1183/13993003.01625-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) is characterised by loss of microvessels. The Wnt pathways control pulmonary angiogenesis but their role in PAH is incompletely understood. We hypothesised that Wnt activation in pulmonary microvascular endothelial cells (PMVECs) is required for pulmonary angiogenesis, and its loss contributes to PAH. METHODS Lung tissue and PMVECs from healthy and PAH patients were screened for Wnt production. Global and endothelial-specific Wnt7a -/- mice were generated and exposed to chronic hypoxia and Sugen-hypoxia (SuHx). RESULTS Healthy PMVECs demonstrated >6-fold Wnt7a expression during angiogenesis that was absent in PAH PMVECs and lungs. Wnt7a expression correlated with the formation of tip cells, a migratory endothelial phenotype critical for angiogenesis. PAH PMVECs demonstrated reduced vascular endothelial growth factor (VEGF)-induced tip cell formation as evidenced by reduced filopodia formation and motility, which was partially rescued by recombinant Wnt7a. We discovered that Wnt7a promotes VEGF signalling by facilitating Y1175 tyrosine phosphorylation in vascular endothelial growth factor receptor 2 (VEGFR2) through receptor tyrosine kinase-like orphan receptor 2 (ROR2), a Wnt-specific receptor. We found that ROR2 knockdown mimics Wnt7a insufficiency and prevents recovery of tip cell formation with Wnt7a stimulation. While there was no difference between wild-type and endothelial-specific Wnt7a -/- mice under either chronic hypoxia or SuHx, global Wnt7a +/- mice in hypoxia demonstrated higher pulmonary pressures and severe right ventricular and lung vascular remodelling. Similar to PAH, Wnt7a +/- PMVECs exhibited an insufficient angiogenic response to VEGF-A that improved with Wnt7a. CONCLUSIONS Wnt7a promotes VEGF signalling in lung PMVECs and its loss is associated with an insufficient VEGF-A angiogenic response. We propose that Wnt7a deficiency contributes to progressive small vessel loss in PAH.
Collapse
Affiliation(s)
- Ananya Chakraborty
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
- These authors contributed equally
| | - Abinaya Nathan
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
- These authors contributed equally
| | - Mark Orcholski
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | - Natasha Auer
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Ankita Mitra
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | - Gowri Swaminathan
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - David F Condon
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Joyce Yu
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Matthew McCarra
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Nicholas H Juul
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | | | | - Ke Yuan
- Boston Children's Hospital, Boston, MA, USA
| | | | - Joseph T Crossno
- Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Paul B Yu
- Brigham and Women's Hospital, Boston, MA, USA
| | | | - Robert A Winn
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, USA
| | | | | | - Tim Lahm
- National Jewish Center, Denver, CO, USA
| | - Haley Hedlin
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Jeffrey R Fineman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Robert Lafyatis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carsten N F Knutsen
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - Cristina M Alvira
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - David N Cornfield
- Division of Pediatric Pulmonary and Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
3
|
Chen M, Yi F, Qi Y, Zhao B, Zhang Z, He X, Yuan D, Jin T. Whole-exome sequencing in searching for novel variants associated with the development of high altitude pulmonary edema. Gene 2023; 870:147384. [PMID: 37001572 DOI: 10.1016/j.gene.2023.147384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND High altitude pulmonary edema (HAPE) is a high-altitude idiopathic disease with serious consequences due to hypoxia at high altitude, and there is individual genetic susceptibility. Whole-exome sequencing (WES) is an effective tool for studying the genetic etiology of HAPE and can identify potentially novel mutations that may cause protein instability and may contribute to the development of HAPE. MATERIALS AND METHODS A total of 50 unrelated HAPE patients were examined using WES, and the available bioinformatics tools were used to perform an analysis of exonic regions. Using the Phenolyzer program, disease candidate gene analysis was carried out. SIFT, PolyPhen-2, Mutation Taster, CADD, DANN, and I-Mutant software were used to assess the effects of genetic variations on protein function. RESULTS The results showed that rs368502694 (p. R1022Q) located in NOS3, rs1595850639 (p. G61S) located in MYBPC3, and rs1367895529 (p. R333H) located in ITGAV were correlated with a high risk of HAPE, and thus could be regarded as potential genetic variations associated with HAPE. CONCLUSION WES was used in this study for the first time to directly screen genetic variations related to HAPE. Notably, our study offers fresh information for the subsequent investigation into the etiology of HAPE.
Collapse
Affiliation(s)
- Mingyue Chen
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Faling Yi
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Yijin Qi
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Beibei Zhao
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Zhanhao Zhang
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Xue He
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Dongya Yuan
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China.
| | - Tianbo Jin
- Key Laboratory of High Altitude Hypoxia Environment and Life Health, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi'an, Shaanxi 710069, China; Shaanxi Provincial Key Laboratory of Biotechnology, Northwest University, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
4
|
Blanchard N, Link PA, Farkas D, Harmon B, Hudson J, Bogamuwa S, Piper B, Authelet K, Cool CD, Heise RL, Freishtat R, Farkas L. Dichotomous role of integrin-β5 in lung endothelial cells. Pulm Circ 2022; 12:e12156. [PMID: 36438452 PMCID: PMC9684688 DOI: 10.1002/pul2.12156] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, devastating disease, and its main histological manifestation is an occlusive pulmonary arteriopathy. One important functional component of PAH is aberrant endothelial cell (EC) function including apoptosis-resistance, unchecked proliferation, and impaired migration. The mechanisms leading to and maintaining physiologic and aberrant EC function are not fully understood. Here, we tested the hypothesis that in PAH, ECs have increased expression of the transmembrane protein integrin-β5, which contributes to migration and survival under physiologic and pathological conditions, but also to endothelial-to-mesenchymal transition (EnMT). We found that elevated integrin-β5 expression in pulmonary artery lesions and lung tissue from PAH patients and rats with PH induced by chronic hypoxia and injection of CD117+ rat lung EC clones. These EC clones exhibited elevated expression of integrin-β5 and its heterodimerization partner integrin-αν and showed accelerated barrier formation. Inhibition of integrin-ανβ5 in vitro partially blocked transforming growth factor (TGF)-β1-induced EnMT gene expression in rat lung control ECs and less in rat lung EC clones and human lung microvascular ECs. Inhibition of integrin-ανβ5 promoted endothelial dysfunction as shown by reduced migration in a scratch assay and increased apoptosis in synergism with TGF-β1. In vivo, blocking of integrin-ανβ5 exaggerated PH induced by chronic hypoxia and CD117+ EC clones in rats. In summary, we found a role for integrin-ανβ5 in lung endothelial survival and migration, but also a partial contribution to TGF-β1-induced EnMT gene expression. Our results suggest that integrin-ανβ5 is required for physiologic function of ECs and lung vascular homeostasis.
