1
|
Liu Z, Bai Z, Chen X, Chen Y, Chen Z, Wang L, He Y, Guo Y. Advances and applications of biosensors in pulmonary hypertension. Respir Res 2025; 26:147. [PMID: 40234824 PMCID: PMC11998464 DOI: 10.1186/s12931-025-03221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/05/2025] [Indexed: 04/17/2025] Open
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by elevated pulmonary artery pressure, with its prevalence and incidence continuously increasing, posing a threat to the lives of many patients worldwide. Due to the complex etiology of PH and the lack of specificity in clinical manifestations, there is currently a lack of effective and specific methods for early diagnosis in clinical practice. Biosensors hold significant promise for the early detection, therapeutic monitoring, prognostic evaluation, and personalized treatment of PH, owing to their rapid, sensitive, and highly selective characteristics. The rapid development of various types of biosensors, such as electrochemical biosensors, optical biosensors, microfluidic biosensors, and wireless biosensors, combined with the use of nanomaterials, makes the rapid and accurate detection of PH-related biomarkers possible. Despite the broad application prospects of biosensors in the field of PH, challenges remain in terms of sensitivity, selectivity, stability, and regulation. This article reviews the main pathophysiological mechanisms and commonly used biomarkers of PH, the types and principles of biosensors, and summarizes the progress of biosensors in PH research as well as the current challenges, in order to promote further in-depth research and the development of biosensor technology, thereby improving the diagnosis and treatment effects of PH. Clinical trial number: Not applicable.
Collapse
Affiliation(s)
- Zhi Liu
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Zhuojun Bai
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Xiang Chen
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China
| | - Yajie Chen
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhu Chen
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Li Wang
- Department of Laboratory, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| | - Yi He
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| | - Yuan Guo
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Department of Cardiovascular Medicine, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116 South Changjiang Road, Zhuzhou, 412007, Hunan, China.
| |
Collapse
|
2
|
Jacquet J, Marcos E, Lipskaia L, Gros V, Born E, Houssaini A, Adnot S, Boyer L. [Senescence of the pulmonary endothelial cells: VEGF, a new target in pulmonary pathologies and aging]. Rev Mal Respir 2025; 42:134-137. [PMID: 40023716 DOI: 10.1016/j.rmr.2025.02.008] [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] [Indexed: 03/04/2025]
Abstract
Vascular aging leading to microvessel depletion is a key element of organismal aging. The proposed mechanism is a deficiency of vascular endothelial growth factor (VEGF) signaling in the endothelial cells (EC), linked to the increase of a receptor in a soluble form (sVEGFR1) preventing VEGF from binding to its active receptor (VEGFR2). Without the VEGF survival signal, ECs may become senescent, contributing to aging and to various pulmonary pathologies. Deficiency of VEGF signaling in EC senescence could represent a determining element of lung aging and diseases such as pulmonary hypertension (PH) and emphysema.
Collapse
Affiliation(s)
| | - E Marcos
- IMRB, Inserm U955, Créteil, France
| | | | - V Gros
- IMRB, Inserm U955, Créteil, France
| | - E Born
- IMRB, Inserm U955, Créteil, France
| | | | - S Adnot
- IMRB, Inserm U955, Créteil, France; Institute for Lung Health, Justus Liebig University, Giessen, Allemagne
| | - L Boyer
- IMRB, Inserm U955, Créteil, France
| |
Collapse
|
3
|
Lammi MR, Kolstad KD, Saketkoo LA, Khatri A, Utz PJ, Steen VD, Chung L. Endothelial Biomarkers of Systemic Sclerosis-Associated Pulmonary Hypertension. Arthritis Care Res (Hoboken) 2025; 77:266-272. [PMID: 37365746 DOI: 10.1002/acr.25180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/23/2023] [Accepted: 06/22/2023] [Indexed: 06/28/2023]
Abstract
OBJECTIVE Despite efforts at early detection, patients with systemic sclerosis (SSc) pulmonary hypertension (PH) present with advanced disease. We sought to determine whether endothelial biomarkers (asymmetric dimethylarginine [ADMA], soluble endoglin [sEng], and pentraxin-3 [PTX-3]) can determine SSc-PH risk or differentiate between SSc-PH subgroups. METHODS ADMA, sEng, and PTX-3 were measured by enzyme-linked immunosorbent assay in four groups: 1) 18 healthy controls, 2) 74 patients with SSc-PH, 3) 44 patients at high risk for PH features, and 4) 10 patients with low risk for PH features. High-risk features included a diffusion capacity (DLco) less than 55% with a forced vital capacity (FVC) greater than 70%, an FVC/DLco ratio of >1.6, or a right ventricular systolic pressure on an echocardiogram greater than or equal to 40 mm Hg. ADMA, sEng, and PTX-3 were compared between these four groups as well as stratified based on the three SSc-PH clinical classification groups (pulmonary arterial hypertension [PAH], left-heart disease, and interstitial lung disease [ILD]). RESULTS PTX-3 was significantly lower in subjects with SSc at low risk for PH (median 27.0 pg/ml [interquartile range (IQR) 19.0-47.3]; P < 0.003) than the other groups. The area under the receiver operating characteristic curve was 0.87 (95% confidence interval 0.76-0.98, P = 0.0002) to differentiate low risk from high risk for patients with PH. PTX-3 was significantly lower in SSc-PH from disease of the left side of the heart (57.5 pg/ml [IQR 39.8-79.0]; P < 0.01) compared to SSc-PH from either PAH (85.5 pg/ml [IQR 56.3-104.5]) or ILD (90.3 pg/ml [IQR 74.9-111.0]). Neither ADMA nor sEng differed between the four groups. CONCLUSION PTX-3 is a promising biomarker of PH risk status in patients with SSc as well as a possible marker of precapillary PH, which should be validated in an external cohort.
Collapse
Affiliation(s)
- Matthew R Lammi
- Louisiana State University Health Sciences Center, University Medical Center-New Orleans, and New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, Louisiana
| | | | - Lesley Ann Saketkoo
- University Medical Center-New Orleans, New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, and Tulane University School of Medicine, New Orleans, Louisiana
| | - Avani Khatri
- Stanford University School of Medicine, Palo Alto, California
| | - Paul J Utz
- Stanford University School of Medicine, Palo Alto, California
| | | | - Lorinda Chung
- Stanford University School of Medicine and Palo Alto VA Health Care System, Palo Alto, California
| |
Collapse
|
4
|
Blaha V, Rathouska JU, Langrova H, Blaha M, Studnicka J, Andrys C, Loefflerova V, Lanska M, Vejrazkova E, Nachtigal P, Stepanov A. Soluble endoglin as a biomarker of successful rheopheresis treatment in patients with age-related macular degeneration. Sci Rep 2024; 14:28902. [PMID: 39572693 PMCID: PMC11582623 DOI: 10.1038/s41598-024-80375-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
Age-related macular degeneration (AMD) is a progressive chronic disease causing visual impairment or central vision loss in the elderly. We hypothesized that successful rheopheresis would be associated with positive changes in soluble endoglin (sENG), PSCK9, alpha-2-macroglobulin (A2M), and hs-CRP levels. 31 elderly patients with the dry form of AMD, treated with rheopheresis with a follow-up period of at least 5 years and an average age of 68 ± 4 years, were evaluated. Each treated patient received a series of 8 procedures in 10 weeks and, after the 2-year period, another 2 procedures within 1 week. Then, the patients were followed up every 6 months and divided into the successfully treated and therapeutic failure group according to best-corrected visual acuity (BCVA), size of the drusen area, and the drusenoid pigment epithelium detachment (DPED). Based on the ophthalmological assessment, rheopheresis treatment was successful in 73% of AMD patients. The therapy was associated with a significant decrease in total cholesterol, LDL-C, HDL-C, apoprotein B, lipoprotein (a) levels, and rheologically important parameters, irrespective of the therapy's success or failure. The success of rheopheresis therapy was exclusively related to a significant decrease in sENG and A2M levels. Over the long term, rheopheresis prevented the decline of BCVA, reduced the DPED and area of macular drusen, and improved the preservation of an intact photoreceptor ellipsoid zone in most patients. Moreover, we showed for the first time that sENG and A2M could be potentially sensitive biomarkers of successful rheopheresis procedure, irrespective of lipid parameters changes.