Collapse
Affiliation(s)
- Neil Blanchard
- Department of Orthopedic SurgeryUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Patrick A. Link
- Departments of Physiology and Biomedical EngineeringMayo ClinicRochesterMichiganUSA
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Daniela Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Brennan Harmon
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Jaylen Hudson
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Srimathi Bogamuwa
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bryce Piper
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Kayla Authelet
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Carlyne D. Cool
- Department of PathologyUniversity of Colorado at DenverDenverColoradoUSA
| | - Rebecca L. Heise
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Robert Freishtat
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Laszlo Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Physiology and BiophysicsVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
5
|
Pienkos S, Gallego N, Condon DF, Cruz-Utrilla A, Ochoa N, Nevado J, Arias P, Agarwal S, Patel H, Chakraborty A, Lapunzina P, Escribano P, Tenorio-Castaño J, de Jesús Pérez VA. Novel TNIP2 and TRAF2 Variants Are Implicated in the Pathogenesis of Pulmonary Arterial Hypertension. Front Med (Lausanne) 2021; 8:625763. [PMID: 33996849 PMCID: PMC8119639 DOI: 10.3389/fmed.2021.625763] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Pulmonary arterial hypertension (PAH) is a rare disease characterized by pulmonary vascular remodeling and right heart failure. Specific genetic variants increase the incidence of PAH in carriers with a family history of PAH, those who suffer from certain medical conditions, and even those with no apparent risk factors. Inflammation and immune dysregulation are related to vascular remodeling in PAH, but whether genetic susceptibility modifies the PAH immune response is unclear. TNIP2 and TRAF2 encode for immunomodulatory proteins that regulate NF-κB activation, a transcription factor complex associated with inflammation and vascular remodeling in PAH. Methods: Two unrelated families with PAH cases underwent whole-exome sequencing (WES). A custom pipeline for variant prioritization was carried out to obtain candidate variants. To determine the impact of TNIP2 and TRAF2 in cell proliferation, we performed an MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] assay on healthy lung pericytes transfected with siRNA specific for each gene. To measure the effect of loss of TNIP2 and TRAF2 on NF-kappa-beta (NF-κB) activity, we measured levels of Phospho-p65-NF-κB in siRNA-transfected pericytes using western immunoblotting. Results: We discovered a novel missense variant in the TNIP2 gene in two affected individuals from the same family. The two patients had a complex form of PAH with interatrial communication and scleroderma. In the second family, WES of the proband with PAH and primary biliary cirrhosis revealed a de novo protein-truncating variant in the TRAF2. The knockdown of TNIP2 and TRAF2 increased NF-κB activity in healthy lung pericytes, which correlated with a significant increase in proliferation over 24 h. Conclusions: We have identified two rare novel variants in TNIP2 and TRAF2 using WES. We speculate that loss of function in these genes promotes pulmonary vascular remodeling by allowing overactivation of the NF-κB signaling activity. Our findings support a role for WES in helping identify novel genetic variants associated with dysfunctional immune response in PAH.
Collapse
Affiliation(s)
- Shaun Pienkos
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Natalia Gallego
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Alejandro Cruz-Utrilla
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Nuria Ochoa
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Julián Nevado
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pedro Arias
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| | - Pablo Lapunzina
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Pilar Escribano
- Pulmonary Hypertension Unit, Department of Cardiology, Hospital Universitario Doce de Octubre, Madrid, Spain
- Centro de Investigación Biomedica en Red en Enfermedades Cardiovasculares, Instituto de Salud Carlos III (CIBERCV), Madrid, Spain
| | - Jair Tenorio-Castaño
- Medical and Molecular Genetics Institute (INGEMM), IdiPaz, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
- Intellectual Disability, TeleHealth, Autism and Congenital Anomalies (ITHACA), European Reference Network on Rare Congenital Malformations and Rare Intellectual Disability, Brussels, Belgium
| | - Vinicio A. de Jesús Pérez
- Division of Pulmonary and Critical Care Medicine and Department of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
6
|
Jandl K, Marsh LM, Hoffmann J, Mutgan AC, Baum O, Bloch W, Thekkekara-Puthenparampil H, Kolb D, Sinn K, Klepetko W, Heinemann A, Olschewski A, Olschewski H, Kwapiszewska G. Basement Membrane Remodeling Controls Endothelial Function in Idiopathic Pulmonary Arterial Hypertension. Am J Respir Cell Mol Biol 2020; 63:104-117. [PMID: 32160015 DOI: 10.1165/rcmb.2019-0303oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix (ECM) increasingly emerges as an active driver in several diseases, including idiopathic pulmonary arterial hypertension (IPAH). The basement membrane (BM) is a specialized class of ECM proteins. In pulmonary arteries, the BM is in close contact and direct proximity to vascular cells, including endothelial cells. So far, the role of the BM has remained underinvestigated in IPAH. Here, we aimed to shed light on the involvement of the BM in IPAH, by addressing its structure, composition, and function. On an ultrastructural level, we observed a marked increase in BM thickness in IPAH pulmonary vessels. BM composition was distinct in small and large vessels and altered in IPAH. Proteoglycans were mostly responsible for distinction between smaller and larger vessels, whereas BM collagens and laminins were more abundantly expressed in IPAH. Type IV collagen and laminin both strengthened endothelial barrier integrity. However, only type IV collagen concentration dependently increased cell adhesion of both donor and IPAH-derived pulmonary arterial endothelial cells (PAECs) and induced nuclear translocation of mechanosensitive transcriptional coactivator of the hippo pathway YAP (Yes-activated protein). On the other hand, laminin caused cytoplasmic retention of YAP in IPAH PAECs. Accordingly, silencing of COL4A5 and LAMC1, respectively, differentially affected tight junction formation and barrier integrity in both donor and IPAH PAECs. Collectively, our results highlight the importance of a well-maintained BM homeostasis. By linking changes in BM structure and composition to altered endothelial cell function, we here suggest an active involvement of the BM in IPAH pathogenesis.
Collapse
Affiliation(s)
- Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Division of Pharmacology and
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Julia Hoffmann
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | | | - Oliver Baum
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wilhelm Bloch
- German Sports University Cologne, Cologne, Germany; and
| | | | | | - Katharina Sinn
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria.,Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, and
| | - Horst Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 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, Austria.,Division of Physiology, Otto Loewi Research Center
| |
Collapse
|
7
|
Miao Y, Tian L, Martin M, Paige SL, Galdos FX, Li J, Klein A, Zhang H, Ma N, Wei Y, Stewart M, Lee S, Moonen JR, Zhang B, Grossfeld P, Mital S, Chitayat D, Wu JC, Rabinovitch M, Nelson TJ, Nie S, Wu SM, Gu M. Intrinsic Endocardial Defects Contribute to Hypoplastic Left Heart Syndrome. Cell Stem Cell 2020; 27:574-589.e8. [PMID: 32810435 PMCID: PMC7541479 DOI: 10.1016/j.stem.2020.07.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/21/2020] [Accepted: 07/15/2020] [Indexed: 01/03/2023]
Abstract
Hypoplastic left heart syndrome (HLHS) is a complex congenital heart disease characterized by abnormalities in the left ventricle, associated valves, and ascending aorta. Studies have shown intrinsic myocardial defects but do not sufficiently explain developmental defects in the endocardial-derived cardiac valve, septum, and vasculature. Here, we identify a developmentally impaired endocardial population in HLHS through single-cell RNA profiling of hiPSC-derived endocardium and human fetal heart tissue with an underdeveloped left ventricle. Intrinsic endocardial defects contribute to abnormal endothelial-to-mesenchymal transition, NOTCH signaling, and extracellular matrix organization, key factors in valve formation. Endocardial abnormalities cause reduced cardiomyocyte proliferation and maturation by disrupting fibronectin-integrin signaling, consistent with recently described de novo HLHS mutations associated with abnormal endocardial gene and fibronectin regulation. Together, these results reveal a critical role for endocardium in HLHS etiology and provide a rationale for considering endocardial function in regenerative strategies.
Collapse
Affiliation(s)
- Yifei Miao
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lei Tian
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Marcy Martin
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sharon L Paige
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Francisco X Galdos
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jibiao Li
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alyssa Klein
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Hao Zhang
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ning Ma
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Yuning Wei
- Center for Personal Dynamic Regulomes, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Maria Stewart
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Soah Lee
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jan-Renier Moonen
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Bing Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Xin Hua Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Paul Grossfeld
- Department of Pediatrics, UCSD School of Medicine, La Jolla, CA 92093, USA
| | - Seema Mital
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - David Chitayat
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada; The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Timothy J Nelson
- Division of General Internal Medicine, Division of Pediatric Cardiology, and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuyi Nie
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sean M Wu
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell and Regenerative Biology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Mingxia Gu
- Department of Pediatrics, Division of Pediatric Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA; Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford School of Medicine, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA; Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
8
|
Wang M, Zhuang D, Mei M, Ma H, Li Z, He F, Cheng G, Lin G, Zhou W. Frequent mutation of hypoxia-related genes in persistent pulmonary hypertension of the newborn. Respir Res 2020; 21:53. [PMID: 32054482 PMCID: PMC7020588 DOI: 10.1186/s12931-020-1314-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 02/04/2020] [Indexed: 12/16/2022] Open
Abstract
Aims Persistent pulmonary hypertension of the newborn (PPHN) is characterized by sustained high levels of pulmonary vascular resistance after birth with etiology unclear; Arterial blood oxygen saturation of Tibetan newborns at high latitudes is higher than that of Han newborns at low latitudes, suggesting that genetic adaptation may allow sufficient oxygen to confer Tibetan populations with resistance to pulmonary hypertension; We have previously identified genetic factors related to PPHN through candidate gene sequencing; In this study, we first performed whole exome sequencing in PPHN patients to screen for genetic-related factors. Methods and results In this two-phase genetic study, we first sequenced the whole exome of 20 Tibetan PPHN patients and compared it with the published genome sequences of 50 healthy high-altitude Tibetanshypoxia-related genes, a total of 166 PPHN-related variants were found, of which 49% were from 43 hypoxia-related genes; considering many studies have shown that the differences in the genetic background between Tibet and Han are characterized by hypoxia-related genetic polymorphisms, so it is necessary to further verify whether the association between hypoxia-related variants and PPHN is independent of high-altitude life. During the validation phase, 237 hypoxia-related genes were sequenced in another 80 Han PPHN patients living in low altitude areas, including genes at the discovery stage and known hypoxia tolerance, of which 413 variants from 127 of these genes were shown to be significantly associated with PPHN.hypoxia-related genes. Conclusions Our results indicates that the association of hypoxia-related genes with PPHN does not depend on high-altitude life, at the same time, 21 rare mutations associated with PPHN were also found, including three rare variants of the tubulin tyrosine ligase-like family member 3 gene (TTLL3:p.E317K, TTLL3:p.P777S) and the integrin subunit alpha M gene (ITGAM:p.E1071D). These novel findings provide important information on the genetic basis of PPHN.
Collapse
Affiliation(s)
- Mingbang Wang
- Shanghai Key Laboratory of Birth Defects, National Health Commision (NHC) Key Laboratory of Neonatal Diseases, Division of Neonatology, National Center for Children's Health, Children's Hospital of Fudan University, Shanghai, 201102, China.
| | - Deyi Zhuang
- Xiamen Key Laboratory of Neonatal Diseases, Neonatal Medical Center, Xiamen Children's Hospital, Children's Hospital of Fudan University (Xiamen Branch), Xiamen, 361006, Fujian, China
| | - Mei Mei
- Division of Pulmonology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Haiyan Ma
- Zhuhai Maternal and Children's Hospital, Zhuhai, 519001, Guangdong, China
| | - Zixiu Li
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | | | - Guoqiang Cheng
- Division of Neonatology, Children's Hospital of Fudan University, Shanghai, 201102, China.,Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai, 200436, China
| | - Guang Lin
- Zhuhai Maternal and Children's Hospital, Zhuhai, 519001, Guangdong, China.
| | - Wenhao Zhou
- Shanghai Key Laboratory of Birth Defects, National Health Commision (NHC) Key Laboratory of Neonatal Diseases, Division of Neonatology, National Center for Children's Health, Children's Hospital of Fudan University, Shanghai, 201102, China.