Collapse
Affiliation(s)
- Vladimir Blaha
- Faculty of Medicine in Hradec Kralove, 3rd Department of Internal Medicine - Metabolism and Gerontology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Jana Urbankova Rathouska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05, Hradec Kralove, Czech Republic
| | - Hana Langrova
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Milan Blaha
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Jan Studnicka
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Ctirad Andrys
- Department of Immunology and Allergology, Faculty of Medicine in Hradec Kralove, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | | | - Miriam Lanska
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Eva Vejrazkova
- Faculty of Medicine in Hradec Kralove, 4th Department of Internal Medicine - Hematology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05, Hradec Kralove, Czech Republic.
| | - Alexandr Stepanov
- Faculty of Medicine in Hradec Kralove, Department of Ophthalmology, University Hospital Hradec Kralove and Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
5
|
Klein F, Dinesh S, Fiedler D, Grün K, Schrepper A, Bogoviku J, Bäz L, Pfeil A, Kretzschmar D, Schulze PC, Möbius-Winkler S, Franz M. Identification of Serum Interleukin-22 as Novel Biomarker in Pulmonary Hypertension: A Translational Study. Int J Mol Sci 2024; 25:3985. [PMID: 38612795 PMCID: PMC11012889 DOI: 10.3390/ijms25073985] [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: 03/03/2024] [Revised: 03/30/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Growing evidence suggests the crucial involvement of inflammation in the pathogenesis of pulmonary hypertension (PH). The current study analyzed the expression of interleukin (IL)-17a and IL-22 as potential biomarkers for PH in a preclinical rat model of PH as well as the serum levels in a PH patient collective. PH was induced by monocrotalin (60 mg/kg body weight s.c.) in 10 Sprague Dawley rats (PH) and compared to 6 sham-treated controls (CON) as well as 10 monocrotalin-induced, macitentan-treated rats (PH_MAC). Lung and cardiac tissues were subjected to histological and immunohistochemical analysis for the ILs, and their serum levels were quantified using ELISA. Serum IL levels were also measured in a PH patient cohort. IL-22 expression was significantly increased in the lungs of the PH and PH_MAC groups (p = 0.002), whereas increased IL17a expression was demonstrated only in the lungs and RV of the PH (p < 0.05) but not the PH_MAC group (p = n.s.). The PH group showed elevated serum concentrations for IL-22 (p = 0.04) and IL-17a (p = 0.008). Compared to the PH group, the PH_MAC group demonstrated a decrease in IL-22 (p = 0.021) but not IL17a (p = n.s.). In the PH patient collective (n = 92), increased serum levels of IL-22 but not IL-17a could be shown (p < 0.0001). This elevation remained significant across the different etiological groups (p < 0.05). Correlation analysis revealed multiple significant relations between IL-22 and various clinical, laboratory, functional and hemodynamic parameters. IL-22 could serve as a promising inflammatory biomarker of PH with potential value for initial diagnosis, functional classification or even prognosis estimation. Its validation in larger patients' cohorts regarding outcome and survival data, as well as the probability of promising therapeutic target structures, remains the object of further studies.
Collapse
Affiliation(s)
- Friederike Klein
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Sandesh Dinesh
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Desiree Fiedler
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Katja Grün
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Jürgen Bogoviku
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Laura Bäz
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Alexander Pfeil
- Department of Internal Medicine III, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Daniel Kretzschmar
- Herz-und Gefäßmedizin Goslar (HUGG), Goslar, Fleischscharren 4, 38640 Goslar, Germany
| | - P. Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Sven Möbius-Winkler
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
| | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (F.K.)
- Department of Cardiology, Angiology and Intensive Care Medicine, Cardiovascular Center Rotenburg Klinikum Hersfeld-Rotenburg, Heinz-Meise-Straße 100, 36199 Rotenburg an der Fulda, Germany
| |
Collapse
|
6
|
Favoino E, Prete M, Liakouli V, Leone P, Sisto A, Navarini L, Vomero M, Ciccia F, Ruscitti P, Racanelli V, Giacomelli R, Perosa F. Idiopathic and connective tissue disease-associated pulmonary arterial hypertension (PAH): Similarities, differences and the role of autoimmunity. Autoimmun Rev 2024; 23:103514. [PMID: 38181859 DOI: 10.1016/j.autrev.2024.103514] [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: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
Pre-capillary pulmonary arterial hypertension (PAH) is hemodynamically characterized by a mean pulmonary arterial pressure (mPAP) ≥ 20 mmHg, pulmonary capillary wedge pressure (PAWP) ≤15 mmHg and pulmonary vascular resistance (PVR) > 2. PAH is classified in six clinical subgroups, including idiopathic PAH (IPAH) and PAH associated to connective tissue diseases (CTD-PAH), that will be the main object of this review. The aim is to compare these two PAH subgroups in terms of epidemiology, histological and pathogenic findings in an attempt to define disease-specific features, including autoimmunity, that may explain the heterogeneity of response to therapy between IPAH and CTD-PAH.
Collapse
Affiliation(s)
- Elvira Favoino
- Laboratory of Cellular and Molecular Immunology, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy.
| | - Marcella Prete
- Internal Medicine Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Vasiliki Liakouli
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Patrizia Leone
- Internal Medicine Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Adriana Sisto
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy
| | - Luca Navarini
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Marta Vomero
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Francesco Ciccia
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vito Racanelli
- Centre for Medical Sciences, University of Trento and Internal Medicine Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| | - Roberto Giacomelli
- Clinical and research section of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Via Álvaro del Portillo 200, 00128, Rome, Italy; Rheumatology and Clinical Immunology, Department of Medicine, University of Rome "Campus Biomedico", School of Medicine, Rome, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, University of Bari Medical School, Bari, Italy.
| |
Collapse
|
7
|
Welch CL, Aldred MA, Balachandar S, Dooijes D, Eichstaedt CA, Gräf S, Houweling AC, Machado RD, Pandya D, Prapa M, Shaukat M, Southgate L, Tenorio-Castano J, Chung WK. Defining the clinical validity of genes reported to cause pulmonary arterial hypertension. Genet Med 2023; 25:100925. [PMID: 37422716 PMCID: PMC10766870 DOI: 10.1016/j.gim.2023.100925] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
PURPOSE Pulmonary arterial hypertension (PAH) is a rare, progressive vasculopathy with significant cardiopulmonary morbidity and mortality. Genetic testing is currently recommended for adults diagnosed with heritable, idiopathic, anorexigen-, hereditary hemorrhagic telangiectasia-, and congenital heart disease-associated PAH, PAH with overt features of venous/capillary involvement, and all children diagnosed with PAH. Variants in at least 27 genes have putative evidence for PAH causality. Rigorous assessment of the evidence is needed to inform genetic testing. METHODS An international panel of experts in PAH applied a semi-quantitative scoring system developed by the NIH Clinical Genome Resource to classify the relative strength of evidence supporting PAH gene-disease relationships based on genetic and experimental evidence. RESULTS Twelve genes (BMPR2, ACVRL1, ATP13A3, CAV1, EIF2AK4, ENG, GDF2, KCNK3, KDR, SMAD9, SOX17, and TBX4) were classified as having definitive evidence and 3 genes (ABCC8, GGCX, and TET2) with moderate evidence. Six genes (AQP1, BMP10, FBLN2, KLF2, KLK1, and PDGFD) were classified as having limited evidence for causal effects of variants. TOPBP1 was classified as having no known PAH relationship. Five genes (BMPR1A, BMPR1B, NOTCH3, SMAD1, and SMAD4) were disputed because of a paucity of genetic evidence over time. CONCLUSION We recommend that genetic testing includes all genes with definitive evidence and that caution be taken in the interpretation of variants identified in genes with moderate or limited evidence. Genes with no known evidence for PAH or disputed genes should not be included in genetic testing.