| |
Collapse
|
9
|
Yuan K, Shamskhou EA, Orcholski ME, Nathan A, Reddy S, Honda H, Mani V, Zeng Y, Ozen MO, Wang L, Demirci U, Tian W, Nicolls MR, de Jesus Perez VA. Loss of Endothelium-Derived Wnt5a Is Associated With Reduced Pericyte Recruitment and Small Vessel Loss in Pulmonary Arterial Hypertension. Circulation 2020; 139:1710-1724. [PMID: 30586764 DOI: 10.1161/circulationaha.118.037642] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a life-threatening disorder of the pulmonary circulation associated with loss and impaired regeneration of microvessels. Reduced pericyte coverage of pulmonary microvessels is a pathological feature of PAH and is caused partly by the inability of pericytes to respond to signaling cues from neighboring pulmonary microvascular endothelial cells (PMVECs). We have shown that activation of the Wnt/planar cell polarity pathway is required for pericyte recruitment, but whether production and release of specific Wnt ligands by PMVECs are responsible for Wnt/planar cell polarity activation in pericytes is unknown. METHODS Isolation of pericytes and PMVECs from healthy donor and PAH lungs was carried out with 3G5 or CD31 antibody-conjugated magnetic beads. Wnt expression profile of PMVECs was documented via quantitative polymerase chain reaction with a Wnt primer library. Exosome purification from PMVEC media was carried out with the ExoTIC device. Hemodynamic profile, right ventricular function, and pulmonary vascular morphometry were obtained in a conditional endothelium-specific Wnt5a knockout ( Wnt5aECKO) mouse model under normoxia, chronic hypoxia, and hypoxia recovery. RESULTS Quantification of Wnt ligand expression in healthy PMVECs cocultured with pericytes demonstrated a 35-fold increase in Wnt5a, a known Wnt/planar cell polarity ligand. This Wnt5a spike was not seen in PAH PMVECs, which correlated with an inability to recruit pericytes in Matrigel coculture assays. Exosomes purified from media demonstrated an increase in Wnt5a content when healthy PMVECs were cocultured with pericytes, a finding that was not observed in exosomes of PAH PMVECs. Furthermore, the addition of either recombinant Wnt5a or purified healthy PMVEC exosomes increased pericyte recruitment to PAH PMVECs in coculture studies. Although no differences were noted in normoxia and chronic hypoxia, Wnt5aECKO mice demonstrated persistent pulmonary hypertension and right ventricular failure 4 weeks after recovery from chronic hypoxia, which correlated with significant reduction, muscularization, and decreased pericyte coverage of microvessels. CONCLUSIONS We identify Wnt5a as a key mediator for the establishment of pulmonary endothelium-pericyte interactions, and its loss could contribute to PAH by reducing the viability of newly formed vessels. We speculate that therapies that mimic or restore Wnt5a production could help prevent loss of small vessels in PAH.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Elya A Shamskhou
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Mark E Orcholski
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Abinaya Nathan
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| | - Sushma Reddy
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Division of Pediatric Cardiology (S.R.), Stanford University, Palo Alto, CA
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, Japan (H.H.)
| | - Vigneshwaran Mani
- Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Yitian Zeng
- Department of Materials Science and Engineering (Y.Z.), Stanford University, Palo Alto, CA
| | - Mehmet O Ozen
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Lingli Wang
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Pediatrics (L.W.), Stanford University, Palo Alto, CA
| | - Utkan Demirci
- Department of Radiology, Canary Center for Early Cancer Detection (V.M., M.O.O., U.D.), Stanford University, Palo Alto, CA
| | - Wen Tian
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Medicine, VA Palo Alto Health Care System/Stanford University, CA (W.T., M.R.N.)
| | - Mark R Nicolls
- Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Department of Medicine, VA Palo Alto Health Care System/Stanford University, CA (W.T., M.R.N.)
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine (K.Y., E.A.S., M.E.O., A.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Stanford Cardiovascular Institute (K.Y., E.A.S., M.E.O., A.N., S.R., M.O.O, L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA.,Wall Center for Pulmonary Vascular Research (K.Y., E.A.S., M.E.O., A.N., L.W., W.T., M.R.N., V.A.d.J.P.), Stanford University, Palo Alto, CA
| |
Collapse
|
10
|
Tielemans B, Stoian L, Gijsbers R, Michiels A, Wagenaar A, Farre Marti R, Belge C, Delcroix M, Quarck R. Cytokines trigger disruption of endothelium barrier function and p38 MAP kinase activation in BMPR2-silenced human lung microvascular endothelial cells. Pulm Circ 2019; 9:2045894019883607. [PMID: 31692724 PMCID: PMC6811766 DOI: 10.1177/2045894019883607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
The bone morphogenetic protein receptor II (BMPRII) signaling pathway is impaired
in pulmonary arterial hypertension and mutations in the BMPR2
gene have been observed in both heritable and idiopathic pulmonary arterial
hypertension. However, all BMPR2 mutation carriers do not
develop pulmonary arterial hypertension, and inflammation could trigger the
development of the disease in BMPR2 mutation carriers.
Circulating levels and/or lung tissue expression of cytokines such as tumor
necrosis factor-α or interleukin-18 are elevated in patients with pulmonary
arterial hypertension and could be involved in the pathogenesis of pulmonary
arterial hypertension. We consequently hypothesized that cytokines could trigger
endothelial dysfunction in addition to impaired BMPRII signaling. Our aim was to
determine whether impairment of BMPRII signaling might affect endothelium
barrier function and adhesiveness to monocytes, in response to cytokines.
BMPR2 was silenced in human lung microvascular endothelial
cells (HLMVECs) using lentiviral vectors encoding microRNA-based hairpins.
Effects of tumor necrosis factor-α and interleukin-18 on HLMVEC adhesiveness to
the human monocyte cell line THP-1, adhesion molecule expression, endothelial
barrier function and activation of P38MAPK were investigated in vitro. Stable
BMPR2 silencing in HLMVECs resulted in impaired endothelial
barrier function and constitutive activation of P38MAPK. Adhesiveness of
BMPR2-silenced HLMVECs to THP-1 cells was enhanced by tumor
necrosis factor-α and interleukin-18 through ICAM-1 adhesion molecule.