Collapse
Affiliation(s)
- Carrie L Welch
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Micheala A Aldred
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Srimmitha Balachandar
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine, IN
| | - Dennis Dooijes
- Department of Genetics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Christina A Eichstaedt
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Stefan Gräf
- NIHR BioResource for Translational Research - Rare Diseases, Department of Haemotology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Arjan C Houweling
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Rajiv D Machado
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Divya Pandya
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Matina Prapa
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, United Kingdom; St. George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Memoona Shaukat
- Center for Pulmonary Hypertension, Thoraxklinik-Heidelberg gGmbH, at Heidelberg University Hospital and Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany; Laboratory for Molecular Genetic Diagnostics, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Laura Southgate
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Jair Tenorio-Castano
- Institute of Medical and Molecular Genetics (INGEMM), Hospital Universitario La Paz, IDiPAZ, Universidad Autonoma de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain; ITHACA, European Reference Network, Brussels, Belgium
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY; Department of Medicine, Columbia University Irving Medical Center, New York, NY.
| |
Collapse
|
8
|
Graham BB, Hilton JF, Lee MH, Kumar R, Balladares DF, Rahaghi FN, Estépar RSJ, Mickael C, Lima RLB, Loureiro CM, Lucena J, Oliveira RK, Corrêa RDA. Is pulmonary arterial hypertension associated with schistosomiasis distinct from pulmonary arterial hypertension associated with portal hypertension? JHLT OPEN 2023; 1:100007. [PMID: 38050478 PMCID: PMC10695267 DOI: 10.1016/j.jhlto.2023.100007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Pulmonary arterial hypertension associated with schistosomiasis (SchPAH) and pulmonary arterial hypertension associated with portal hypertension (PoPAH) are lung diseases that develop in the presence of liver diseases. However, mechanistic pathways by which the underlying liver conditions and other drivers contribute to the development and progression of pulmonary arterial hypertension (PAH) are unclear for both etiologies. In turn, these unknowns limit certainty of strategies to prevent, diagnose, and reverse the resultant PAH. Here we consider specific mechanisms that contribute to SchPAH and PoPAH, identifying those that may be shared and those that appear to be unique to each etiology, in the hope that this exploration will both highlight known causal drivers and identify knowledge gaps appropriate for future research. Overall, the key pathophysiologic differences that we identify between SchPAH and PoPAH suggest that they are not variants of a single condition.
Collapse
Affiliation(s)
- Brian B. Graham
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Joan F. Hilton
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California
| | - Michael H. Lee
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Rahul Kumar
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Dara Fonseca Balladares
- Lung Biology Center, University of California San Francisco, San Francisco, California
- Pulmonary Division, San Francisco General Hospital, San Francisco, California
| | - Farbod N. Rahaghi
- Pulmonary Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Claudia Mickael
- Pulmonary and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | - Juliana Lucena
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Rudolf K.F. Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo de Amorim Corrêa
- Internal Medicine/Pulmonary Division, Medical School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
9
|
Zhong (钟颖) Y, Yu (游博群) PB. Angiogenesis Redux: An Overall Protective Role of VEGF/KDR Signaling in the Microvasculature in Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2023; 43:1784-1787. [PMID: 37675636 PMCID: PMC10803133 DOI: 10.1161/atvbaha.123.319839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Affiliation(s)
- Ying Zhong (钟颖)
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul B. Yu (游博群)
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Dimitry MO, Soliman YMA, ElKorashy RI, Raslan HM, Kamel SA, Hassan EM, Ahmed FE, Yousef RN, Awadallah EA. Role of micro-RNAs 21, 124 and other novel biomarkers in distinguishing between group 1 WHO pulmonary hypertension and group 2, 3 WHO pulmonary hypertension. Egypt Heart J 2023; 75:76. [PMID: 37646902 PMCID: PMC10468479 DOI: 10.1186/s43044-023-00395-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/29/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Pulmonary hypertension "PH" is considered a serious cardiovascular disease. World Health Organization divided PH into groups depending on many factors like pathological, hemodynamic, and clinical pictures. Lately, various micro-RNAs "miRNAs" and other novel biomarkers like endoglin and asymmetric dimethylarginine "ADMA" might have a role in diagnosis of PH and may differentiate between pulmonary arterial hypertension "PAH" and non-PAH. The purpose of the study is to show the role of miR-21, miR-124, endoglin and ADMA in the diagnosis of PH and distinguishing between WHO group 1 PH and WHO group 2 and 3 PH and to identify patients who might benefit from non-invasive and inexpensive tools to diagnose PAH. RESULTS miR-21 was upregulated in group 1 PH, and there was significant difference between group 1 PH as compared with group 2 PH, group 3 PH and control; miR-124 was down-regulated in group 1 PH with highly significant difference between group 1 and group 2 PH and control but no significant difference with group 3 PH, endoglin was elevated in group 1 PH with a significant difference as compared to group 2 PH, group 3 PH and control. ADMA was elevated in group 1 PH as compared to control; however, there was no significant difference between it and group 2, 3 PH. CONCLUSIONS miR-21, miR-124, endoglin and ADMA are good biomarkers to diagnose PH; however, only miR-21 and endoglin could distinguish group 1 PH from group 2 and 3 PH.
Collapse
Affiliation(s)
- Mark O Dimitry
- Cardiology Unit, Department of Internal Medicine, National Research Center, Cairo, Egypt.
| | - Youssef M A Soliman
- Pulmonary Vascular Disease Unit, Department of Pulmonology, Cairo University, Cairo, Egypt
| | - Reem I ElKorashy
- Pulmonary Vascular Disease Unit, Department of Pulmonology, Cairo University, Cairo, Egypt
| | - Hala M Raslan
- Cardiology Unit, Department of Internal Medicine, National Research Center, Cairo, Egypt
| | - Solaf A Kamel
- Department of Clinical and Chemical Pathology, National Research Center, Cairo, Egypt
| | - Eman M Hassan
- Department of Clinical and Chemical Pathology, National Research Center, Cairo, Egypt
| | - Fatma Elzahraa Ahmed
- Pulmonary Vascular Disease Unit, Department of Pulmonology, Cairo University, Cairo, Egypt
| | - Rasha N Yousef
- Department of Clinical and Chemical Pathology, National Research Center, Cairo, Egypt
| | - Eman A Awadallah
- Department of Clinical and Chemical Pathology, National Research Center, Cairo, Egypt
| |
Collapse
|
11
|
Jose A, Elwing JM, Kawut SM, Pauciulo MW, Sherman KE, Nichols WC, Fallon MB, McCormack FX. Human liver single nuclear RNA sequencing implicates BMPR2, GDF15, arginine, and estrogen in portopulmonary hypertension. Commun Biol 2023; 6:826. [PMID: 37558836 PMCID: PMC10412637 DOI: 10.1038/s42003-023-05193-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Portopulmonary hypertension (PoPH) is a type of pulmonary vascular disease due to portal hypertension that exhibits high morbidity and mortality. The mechanisms driving disease are unknown, and transcriptional characteristics unique to the PoPH liver remain unexplored. Here, we apply single nuclear RNA sequencing to compare cirrhotic livers from patients with and without PoPH. We identify characteristics unique to PoPH in cells surrounding the central hepatic vein, including increased growth differentiation factor signaling, enrichment of the arginine biosynthesis pathway, and differential expression of the bone morphogenic protein type II receptor and estrogen receptor type I genes. These results provide insight into the transcriptomic characteristics of the PoPH liver and mechanisms by which PoPH cellular dysfunction might contribute to pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Arun Jose
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Jean M Elwing
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kenneth E Sherman
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Francis X McCormack
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
12
|
Andersson-Rusch C, Liu B, Quist-Løkken I, Upton PD, Olsen OE, Hella H, Yang X, Tong Z, Morrell NW, Holien T, Li W. High concentrations of soluble endoglin can inhibit BMP9 signaling in non-endothelial cells. Sci Rep 2023; 13:6639. [PMID: 37095146 PMCID: PMC10126157 DOI: 10.1038/s41598-023-33352-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Endoglin (ENG) is a single-pass transmembrane protein highly expressed on vascular endothelial cells, although low expression levels can be detected in many other cell types. Its extracellular domain can be found in circulation known as soluble endoglin (sENG). Levels of sENG are elevated in many pathological conditions, in particular preeclampsia. We have shown that while loss of cell surface ENG decreases BMP9 signaling in endothelial cells, knocking down ENG in blood cancer cells enhances BMP9 signaling. Despite sENG binding to BMP9 with high affinity and blocking the type II receptor binding site on BMP9, sENG did not inhibit BMP9 signaling in vascular endothelial cells, but the dimeric form of sENG inhibited BMP9 signaling in blood cancer cells. Here we report that in non-endothelial cells such as human multiple myeloma cell lines and the mouse myoblast cell line C2C12, both monomeric and dimeric forms of sENG inhibit BMP9 signaling when present at high concentrations. Such inhibition can be alleviated by the overexpression of ENG and ACVRL1 (encoding ALK1) in the non-endothelial cells. Our findings suggest that the effects of sENG on BMP9 signaling is cell-type specific. This is an important consideration when developing therapies targeting the ENG and ALK1 pathway.
Collapse
Affiliation(s)
- Clara Andersson-Rusch
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway
| | - Bin Liu
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Ingrid Quist-Løkken
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Paul D Upton
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Oddrun Elise Olsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Hanne Hella
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Xudong Yang
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Zhen Tong
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Nicholas W Morrell
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Toril Holien
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway.
- Department of Hematology, St. Olav's University Hospital, Trondheim, Norway.
- Department of Biomedical Laboratory Science, NTNU, Trondheim, Norway.
- Department of Immunology and Transfusion Medicine, St. Olav's University Hospital, Trondheim, Norway.
| | - Wei Li
- Department of Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
13
|
Hirsch K, Nolley S, Ralph DD, Zheng Y, Altemeier WA, Rhodes CJ, Morrell NW, Wilkins MR, Leary PJ, Rayner SG. Circulating markers of inflammation and angiogenesis and clinical outcomes across subtypes of pulmonary arterial hypertension. J Heart Lung Transplant 2023; 42:173-182. [PMID: 36470771 PMCID: PMC9840657 DOI: 10.1016/j.healun.2022.10.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 10/09/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Subtypes of pulmonary arterial hypertension (PAH) differ in both fundamental disease features and clinical outcomes. Angiogenesis and inflammation represent disease features that may differ across subtypes and are of special interest in connective tissue disease-associated PAH (CTD-PAH). We compared inflammatory and angiogenic biomarker profiles across different etiologies of PAH and related them to clinical outcomes. METHODS Participants with idiopathic PAH, CTD-PAH, toxin-associated PAH (tox-PAH), or congenital heart disease-associated PAH (CHD-PAH) were enrolled into a prospective observational cohort. Baseline serum concentrations of 33 biomarkers were related to 3-year mortality, echocardiogram, REVEAL score, and 6-minute walk distance (6MWD). Findings were validated using plasma proteomic data from the UK PAH Cohort Study. RESULTS One hundred twelve patients were enrolled: 45 idiopathic, 27 CTD-PAH, 20 tox-PAH, and 20 CHD-PAH. Angiogenic and inflammatory biomarkers were distinctly elevated within the CTD-PAH cohort. Six biomarkers were associated with mortality within the entire PAH cohort: interleukin-6 (IL-6, HR:1.6, 95% CI:1.18-2.18), soluble fms-like tyrosine kinase 1 (sFlt-1, HR:1.35, 95% CI:1.02-1.80), placental growth factor (PlGF, HR:1.55, 95% CI:1.07-2.25), interferon gamma-induced protein 10 (IP-10, HR:1.44, 95% CI:1.04-1.99), tumor necrosis factor-beta (TNF-β, HR:1.81, 95% CI:1.11-2.95), and NT-proBNP (HR:2.19, 95% CI:1.52-3.14). Only IL-6 and NT-proBNP remained significant after controlling for multiple comparisons. IL-6, IP-10, and sFlt-1 significantly associated with mortality in CTD-PAH, but not non-CTD-PAH subgroups. In the UK cohort, IP-10, PlGF, TNF-β, and NT-proBNP significantly associated with 5-year survival. CONCLUSION Levels of angiogenic and inflammatory biomarkers are elevated in CTD-PAH, compared with other etiologies of PAH, and may correlate with clinical outcomes including mortality.
Collapse
Affiliation(s)
- Kellen Hirsch
- Department of Medicine, University of Washington, Seattle, Washington
| | - Stephanie Nolley
- Department of Medicine, University of Washington, Seattle, Washington
| | - David D Ralph
- Department of Medicine, University of Washington, Seattle, Washington
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, Washington
| | - William A Altemeier
- Department of Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington, Seattle, Washington
| | - Christopher J Rhodes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas W Morrell
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J Leary
- Department of Medicine, University of Washington, Seattle, Washington; Department of Epidemiology, University of Washington, Seattle, Washington
| | - Samuel G Rayner
- Department of Medicine, University of Washington, Seattle, Washington; Department of Bioengineering, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington, Seattle, Washington.
| |
Collapse
|
14
|
Hojda SE, Chis IC, Clichici S. Biomarkers in Pulmonary Arterial Hypertension. Diagnostics (Basel) 2022; 12:diagnostics12123033. [PMID: 36553040 PMCID: PMC9776459 DOI: 10.3390/diagnostics12123033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe medical condition characterized by elevated pulmonary vascular resistance (PVR), right ventricular (RV) failure, and death in the absence of appropriate treatment. The progression and prognosis are strictly related to the etiology, biochemical parameters, and treatment response. The gold-standard test remains right-sided heart catheterization, but dynamic monitoring of systolic pressure in the pulmonary artery is performed using echocardiography. However, simple and easily accessible non-invasive assays are also required in order to monitor this pathology. In addition, research in this area is in continuous development. In recent years, more and more biomarkers have been studied and included in clinical guidelines. These biomarkers can be categorized based on their associations with inflammation, endothelial cell dysfunction, cardiac fibrosis, oxidative stress, and metabolic disorders. Moreover, biomarkers can be easily detected in blood and urine and correlated with disease severity, playing an important role in diagnosis, prognosis, and disease progression.
Collapse
|
15
|
Santos-Gomes J, Gandra I, Adão R, Perros F, Brás-Silva C. An Overview of Circulating Pulmonary Arterial Hypertension Biomarkers. Front Cardiovasc Med 2022; 9:924873. [PMID: 35911521 PMCID: PMC9333554 DOI: 10.3389/fcvm.2022.924873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), also known as Group 1 Pulmonary Hypertension (PH), is a PH subset characterized by pulmonary vascular remodeling and pulmonary arterial obstruction. PAH has an estimated incidence of 15-50 people per million in the United States and Europe, and is associated with high mortality and morbidity, with patients' survival time after diagnosis being only 2.8 years. According to current guidelines, right heart catheterization is the gold standard for diagnostic and prognostic evaluation of PAH patients. However, this technique is highly invasive, so it is not used in routine clinical practice or patient follow-up. Thereby, it is essential to find new non-invasive strategies for evaluating disease progression. Biomarkers can be an effective solution for determining PAH patient prognosis and response to therapy, and aiding in diagnostic efforts, so long as their detection is non-invasive, easy, and objective. This review aims to clarify and describe some of the potential new candidates as circulating biomarkers of PAH.
Collapse
Affiliation(s)
- Joana Santos-Gomes
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Inês Gandra
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Rui Adão
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Frédéric Perros
- Paris-Porto Pulmonary Hypertension Collaborative Laboratory (3PH), UMR_S 999, INSERM, Université Paris-Saclay, Paris, France
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Carmen Brás-Silva
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| |
Collapse
|
16
|
Harbaum L, Rhodes CJ, Wharton J, Lawrie A, Karnes JH, Desai AA, Nichols WC, Humbert M, Montani D, Girerd B, Sitbon O, Boehm M, Novoyatleva T, Schermuly RT, Ghofrani HA, Toshner M, Kiely DG, Howard LS, Swietlik EM, Gräf S, Pietzner M, Morrell NW, Wilkins MR. Mining the Plasma Proteome for Insights into the Molecular Pathology of Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2022; 205:1449-1460. [PMID: 35394406 PMCID: PMC9875902 DOI: 10.1164/rccm.202109-2106oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 04/07/2022] [Indexed: 01/29/2023] Open
Abstract
Rationale: Pulmonary arterial hypertension (PAH) is characterized by structural remodeling of pulmonary arteries and arterioles. Underlying biological processes are likely reflected in a perturbation of circulating proteins. Objectives: To quantify and analyze the plasma proteome of patients with PAH using inherited genetic variation to inform on underlying molecular drivers. Methods: An aptamer-based assay was used to measure plasma proteins in 357 patients with idiopathic or heritable PAH, 103 healthy volunteers, and 23 relatives of patients with PAH. In discovery and replication subgroups, the plasma proteomes of PAH and healthy individuals were compared, and the relationship to transplantation-free survival in PAH was determined. To examine causal relationships to PAH, protein quantitative trait loci (pQTL) that influenced protein levels in the patient population were used as instruments for Mendelian randomization (MR) analysis. Measurements and Main Results: From 4,152 annotated plasma proteins, levels of 208 differed between patients with PAH and healthy subjects, and 49 predicted long-term survival. MR based on cis-pQTL located in proximity to the encoding gene for proteins that were prognostic and distinguished PAH from health estimated an adverse effect for higher levels of netrin-4 (odds ratio [OR], 1.55; 95% confidence interval [CI], 1.16-2.08) and a protective effect for higher levels of thrombospondin-2 (OR, 0.83; 95% CI, 0.74-0.94) on PAH. Both proteins tracked the development of PAH in previously healthy relatives and changes in thrombospondin-2 associated with pulmonary arterial pressure at disease onset. Conclusions: Integrated analysis of the plasma proteome and genome implicates two secreted matrix-binding proteins, netrin-4 and thrombospondin-2, in the pathobiology of PAH.