Interestingly, tumor necrosis factor-α induced activation of P38MAPK and
disrupted endothelial barrier function in BMPR2-silenced
HLMVECs. Altogether, our findings showed that stable BMPR2
silencing resulted in impaired endothelial barrier function and activation of
P38MAPK in HLMVECs. In BMPR2-silenced HLMVECs, cytokines
enhanced adhesiveness capacities, activation of P38MAPK and impaired endothelial
barrier function suggesting that cytokines could trigger the development of
pulmonary arterial hypertension in a context of impaired BMPRII signaling
pathway.
Collapse
Affiliation(s)
- Birger Tielemans
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Leanda Stoian
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium.,Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Annelies Michiels
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven - University of Leuven, Leuven, Belgium
| | - Allard Wagenaar
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Ricard Farre Marti
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Catharina Belge
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Marion Delcroix
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rozenn Quarck
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Lee HW, Park SH. Elevated microRNA-135a is associated with pulmonary arterial hypertension in experimental mouse model. Oncotarget 2018; 8:35609-35618. [PMID: 28415675 PMCID: PMC5482602 DOI: 10.18632/oncotarget.16011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
Multiple causes are associated with the complex mechanism of pathogenesis of pulmonary arterial hypertension (PAH), but the molecular pathway in the pathogenesis of PAH is still insufficiently understood. In this study, we investigated epigenetic changes that cause PAH induced by exposure to combined Th2 antigen (Ovalbumin, OVA) and urban particulate matter (PM) in mice. To address that, we focused on the epigenetic mechanism, linked to microRNA (miR)-135a. We found that miR-135a levels were significantly increased, and levels of bone morphogenetic protein receptor type II (BMPR2) which is the target of miR-135a, were significantly decreased in this experimental PAH mouse model. Therefore to evaluate the role of miR-135a, we injected AntagomiR-135a into this mouse model. AntagomiR-135a injected mice showed decreased right ventricular systolic pressures (RVSPs), right ventricular hypertrophy (RVH), and the percentage of severely thickened pulmonary arteries compared to control scrambled miRNA injected mice. Both mRNA and protein expression of BMPR2 were recovered in the AntagomiR-135a injected mice compared to control mice. Our study understands if miR-135a could serve as a biomarker helping to manage PAH. The blocking of miR-135a could lead to new therapeutic modalities to alleviate exacerbation of PAH caused by exposure to Th2 antigen and urban air pollution.
Collapse
Affiliation(s)
- Hyun-Wook Lee
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| | - Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York, USA
| |
Collapse
|
12
|
Ranchoux B, Harvey LD, Ayon RJ, Babicheva A, Bonnet S, Chan SY, Yuan JXJ, Perez VDJ. Endothelial dysfunction in pulmonary arterial hypertension: an evolving landscape (2017 Grover Conference Series). Pulm Circ 2018; 8:2045893217752912. [PMID: 29283043 PMCID: PMC5798691 DOI: 10.1177/2045893217752912] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Endothelial dysfunction is a major player in the development and progression of vascular pathology in pulmonary arterial hypertension (PAH), a disease associated with small vessel loss and obstructive vasculopathy that leads to increased pulmonary vascular resistance, subsequent right heart failure, and premature death. Over the past ten years, there has been tremendous progress in our understanding of pulmonary endothelial biology as it pertains to the genetic and molecular mechanisms that orchestrate the endothelial response to direct or indirect injury, and how their dysregulation can contribute to the pathogenesis of PAH. As one of the major topics included in the 2017 Grover Conference Series, discussion centered on recent developments in four areas of pulmonary endothelial biology: (1) angiogenesis; (2) endothelial-mesenchymal transition (EndMT); (3) epigenetics; and (4) biology of voltage-gated ion channels. The present review will summarize the content of these discussions and provide a perspective on the most promising aspects of endothelial dysfunction that may be amenable for therapeutic development.
Collapse
Affiliation(s)
| | - Lloyd D. Harvey
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Ramon J. Ayon
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Aleksandra Babicheva
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | | | - Stephen Y. Chan
- University of Pittsburgh Vascular Medicine Institute Division of Cardiology, Pittsburgh, PA, USA
| | - Jason X.-J. Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, AZ, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
- The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford University Medical Center, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University Medical Center, Stanford, CA, USA
| |
Collapse
|
13
|
Bonnet S, Provencher S, Guignabert C, Perros F, Boucherat O, Schermuly RT, Hassoun PM, Rabinovitch M, Nicolls MR, Humbert M. Translating Research into Improved Patient Care in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2017; 195:583-595. [PMID: 27649290 PMCID: PMC5440916 DOI: 10.1164/rccm.201607-1515pp] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Sébastien Bonnet
- 1 Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada.,2 Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Steeve Provencher
- 1 Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada.,2 Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Christophe Guignabert
- 3 INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, Paris, France.,4 Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, Paris, France
| | - Frédéric Perros
- 3 INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, Paris, France.,4 Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, Paris, France
| | - Olivier Boucherat
- 1 Pulmonary Hypertension Research Group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, Quebec, Canada
| | - Ralph Theo Schermuly
- 5 Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University Giessen, Giessen, Germany
| | - Paul M Hassoun
- 6 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | - Mark R Nicolls
- 8 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California.,9 VA Palo Alto Health Care System, Palo Alto, California; and
| | - Marc Humbert
- 3 INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, Paris, France.,4 Université Paris-Sud and Université Paris-Saclay, Kremlin-Bicêtre, Paris, France.,10 Assistance Publique-Hôpitaux de Paris, Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, Département Hospitalo-Universitaire Thorax Innovation, Hôpital de Bicêtre, Paris, France
| |
Collapse
|
14
|
Sa S, Gu M, Chappell J, Shao NY, Ameen M, Elliott KAT, Li D, Grubert F, Li CG, Taylor S, Cao A, Ma Y, Fong R, Nguyen L, Wu JC, Snyder MP, Rabinovitch M. Induced Pluripotent Stem Cell Model of Pulmonary Arterial Hypertension Reveals Novel Gene Expression and Patient Specificity. Am J Respir Crit Care Med 2017; 195:930-941. [PMID: 27779452 DOI: 10.1164/rccm.201606-1200oc] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RATIONALE Idiopathic or heritable pulmonary arterial hypertension is characterized by loss and obliteration of lung vasculature. Endothelial cell dysfunction is pivotal to the pathophysiology, but different causal mechanisms may reflect a need for patient-tailored therapies. OBJECTIVES Endothelial cells differentiated from induced pluripotent stem cells were compared with pulmonary arterial endothelial cells from the same patients with idiopathic or heritable pulmonary arterial hypertension, to determine whether they shared functional abnormalities and altered gene expression patterns that differed from those in unused donor cells. We then investigated whether endothelial cells differentiated from pluripotent cells could serve as surrogates to test emerging therapies. METHODS Functional changes assessed included adhesion, migration, tube formation, and propensity to apoptosis. Expression of bone morphogenetic protein receptor type 2 (BMPR2) and its target, collagen IV, signaling of the phosphorylated form of the mothers against decapentaplegic proteins (pSMAD1/5), and transcriptomic profiles were also analyzed. MEASUREMENTS AND MAIN RESULTS Native pulmonary arterial and induced pluripotent stem cell-derived endothelial cells from patients with idiopathic and heritable pulmonary arterial hypertension compared with control subjects showed a similar reduction in adhesion, migration, survival, and tube formation, and decreased BMPR2 and downstream signaling and collagen IV expression. Transcriptomic profiling revealed high kisspeptin 1 (KISS1) related to reduced migration and low carboxylesterase 1 (CES1), to impaired survival in patient cells. A beneficial angiogenic response to potential therapies, FK506 and Elafin, was related to reduced slit guidance ligand 3 (SLIT3), an antimigratory factor. CONCLUSIONS Despite the site of disease in the lung, our study indicates that induced pluripotent stem cell-derived endothelial cells are useful surrogates to uncover novel features related to disease mechanisms and to better match patients to therapies.