Collapse
Affiliation(s)
- Lars Harbaum
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Christopher J. Rhodes
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - John Wharton
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Allan Lawrie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, United Kingdom
| | - Jason H. Karnes
- Department of Pharmacy Practice and Science, University of Arizona College of Pharmacy, Tucson, Arizona
| | - Ankit A. Desai
- Department of Medical and Molecular Genetics, and Krannert Institute of Cardiology, Department of Medicine, Indiana University, Indianapolis, Indiana
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc Humbert
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - David Montani
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Barbara Girerd
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Olivier Sitbon
- Université Paris–Saclay, AP-HP, INSERM UMR_S 999, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Centre, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Mario Boehm
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tatyana Novoyatleva
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Ralph T. Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | | | - Mark Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - David G. Kiely
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Medical School, Sheffield, United Kingdom
| | - Luke S. Howard
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Emilia M. Swietlik
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Stefan Gräf
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- National Institute for Health Research BioResource for Translational Research, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Maik Pietzner
- Computational Medicine, Berlin Institute of Health (BIH) at Charité–Universitätsmedizin Berlin, Germany; and
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Martin R. Wilkins
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
17
|
Smits AJ, Botros L, Mol MA, Ziesemer KA, Wilkins MR, Vonk Noordegraaf A, Bogaard HJ, Aman J. A Systematic Review with Meta-analysis of Biomarkers for detection of Pulmonary Arterial Hypertension. ERJ Open Res 2022; 8:00009-2022. [PMID: 35651362 PMCID: PMC9149393 DOI: 10.1183/23120541.00009-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/04/2022] [Indexed: 11/20/2022] Open
Abstract
Rationale The blood is a rich source of potential biomarkers for the diagnosis of idiopathic and hereditary pulmonary arterial hypertension (iPAH and hPAH, referred to as “PAH”). While a lot of biomarkers have been identified for PAH, the clinical utility of these biomarkers often remains unclear. Here, we performed an unbiased meta-analysis of published biomarkers to identify biomarkers with the highest performance for detection of PAH. Methods A literature search (in PubMed, Embase.com, Clarivate Analytics/Web of Science Core Collection and Wiley/Cochrane Library) was performed up to 28 January 2021. Primary end points were blood biomarker levels in PAH versus asymptomatic controls or patients suspected of pulmonary hypertension (PH) with proven normal haemodynamic profiles. Results 149 articles were identified by the literature search. Meta-analysis of 26 biomarkers yielded 17 biomarkers that were differentially expressed in PAH and non-PH control subjects. Red cell distribution width, low density lipid-cholesterol, d-dimer, N-terminal prohormone of brain natriuretic protein (NT-proBNP), interleukin-6 (IL-6) and uric acid were biomarkers with the largest observed differences, largest sample sizes and a low risk of publication bias. Receiver operating characteristic curves and sensitivity/specificity analyses demonstrated that NT-proBNP had a high sensitivity, but low specificity for PAH. For the other biomarkers, insufficient data on diagnostic accuracy with receiver operating characteristic curves were available for meta-analysis. Conclusion This meta-analysis validates NT-proBNP as a biomarker with high sensitivity for PAH, albeit with low specificity. The majority of biomarkers evaluated in this meta-analysis lacked either external validation or data on diagnostic accuracy. Further validation studies are required as well as studies that test combinations of biomarkers to improve specificity. Meta-analysis of 26 biomarkers yielded 17 differentially expressed biomarkers in PAH. NT-proBNP had the highest diagnostic accuracy but had a low specificity for PAH. Other markers, including IL-6, RDW, LDL-c, D-dimer and UA, lacked clinical validation.https://bit.ly/3J4YAyC
Collapse
|
18
|
Elfsmark L, Ågren L, Akfur C, Jonasson S. Ammonia exposure by intratracheal instillation causes severe and deteriorating lung injury and vascular effects in mice. Inhal Toxicol 2022; 34:145-158. [PMID: 35452355 DOI: 10.1080/08958378.2022.2064566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Ammonia (NH3) is a corrosive alkaline gas that can cause life-threatening injuries by inhalation. The aim was to establish a disease model for NH3-induced injuries similar to acute lung injury (ALI) described in exposed humans and investigate the progression of lung damage, respiratory dysfunction and evaluate biomarkers for ALI and inflammation over time. METHODS Female BALB/c mice were exposed to an NH3 dose of 91.0 mg/kg·bw using intratracheal instillation and the pathological changes were followed for up to 7 days. RESULTS NH3 instillation resulted in the loss of body weight along with a significant increase in pro-inflammatory mediators in both bronchoalveolar lavage fluid (e.g. IL-1β, IL-6, KC, MMP-9, SP-D) and blood (e.g. IL-6, Fibrinogen, PAI-1, PF4/CXCL4, SP-D), neutrophilic lung inflammation, alveolar damage, increased peripheral airway resistance and methacholine-induced airway hyperresponsiveness compared to controls at 20 h. On day 7 after exposure, deteriorating pathological changes such as increased macrophage lung infiltration, heart weights, lung hemorrhages and coagulation abnormalities (elevated plasma levels of PAI-1, fibrinogen, endothelin and thrombomodulin) were observed but no increase in lung collagen. Some of the analyzed blood biomarkers (e.g. RAGE, IL-1β) were unaffected despite severe ALI and may not be significant for NH3-induced damages. CONCLUSIONS NH3 induces severe acute lung injuries that deteriorate over time and biomarkers in lungs and blood that are similar to those found in humans. Therefore, this model has potential use for developing diagnostic tools for NH3-induced ALI and for finding new therapeutic treatments, since no specific antidote has been identified yet.
Collapse
Affiliation(s)
- Linda Elfsmark
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | - Lina Ågren
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | - Christine Akfur
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| | - Sofia Jonasson
- Swedish Defence Research Agency, CBRN Defence and Security, Umeå, Sweden
| |
Collapse
|
19
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
20
|
Lucero García Rojas EY, Villanueva C, Bond RA. Hypoxia Inducible Factors as Central Players in the Pathogenesis and Pathophysiology of Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:709509. [PMID: 34447792 PMCID: PMC8382733 DOI: 10.3389/fcvm.2021.709509] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular (CV) diseases are the major cause of death in industrialized countries. The main function of the CV system is to deliver nutrients and oxygen to all tissues. During most CV pathologies, oxygen and nutrient delivery is decreased or completely halted. Several mechanisms, including increased oxygen transport and delivery, as well as increased blood flow are triggered to compensate for the hypoxic state. If the compensatory mechanisms fail to sufficiently correct the hypoxia, irreversible damage can occur. Thus, hypoxia plays a central role in the pathogenesis and pathophysiology of CV diseases. Hypoxia inducible factors (HIFs) orchestrate the gene transcription for hundreds of proteins involved in erythropoiesis, glucose transport, angiogenesis, glycolytic metabolism, reactive oxygen species (ROS) handling, cell proliferation and survival, among others. The overall regulation of the expression of HIF-dependent genes depends on the severity, duration, and location of hypoxia. In the present review, common CV diseases were selected to illustrate that HIFs, and proteins derived directly or indirectly from their stabilization and activation, are related to the development and perpetuation of hypoxia in these pathologies. We further classify CV diseases into acute and chronic hypoxic states to better understand the temporal relevance of HIFs in the pathogenesis, disease progression and clinical outcomes of these diseases. We conclude that HIFs and their derived factors are fundamental in the genesis and progression of CV diseases. Understanding these mechanisms will lead to more effective treatment strategies leading to reduced morbidity and mortality.
Collapse
Affiliation(s)
| | - Cleva Villanueva
- Instituto Politecnico Nacional, Escuela Superior de Medicina, Mexico City, Mexico
| | - Richard A Bond
- Department of Pharmacology and Pharmaceutical Sciences, University of Houston, Houston, TX, United States
| |
Collapse
|
21
|
Li W, Long L, Yang X, Tong Z, Southwood M, King R, Caruso P, Upton PD, Yang P, Bocobo GA, Nikolic I, Higuera A, Salmon RM, Jiang H, Lodge KM, Hoenderdos K, Baron RM, Yu PB, Condliffe AM, Summers C, Nourshargh S, Chilvers ER, Morrell NW. Circulating BMP9 Protects the Pulmonary Endothelium during Inflammation-induced Lung Injury in Mice. Am J Respir Crit Care Med 2021; 203:1419-1430. [PMID: 33320799 PMCID: PMC8456542 DOI: 10.1164/rccm.202005-1761oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: Pulmonary endothelial permeability contributes to the high-permeability pulmonary edema that characterizes acute respiratory distress syndrome. Circulating BMP9 (bone morphogenetic protein 9) is emerging as an important regulator of pulmonary vascular homeostasis. Objectives:To determine whether endogenous BMP9 plays a role in preserving pulmonary endothelial integrity and whether loss of endogenous BMP9 occurs during LPS challenge. Methods: A BMP9-neutralizing antibody was administrated to healthy adult mice, and lung vasculature was examined. Potential mechanisms were delineated by transcript analysis in human lung endothelial cells. The impact of BMP9 administration was evaluated in a murine acute lung injury model induced by inhaled LPS. Levels of BMP9 were measured in plasma from patients with sepsis and from endotoxemic mice. Measurements and Main Results: Subacute neutralization of endogenous BMP9 in mice (N = 12) resulted in increased lung vascular permeability (P = 0.022), interstitial edema (P = 0.0047), and neutrophil extravasation (P = 0.029) compared with IgG control treatment (N = 6). In pulmonary endothelial cells, BMP9 regulated transcriptome pathways implicated in vascular permeability and cell-membrane integrity. Augmentation of BMP9 signaling in mice (N = 8) prevented inhaled LPS-induced lung injury (P = 0.0027) and edema (P < 0.0001). In endotoxemic mice (N = 12), endogenous circulating BMP9 concentrations were markedly reduced, the causes of which include a transient reduction in hepatic BMP9 mRNA expression and increased elastase activity in plasma. In human patients with sepsis (N = 10), circulating concentratons of BMP9 were also markedly reduced (P < 0.0001). Conclusions: Endogenous circulating BMP9 is a pulmonary endothelial-protective factor, downregulated during inflammation. Exogenous BMP9 offers a potential therapy to prevent increased pulmonary endothelial permeability in lung injury.
Collapse
Affiliation(s)
- Wei Li
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Lu Long
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Xudong Yang
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Zhen Tong
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Mark Southwood
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ross King
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Paola Caruso
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Paul D. Upton
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Ivana Nikolic
- Cardiovascular Medicine Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Angelica Higuera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital–Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Richard M. Salmon
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - He Jiang
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Katharine M. Lodge
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Kim Hoenderdos
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca M. Baron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital–Harvard Medical School, Harvard University, Boston, Massachusetts
| | | | - Alison M. Condliffe
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Charlotte Summers
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Sussan Nourshargh
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Edwin R. Chilvers
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Nicholas W. Morrell
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
22
|
Vicen M, Igreja Sá IC, Tripská K, Vitverová B, Najmanová I, Eissazadeh S, Micuda S, Nachtigal P. Membrane and soluble endoglin role in cardiovascular and metabolic disorders related to metabolic syndrome. Cell Mol Life Sci 2021; 78:2405-2418. [PMID: 33185696 PMCID: PMC11072708 DOI: 10.1007/s00018-020-03701-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/05/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
Membrane endoglin (Eng, CD105) is a transmembrane glycoprotein essential for the proper function of vascular endothelium. It might be cleaved by matrix metalloproteinases to form soluble endoglin (sEng), which is released into the circulation. Metabolic syndrome comprises conditions/symptoms that usually coincide (endothelial dysfunction, arterial hypertension, hyperglycemia, obesity-related insulin resistance, and hypercholesterolemia), and are considered risk factors for cardiometabolic disorders such as atherosclerosis, type II diabetes mellitus, and liver disorders. The purpose of this review is to highlight current knowledge about the role of Eng and sEng in the disorders mentioned above, in vivo and in vitro extent, where we can find a wide range of contradictory results. We propose that reduced Eng expression is a hallmark of endothelial dysfunction development in chronic pathologies related to metabolic syndrome. Eng expression is also essential for leukocyte transmigration and acute inflammation, suggesting that Eng is crucial for the regulation of endothelial function during the acute phase of vascular defense reaction to harmful conditions. sEng was shown to be a circulating biomarker of preeclampsia, and we propose that it might be a biomarker of metabolic syndrome-related symptoms and pathologies, including hypercholesterolemia, hyperglycemia, arterial hypertension, and diabetes mellitus as well, despite the fact that some contradictory findings have been reported. Besides, sEng can participate in the development of endothelial dysfunction and promote the development of arterial hypertension, suggesting that high levels of sEng promote metabolic syndrome symptoms and complications. Therefore, we suggest that the treatment of metabolic syndrome should take into account the importance of Eng in the endothelial function and levels of sEng as a biomarker and risk factor of related pathologies.
Collapse
Affiliation(s)
- Matej Vicen
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Ivone Cristina Igreja Sá
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Katarína Tripská
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Barbora Vitverová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Iveta Najmanová
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Samira Eissazadeh
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic
| | - Stanislav Micuda
- Faculty of Medicine in Hradec Kralove, Department of Pharmacology, Charles University, Simkova 870, Hradec Kralove, 500 03, Czech Republic
| | - Petr Nachtigal
- Faculty of Pharmacy in Hradec Kralove, Department of Biological and Medical Sciences, Charles University, Heyrovskeho 1203, Hradec Kralove, 500 03, Czech Republic.
| |
Collapse
|
23
|
Dara A, Arvanitaki A, Theodorakopoulou M, Athanasiou C, Pagkopoulou E, Boutou A. Non-Invasive Assessment of Endothelial Dysfunction in Pulmonary Arterial Hypertension. Mediterr J Rheumatol 2021; 32:6-14. [PMID: 34386697 PMCID: PMC8314877 DOI: 10.31138/mjr.32.1.6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 01/01/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterised by an increased pressure in the pulmonary arterial circulation, resulting in the elevation of pulmonary vascular resistance. Pulmonary endothelial dysfunction and inflammation, triggered by shear stress and hypoxia, constitute the hallmarks of pulmonary vasculopathy by promoting endothelial and smooth muscle cells proliferation, vasoconstriction, and thrombosis. While research was predominantly focused on pulmonary vasculature, the investigation of peripheral endothelial damage in different vascular beds has attracted the interest over the last years. As a result, effective non-invasive methods that can assess the endothelial function and the architectural integrity have been utilized for the evaluation of pulmonary and peripheral vasculature. Non-invasive plethysmography, pulmonary flow reserve, nailfold videocapillaroscopy, near-infrared spectroscopy, and imaging techniques such as magnetic resonance angiography and perfusion imaging coupled by a number of biomarkers can be used for the assessment of peripheral vascular function in PAH individuals. In this review, we summarise and critically approach the current evidence of more systemic derangement of vascular function in PAH defined by novel, non-invasive methods employed for functional and morphological assessment of endothelium and microcirculation.