Collapse
Affiliation(s)
- Silin Sa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mingxia Gu
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | | | - Ning-Yi Shao
- 2 Cardiovascular Institute.,4 Department of Medicine, and
| | - Mohamed Ameen
- 2 Cardiovascular Institute.,5 Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Kathryn A T Elliott
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Dan Li
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Fabian Grubert
- 5 Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Caiyun G Li
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Shalina Taylor
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Aiqin Cao
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Yu Ma
- 2 Cardiovascular Institute.,5 Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Ryan Fong
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Long Nguyen
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Joseph C Wu
- 2 Cardiovascular Institute.,4 Department of Medicine, and
| | - Michael P Snyder
- 2 Cardiovascular Institute.,5 Department of Genetics, Stanford University School of Medicine, Stanford, California
| | - Marlene Rabinovitch
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| |
Collapse
|
15
|
Patient-Specific iPSC-Derived Endothelial Cells Uncover Pathways that Protect against Pulmonary Hypertension in BMPR2 Mutation Carriers. Cell Stem Cell 2016; 20:490-504.e5. [PMID: 28017794 DOI: 10.1016/j.stem.2016.08.019] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/11/2016] [Accepted: 08/19/2016] [Indexed: 01/15/2023]
Abstract
In familial pulmonary arterial hypertension (FPAH), the autosomal dominant disease-causing BMPR2 mutation is only 20% penetrant, suggesting that genetic variation provides modifiers that alleviate the disease. Here, we used comparison of induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) from three families with unaffected mutation carriers (UMCs), FPAH patients, and gender-matched controls to investigate this variation. Our analysis identified features of UMC iPSC-ECs related to modifiers of BMPR2 signaling or to differentially expressed genes. FPAH-iPSC-ECs showed reduced adhesion, survival, migration, and angiogenesis compared to UMC-iPSC-ECs and control cells. The "rescued" phenotype of UMC cells was related to an increase in specific BMPR2 activators and/or a reduction in inhibitors, and the improved cell adhesion could be attributed to preservation of related signaling. The improved survival was related to increased BIRC3 and was independent of BMPR2. Our findings therefore highlight protective modifiers for FPAH that could help inform development of future treatment strategies.
Collapse
|
16
|
Modulation of BMP signalling by integrins. Biochem Soc Trans 2016; 44:1465-1473. [DOI: 10.1042/bst20160111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/11/2016] [Accepted: 07/15/2016] [Indexed: 12/17/2022]
Abstract
The bone morphogenetic protein (BMP) pathway is a major conserved signalling pathway with diverse roles in development and homeostasis. Given that cells exist in three-dimensional environments, one important area is to understand how the BMP pathway operates within such complex cellular environments. The extracellular matrix contains information regarding tissue architecture and its mechanical properties that is transmitted to the cell via integrin receptors. In this review, I describe various examples of modulation of the BMP pathway by integrins. In the case of the Drosophila embryo and some cell line-based studies, integrins have been found to enhance BMP responses through different mechanisms, such as enhancement of BMP ligand–receptor binding and effects on Smad phosphorylation or stability. In these contexts, BMP-dependent activation of integrins is a common theme. However, I also discuss examples where integrins inhibit the BMP pathway, highlighting the context-dependent nature of integrin–BMP cross-talk.
Collapse
|
17
|
Yuan K, Shao NY, Hennigs JK, Discipulo M, Orcholski ME, Shamskhou E, Richter A, Hu X, Wu JC, de Jesus Perez VA. Increased Pyruvate Dehydrogenase Kinase 4 Expression in Lung Pericytes Is Associated with Reduced Endothelial-Pericyte Interactions and Small Vessel Loss in Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2500-14. [PMID: 27456128 DOI: 10.1016/j.ajpath.2016.05.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 04/26/2016] [Accepted: 05/25/2016] [Indexed: 12/23/2022]
Abstract
Reduced endothelial-pericyte interactions are linked to progressive small vessel loss in pulmonary arterial hypertension (PAH), but the molecular mechanisms underlying this disease remain poorly understood. To identify relevant gene candidates associated with aberrant pericyte behavior, we performed a transcriptome analysis of patient-derived donor control and PAH lung pericytes followed by functional genomics analysis. Compared with donor control cells, PAH pericytes had significant enrichment of genes involved in various metabolic processes, the top hit being PDK4, a gene coding for an enzyme that suppresses mitochondrial activity in favor of glycolysis. Given reports that link reduced mitochondrial activity with increased PAH cell proliferation, we hypothesized that increased PDK4 is associated with PAH pericyte hyperproliferation and reduced endothelial-pericyte interactions. We found that PDK4 gene and protein expression was significantly elevated in PAH pericytes and correlated with reduced mitochondrial metabolism, higher rates of glycolysis, and hyperproliferation. Importantly, reducing PDK4 levels restored mitochondrial metabolism, reduced cell proliferation, and improved endothelial-pericyte interactions. To our knowledge, this is the first study that documents significant differences in gene expression between human donor control and PAH lung pericytes and the link between mitochondrial dysfunction and aberrant endothelial-pericyte interactions in PAH. Comprehensive characterization of these candidate genes could provide novel therapeutic targets to improve endothelial-pericyte interactions and prevent small vessel loss in PAH.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Ning-Yi Shao
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Division of Cardiology, Stanford University, Stanford, California
| | - Jan K Hennigs
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| | - Marielle Discipulo
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | - Mark E Orcholski
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Elya Shamskhou
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Alice Richter
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California
| | - Xinqian Hu
- Department of Genetics, Stanford University, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, California; Division of Cardiology, Stanford University, Stanford, California
| | - Vinicio A de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, California; Stanford Cardiovascular Institute, Stanford University, Stanford, California.