Collapse
Affiliation(s)
- Athanasia Dara
- Fourth Department of Internal Medicine, Hippokration University Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Alexandra Arvanitaki
- Fourth Department of Internal Medicine, Hippokration University Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,First Department of Cardiology, AHEPA University Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Adult Congenital Heart Centre and National Centre for Pulmonary Arterial Hypertension, Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, Imperial College, London, UK
| | | | - Christos Athanasiou
- Fourth Department of Internal Medicine, Hippokration University Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Pagkopoulou
- Fourth Department of Internal Medicine, Hippokration University Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Afroditi Boutou
- Department of Respiratory Medicine, G. Papanikolaou Hospital, Thessaloniki, Greece
| |
Collapse
|
24
|
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: 88] [Impact Index Per Article: 22.0] [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
|
25
|
Igreja Sá IC, Tripska K, Hroch M, Hyspler R, Ticha A, Lastuvkova H, Schreiberova J, Dolezelova E, Eissazadeh S, Vitverova B, Najmanova I, Vasinova M, Pericacho M, Micuda S, Nachtigal P. Soluble Endoglin as a Potential Biomarker of Nonalcoholic Steatohepatitis (NASH) Development, Participating in Aggravation of NASH-Related Changes in Mouse Liver. Int J Mol Sci 2020; 21:E9021. [PMID: 33261044 PMCID: PMC7731045 DOI: 10.3390/ijms21239021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by hepatic steatosis with inflammation and fibrosis. Membrane endoglin (Eng) expression is shown to participate in fibrosis, and plasma concentrations of soluble endoglin (sEng) are increased in patients with hypercholesterolemia and type 2 diabetes mellitus. We hypothesize that NASH increases both hepatic Eng expression and sEng in blood and that high levels of sEng modulate cholesterol and bile acid (BA) metabolism and affect NASH progression. Three-month-old transgenic male mice overexpressing human sEng and their wild type littermates are fed for six months with either a high-saturated fat, high-fructose high-cholesterol (FFC) diet or a chow diet. Evaluation of NASH, Liquid chromatography-mass spectrometry (LC/MS) analysis of BA, hepatic expression of Eng, inflammation, fibrosis markers, enzymes and transporters involved in hepatic cholesterol and BA metabolism are assessed using Real-Time Quantitative Reverse Transcription Polymerase Chain reaction (qRT-PCR) and Western blot. The FFC diet significantly increases mouse sEng levels and increases hepatic expression of Eng. High levels of human sEng results in increased hepatic deposition of cholesterol due to reduced conversion into BA, as well as redirects the metabolism of triglycerides (TAG) to its accumulation in the liver, via reduced TAG elimination by β-oxidation combined with reduced hepatic efflux. We propose that sEng might be a biomarker of NASH development, and the presence of high levels of sEng might support NASH aggravation by impairing the essential defensive mechanism protecting NASH liver against excessive TAG and cholesterol accumulation, suggesting the importance of high sEng levels in patients prone to develop NASH.
Collapse
Affiliation(s)
- Ivone Cristina Igreja Sá
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Milos Hroch
- Department of Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic;
| | - Radomir Hyspler
- Centrum for Research and Development University Hospital, Hradec Kralove, 500 03 Hradec Kralove, Czech Republic; (R.H.); (A.T.)
| | - Alena Ticha
- Centrum for Research and Development University Hospital, Hradec Kralove, 500 03 Hradec Kralove, Czech Republic; (R.H.); (A.T.)
| | - Hana Lastuvkova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.L.); (J.S.); (E.D.)
| | - Jolana Schreiberova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.L.); (J.S.); (E.D.)
| | - Eva Dolezelova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.L.); (J.S.); (E.D.)
| | - Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Iveta Najmanova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Martina Vasinova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| | - Miguel Pericacho
- Biomedical Research Institute of Salamanca and Renal and Cardiovascular Physiopathology Unit, Department of Physiology and Pharmacology, University of Salamanca, 370 06 Salamanca, Spain;
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.L.); (J.S.); (E.D.)
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, 500 05 Hradec Kralove, Czech Republic; (I.C.I.S.); (K.T.); (S.E.); (B.V.); (I.N.); (M.V.)
| |
Collapse
|
26
|
Sun W, Tang Y, Tai YY, Handen A, Zhao J, Speyer G, Al Aaraj Y, Watson A, Romanelli ME, Sembrat J, Rojas M, Simon MA, Zhang Y, Lee J, Xiong Z, Dutta P, Vasamsetti SB, McNamara D, McVerry B, McTiernan CF, Sciurba FC, Kim S, Smith KA, Mazurek JA, Han Y, Vaidya A, Nouraie SM, Kelly NJ, Chan SY. SCUBE1 Controls BMPR2-Relevant Pulmonary Endothelial Function: Implications for Diagnostic Marker Development in Pulmonary Arterial Hypertension. JACC Basic Transl Sci 2020; 5:1073-1092. [PMID: 33294740 PMCID: PMC7691287 DOI: 10.1016/j.jacbts.2020.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022]
Abstract
Utilizing publicly available ribonucleic acid sequencing data, we identified SCUBE1 as a BMPR2-related gene differentially expressed between induced pluripotent stem cell-endothelial cells derived from pulmonary arterial hypertension (PAH) patients carrying pathogenic BMPR2 mutations and control patients without mutations. Endothelial SCUBE1 expression was decreased by known triggers of PAH, and its down-regulation recapitulated known BMPR2-associated endothelial pathophenotypes in vitro. Meanwhile, SCUBE1 concentrations were reduced in plasma obtained from PAH rodent models and patients with PAH, whereas plasma concentrations were tightly correlated with hemodynamic markers of disease severity. Taken together, these data implicate SCUBE1 as a novel contributor to PAH pathogenesis with potential therapeutic, diagnostic, and prognostic applications.
Collapse
Key Words
- BMP, bone morphogenetic protein
- BMPR2
- EC, endothelial cell
- PAEC, pulmonary arterial endothelial cell
- PAH, pulmonary arterial hypertension
- PAP, pulmonary artery pressure
- PCWP, pulmonary capillary wedge pressure
- PH, pulmonary hypertension
- PVR, pulmonary vascular resistance
- RV, right ventricle
- SCUBE1
- WSPH, World Symposium on Pulmonary Hypertension
- endothelium
- iPSC-EC, induced pluripotent stem cell-endothelial cell
- mPAP, mean pulmonary artery pressure
- pulmonary hypertension
Collapse
Affiliation(s)
- Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yi-Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Adam Handen
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Gil Speyer
- Research Computing, Arizona State University, Tempe, Arizona, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Annie Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Makenna E Romanelli
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Marc A Simon
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yingze Zhang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Janet Lee
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zeyu Xiong
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Partha Dutta
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Sathish Badu Vasamsetti
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Dennis McNamara
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Bryan McVerry
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Charles F McTiernan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Frank C Sciurba
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, Roy G. Perry College of Engineering, Prairie View A and M University, Prairie View, Texas, USA
| | - Kerri Akaya Smith
- Division of Pulmonary Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeremy A Mazurek
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuchi Han
- Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anjali Vaidya
- Cardiovascular Division, Temple University Health Systems, Philadelphia, Pennsylvania, USA
| | - Seyed Mehdi Nouraie
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Neil J Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology and Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
27
|
Khandagale A, Åberg M, Wikström G, Bergström Lind S, Shevchenko G, Björklund E, Siegbahn A, Christersson C. Role of Extracellular Vesicles in Pulmonary Arterial Hypertension: Modulation of Pulmonary Endothelial Function and Angiogenesis. Arterioscler Thromb Vasc Biol 2020; 40:2293-2309. [PMID: 32757648 DOI: 10.1161/atvbaha.120.314152] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Extracellular vesicles (EVs) have the potential to act as intercellular communicators. The aims were to characterize circulating EVs in patients with pulmonary arterial hypertension (PAH) and to explore whether these EVs contribute to endothelial activation and angiogenesis. Approach and Results: Patients with PAH (n=70) and healthy controls (HC; n=20) were included in this cross-sectional study. EVs were characterized and human pulmonary endothelial cells (hPAECs) were incubated with purified EVs. Endothelial cell activity and proangiogenic markers were analyzed. Tube formation analysis was performed for hPAECs, and the involvement of PSGL-1 (P-selectin glycoprotein ligand 1) was evaluated. The numbers of CD62P+, CD144+, and CD235a EVs were higher in blood from PAH compared with HC. Thirteen proteins were differently expressed in PAH and HC EVs, where complement fragment C1q was the most significantly elevated protein (P=0.0009) in PAH EVs. Upon EVs-internalization in hPAECs, more PAH compared with HC EVs evaded lysosomes (P<0.01). As oppose to HC, PAH EVs stimulated hPAEC activation and induced transcription and translation of VEGF-A (vascular endothelial growth factor A; P<0.05) and FGF (fibroblast growth factor; P<0.005) which were released in the cell supernatant. These proangiogenic proteins were higher in patient with PAH plasma compered with HC. PAH EVs induced a complex network of angiotubes in vitro, which was abolished by inhibitory PSGL-1antibody. Anti-PSGL-1 also inhibited EV-induced endothelial cell activation and PAH EV dependent increase of VEGF-A. CONCLUSIONS Patients with PAH have higher levels of EVs harboring increased amounts of angiogenic proteins, which induce activation of hPAECs and in vitro angiogenesis. These effects were partly because of platelet-derived EVs evasion of lysosomes upon internalization within hPAEC and through possible involvement of P-selectin-PSGL-1 pathway.