| |
Collapse
|
18
|
Takahashi J, Orcholski M, Yuan K, de Jesus Perez V. PDGF-dependent β-catenin activation is associated with abnormal pulmonary artery smooth muscle cell proliferation in pulmonary arterial hypertension. FEBS Lett 2016; 590:101-9. [PMID: 26787464 DOI: 10.1002/1873-3468.12038] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/30/2015] [Accepted: 12/02/2015] [Indexed: 12/29/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by excessive pulmonary arterial smooth muscle cells (PASMCs) growth, partially in response to PDGF-BB but whether this is dependent on β-catenin (βC) activation is unclear. Compared to healthy cells, PAH PASMCs demonstrate higher levels of proliferation both at baseline and with PDGF-BB that correlate with GSK3β dependent βC activation. We show that βC knockdown but not Wnt5a stimulation reduces PDGF-BB dependent growth and normalizes PAH PASMCs proliferation. These findings support that cross-talk between PDGF and Wnt signaling modulates PASMC proliferation and suggest that βC targeted therapies could treat abnormal vascular remodeling in PAH.
Collapse
Affiliation(s)
- Jack Takahashi
- Division of Pulmonary and Critical Care Medicine, Stanford, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford, CA, USA.,Stanford Cardiovascular Institute, Stanford University Medical Center, CA, USA
| | - Mark Orcholski
- Division of Pulmonary and Critical Care Medicine, Stanford, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford, CA, USA.,Stanford Cardiovascular Institute, Stanford University Medical Center, CA, USA
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford, CA, USA.,Stanford Cardiovascular Institute, Stanford University Medical Center, CA, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford, CA, USA.,The Vera Moulton Wall Center for Pulmonary Vascular Medicine, Stanford, CA, USA.,Stanford Cardiovascular Institute, Stanford University Medical Center, CA, USA
| |
Collapse
|
19
|
Rhodes CJ, Im H, Cao A, Hennigs JK, Wang L, Sa S, Chen PI, Nickel NP, Miyagawa K, Hopper RK, Tojais NF, Li CG, Gu M, Spiekerkoetter E, Xian Z, Chen R, Zhao M, Kaschwich M, Del Rosario PA, Bernstein D, Zamanian RT, Wu JC, Snyder MP, Rabinovitch M. RNA Sequencing Analysis Detection of a Novel Pathway of Endothelial Dysfunction in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2015; 192:356-66. [PMID: 26030479 DOI: 10.1164/rccm.201408-1528oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Pulmonary arterial hypertension is characterized by endothelial dysregulation, but global changes in gene expression have not been related to perturbations in function. OBJECTIVES RNA sequencing was used to discriminate changes in transcriptomes of endothelial cells cultured from lungs of patients with idiopathic pulmonary arterial hypertension versus control subjects and to assess the functional significance of major differentially expressed transcripts. METHODS The endothelial transcriptomes from the lungs of seven control subjects and six patients with idiopathic pulmonary arterial hypertension were analyzed. Differentially expressed genes were related to bone morphogenetic protein type 2 receptor (BMPR2) signaling. Those down-regulated were assessed for function in cultured cells and in a transgenic mouse. MEASUREMENTS AND MAIN RESULTS Fold differences in 10 genes were significant (P < 0.05), four increased and six decreased in patients versus control subjects. No patient was mutant for BMPR2. However, knockdown of BMPR2 by siRNA in control pulmonary arterial endothelial cells recapitulated 6 of 10 patient-related gene changes, including decreased collagen IV (COL4A1, COL4A2) and ephrinA1 (EFNA1). Reduction of BMPR2-regulated transcripts was related to decreased β-catenin. Reducing COL4A1, COL4A2, and EFNA1 by siRNA inhibited pulmonary endothelial adhesion, migration, and tube formation. In mice null for the EFNA1 receptor, EphA2, versus control animals, vascular endothelial growth factor receptor blockade and hypoxia caused more severe pulmonary hypertension, judged by elevated right ventricular systolic pressure, right ventricular hypertrophy, and loss of small arteries. CONCLUSIONS The novel relationship between BMPR2 dysfunction and reduced expression of endothelial COL4 and EFNA1 may underlie vulnerability to injury in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Christopher J Rhodes
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Hogune Im
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Aiqin Cao
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Jan K Hennigs
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Lingli Wang
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Silin Sa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Pin-I Chen
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nils P Nickel
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Kazuya Miyagawa
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rachel K Hopper
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Nancy F Tojais
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Caiyun G Li
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mingxia Gu
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Edda Spiekerkoetter
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Zhaoying Xian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Rui Chen
- 2 Cardiovascular Institute.,4 Department of Genetics, and
| | - Mingming Zhao
- 2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Mark Kaschwich
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| | - Patricia A Del Rosario
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Roham T Zamanian
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- 2 Cardiovascular Institute.,5 Department of Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Marlene Rabinovitch
- 1 Vera Moulton Wall Center for Pulmonary Vascular Diseases.,2 Cardiovascular Institute.,3 Department of Pediatrics
| |
Collapse
|
20
|
de Jesus Perez VA, Yuan K, Lyuksyutova MA, Dewey F, Orcholski ME, Shuffle EM, Mathur M, Yancy L, Rojas V, Li CG, Cao A, Alastalo TP, Khazeni N, Cimprich KA, Butte AJ, Ashley E, Zamanian RT. Whole-exome sequencing reveals TopBP1 as a novel gene in idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2014; 189:1260-72. [PMID: 24702692 DOI: 10.1164/rccm.201310-1749oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Idiopathic pulmonary arterial hypertension (IPAH) is a life-threatening disorder characterized by progressive loss of pulmonary microvessels. Although mutations in the bone morphogenetic receptor 2 (BMPR2) are found in 80% of heritable and ∼15% of patients with IPAH, their low penetrance (∼20%) suggests that other unidentified genetic modifiers are required for manifestation of the disease phenotype. Use of whole-exome sequencing (WES) has recently led to the discovery of novel susceptibility genes in heritable PAH, but whether WES can also accelerate gene discovery in IPAH remains unknown. OBJECTIVES To determine whether WES can help identify novel gene modifiers in patients with IPAH. METHODS Exome capture and sequencing was performed on genomic DNA isolated from 12 unrelated patients with IPAH lacking BMPR2 mutations. Observed genetic variants were prioritized according to their pathogenic potential using ANNOVAR. MEASUREMENTS AND MAIN RESULTS A total of nine genes were identified as high-priority candidates. Our top hit was topoisomerase DNA binding II binding protein 1 (TopBP1), a gene involved in the response to DNA damage and replication stress. We found that TopBP1 expression was reduced in vascular lesions and pulmonary endothelial cells isolated from patients with IPAH. Although TopBP1 deficiency made endothelial cells susceptible to DNA damage and apoptosis in response to hydroxyurea, its restoration resulted in less DNA damage and improved cell survival. CONCLUSIONS WES led to the discovery of TopBP1, a gene whose deficiency may increase susceptibility to small vessel loss in IPAH. We predict that use of WES will help identify gene modifiers that influence an individual's risk of developing IPAH.
Collapse
|
21
|
de Jesus Perez V, Yuan K, Alastalo TP, Spiekerkoetter E, Rabinovitch M. Targeting the Wnt signaling pathways in pulmonary arterial hypertension. Drug Discov Today 2014; 19:1270-6. [PMID: 24955837 DOI: 10.1016/j.drudis.2014.06.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/16/2014] [Indexed: 10/25/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disorder that is associated with elevated pulmonary pressures and right heart failure resulting from progressive loss and thickening of small pulmonary arteries. Despite their ability to improve symptoms, current therapies fail to prevent disease progression, leaving lung transplantation as the only therapy in end-stage PAH. To overcome the limitations of current therapies, there is an active search for disease-modifying agents capable of altering the natural history of, and improving clinical outcomes in, PAH. The Wnt signaling pathways have emerged as attractive treatment targets in PAH given their role in the preservation of pulmonary vascular homeostasis and the recent development of Wnt-specific compounds and biological therapies capable of modulating pathway activity. In this review, we summarize the literature describing the role of Wnt signaling in the pulmonary circulation and discuss promising advances in the field of Wnt therapeutics that could lead to novel clinical therapies capable of preventing and/or reversing pulmonary vascular pathology in patients with this devastating disease.
Collapse
Affiliation(s)
- Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA.
| | - Ke Yuan
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| | - Tero-Pekka Alastalo
- Children's Hospital Helsinki, Tukholmankatu 8, FI-00290 Helsinki, Finland; Biomedicum Helsinski, Tukholmankatu 8, FI-00290 Helsinki, Finland Finland
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Pediatric Cardiology, Stanford University Medical Center, 300 Pasteur Drive Grant S140B, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Koyama M, Furuhashi M, Ishimura S, Mita T, Fuseya T, Okazaki Y, Yoshida H, Tsuchihashi K, Miura T. Reduction of endoplasmic reticulum stress by 4-phenylbutyric acid prevents the development of hypoxia-induced pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2014; 306:H1314-23. [PMID: 24610918 DOI: 10.1152/ajpheart.00869.2013] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by vasoconstriction and vascular remodeling of the pulmonary artery (PA). Recently, endoplasmic reticulum (ER) stress and inappropriate adaptation through the unfolded protein response (UPR) have been disclosed in various types of diseases. Here we examined whether ER stress is involved in the pathogenesis of PAH. Four weeks of chronic normobaric hypoxia increased right ventricular (RV) systolic pressure by 63% compared with that in normoxic controls and induced RV hypertrophy and medial thickening of the PA in C57BL/6J mice. Treatment with 4-phenylbutyric acid (4-PBA), a chemical chaperone, significantly reduced RV systolic pressure by 30%, attenuated RV hypertrophy and PA muscularization, and increased total running distance in a treadmill test by 70% in hypoxic mice. The beneficial effects of 4-PBA were associated with suppressed expression of inflammatory cytokines and ER stress markers, including Grp78 and Grp94 in the activating transcription factor-6 branch, sXbp1 and Pdi in the inositol-requiring enzyme-1 branch and Atf4 in the PKR-like ER kinase branch, and reduced phosphorylation of c-Jun NH2-terminal kinase and eukaryotic translation initiation factor-2α in the lung. The pattern of changes in ER stress and inflammatory markers by 4-PBA in the lung of the PAH model was reproduced in PA smooth muscle cells by chronic stimulation of platelet-derived growth factor-BB or hypoxia. Furthermore, knockdown of each UPR branch sensor activated other branches and promoted proliferation of PA smooth muscle cells. The findings indicate that activation of all branches of the UPR and accompanying inflammation play a major role in the pathogenesis of PAH, and that chemical chaperones are potentially therapeutic agents for PAH.
Collapse
Affiliation(s)
- Masayuki Koyama
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan; and
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Enhanced assay of endothelial exocytosis using extracellular matrix components. Anal Biochem 2014; 452:19-24. [PMID: 24561025 DOI: 10.1016/j.ab.2014.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 01/05/2023]
Abstract
Vascular inflammation plays a key role in the pathogenesis of atherosclerosis. The first step in vascular inflammation is endothelial exocytosis, in which endothelial granules fuse with the plasma membrane, releasing prothrombotic and proinflammatory messenger molecules. The development of cell culture models to study endothelial exocytosis has been challenging because the factors that modulate exocytosis in vitro are not well understood. Here we report a method for studying endothelial exocytosis that optimizes extracellular matrix components, cell density, and duration of culture. Human umbilical vein endothelial cells plated on collagen I-coated plates and cultured in the confluent state for 7-12 days in low-serum medium showed robust secretion of von Willebrand factor when stimulated with various agonists. This exocytosis assay is rapid and applicable to high-throughput screening.
Collapse
|
24
|
|
25
|
|