Collapse
Affiliation(s)
- Avinash Khandagale
- From the Department of Medical Sciences, Cardiology and Clinical Chemistry (A.K.), Uppsala University, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry (M.Å., A.S.), Uppsala University, Sweden
| | - Gerhard Wikström
- Department of Medical Sciences, Cardiology and Internal Medicine (G.W.), Uppsala University, Sweden
| | - Sara Bergström Lind
- Department of Chemistry - BMC, Analytical Chemistry (S.B.L., G.S.), Uppsala University, Sweden
| | - Ganna Shevchenko
- Department of Chemistry - BMC, Analytical Chemistry (S.B.L., G.S.), Uppsala University, Sweden
| | - Erik Björklund
- Department of Medical Sciences, Cardiology (E.B., C.C.), Uppsala University, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry (M.Å., A.S.), Uppsala University, Sweden
| | | |
Collapse
|
28
|
Abstract
Endoglin, known to be expressed in proliferating vessels, is of worth when evaluating microvessel density as a prognostic factor in many types of malignancies, including some subtypes of leukemia cells. In childhood acute lymphoblastic leukemia, endoglin is associated with adverse outcome. In bone marrow, endoglin identifies the repopulating hematopoietic stem cells. Mast cells are a component of normal tissue and play an important role in the regulation of several processes, including inflammation and neoplasia. The aim of this study was to evaluate the use of endoglin as a biological marker of mast cells compared with the gold standard stains. We studied 15 specimens of neurofibroma, 9 of mastocytosis, and 6 of fibrous scar tissue through immunohistochemistry (for endoglin and mast cell tryptase) and histochemical staining using toluidine blue. Quantitative analysis of the cells was performed by counting 5 hotspots. The validity of endoglin as a mast cell marker was assessed by intraclass correlation coefficient. The Kruskal-Wallis test was used to compare mast cell count for each marker. A strong endoglin expression was found in the cytoplasmic granules of mast cells within the 3 groups. Similar results were observed with mast cell tryptase as well as toluidine blue. The intraclass correlation coefficient revealed that endoglin is a highly reliable biomarker of mast cells when compared with mast cell tryptase and toluidine blue. In conclusion, endoglin may assist in the diagnosis and pathogenesis study of various processes associated with mast cells. An endoglin-neutralizing treatment for solid cancers and leukemia could also affect mastocytes and the immunologic system.
Collapse
|
29
|
Tzavlaki K, Moustakas A. TGF-β Signaling. Biomolecules 2020; 10:biom10030487. [PMID: 32210029 PMCID: PMC7175140 DOI: 10.3390/biom10030487] [Citation(s) in RCA: 499] [Impact Index Per Article: 99.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) represents an evolutionarily conserved family of secreted polypeptide factors that regulate many aspects of physiological embryogenesis and adult tissue homeostasis. The TGF-β family members are also involved in pathophysiological mechanisms that underlie many diseases. Although the family comprises many factors, which exhibit cell type-specific and developmental stage-dependent biological actions, they all signal via conserved signaling pathways. The signaling mechanisms of the TGF-β family are controlled at the extracellular level, where ligand secretion, deposition to the extracellular matrix and activation prior to signaling play important roles. At the plasma membrane level, TGF-βs associate with receptor kinases that mediate phosphorylation-dependent signaling to downstream mediators, mainly the SMAD proteins, and mediate oligomerization-dependent signaling to ubiquitin ligases and intracellular protein kinases. The interplay between SMADs and other signaling proteins mediate regulatory signals that control expression of target genes, RNA processing at multiple levels, mRNA translation and nuclear or cytoplasmic protein regulation. This article emphasizes signaling mechanisms and the importance of biochemical control in executing biological functions by the prototype member of the family, TGF-β.
Collapse
|
30
|
Hewes JL, Lee JY, Fagan KA, Bauer NN. The changing face of pulmonary hypertension diagnosis: a historical perspective on the influence of diagnostics and biomarkers. Pulm Circ 2020; 10:2045894019892801. [PMID: 32110383 PMCID: PMC7000867 DOI: 10.1177/2045894019892801] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension is a complex, multifactorial disease that results in right heart failure and premature death. Since the initial reports of pulmonary hypertension in the late 1800s, the diagnosis of pulmonary hypertension has evolved with respect to its definition, screening tools, and diagnostic techniques. This historical perspective traces the earliest roots of pulmonary hypertension detection and diagnosis through to the current recommendations for classification. We highlight the diagnostic tools used in the past and present, and end with a focus on the future directions of early detection. Early detection of pulmonary hypertension and pulmonary arterial hypertension and the proper determination of etiology are vital for the early therapeutic intervention that can prolong life expectancy and improve quality of life. The search for a non-invasive screening tool for the identification and classification of pulmonary hypertension is ongoing, and we discuss the role of animal models of the disease in this search.
Collapse
Affiliation(s)
- Jenny L. Hewes
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Ji Young Lee
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
- Department of Physiology and Cell
Biology, College of Medicine,
University
of South Alabama, Mobile, AL, USA
| | - Karen A. Fagan
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Division of Pulmonary and Critical Care
Medicine, University Hospital,
University
of South Alabama, Mobile, AL, USA
| | - Natalie N. Bauer
- Department of Pharmacology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
- Center for Lung Biology, College of
Medicine,
University
of South Alabama, Mobile, AL, USA
| |
Collapse
|
31
|
Abstract
Endoglin (ENG) is a coreceptor of the transforming growth factor-β (TGFβ) family signaling complex, which is highly expressed on endothelial cells and plays a key role in angiogenesis. Its extracellular domain can be cleaved and released into the circulation as soluble ENG (sENG). High circulating levels of sENG contribute to the pathogenesis of preeclampsia (PE). Circulating bone morphogenetic protein 9 (BMP9), a vascular quiescence and endothelial-protective factor, binds sENG with high affinity, but how sENG participates in BMP9 signaling complexes is not fully resolved. sENG was thought to be a ligand trap for BMP9, preventing type II receptor binding and BMP9 signaling. Here we show that, despite cell-surface ENG being a dimer linked by disulfide bonds, sENG purified from human placenta and plasma from PE patients is primarily in a monomeric form. Incubating monomeric sENG with the circulating form of BMP9 (prodomain-bound form) in solution leads to the release of the prodomain and formation of a sENG:BMP9 complex. Furthermore, we demonstrate that binding of sENG to BMP9 does not inhibit BMP9 signaling. Indeed, the sENG:BMP9 complex signals with comparable potency and specificity to BMP9 on human primary endothelial cells. The full signaling activity of the sENG:BMP9 complex required transmembrane ENG. This study confirms that rather than being an inhibitory ligand trap, increased circulating sENG might preferentially direct BMP9 signaling via cell-surface ENG at the endothelium. This is important for understanding the role of sENG in the pathobiology of PE and other cardiovascular diseases.
Collapse
|
32
|
Gaynitdinova VV, Avdeev SN. [Novel Biomarkers of Pulmonary Hypertension]. ACTA ACUST UNITED AC 2019; 59:84-94. [PMID: 31322094 DOI: 10.18087/cardio.2019.7.10259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 11/18/2022]
Abstract
Pulmonary hypertension (PH) is a clinical syndrome characterized by a progressive increase in pulmonary vascular resistance (PVR), which leads to remodeling of the right ventricle (RV), right heart failure and premature death of patients. Early diagnosis and monitoring of disease progression are crucial for making decisions about the necessary therapy. The gold standard for the diagnosis of pulmonary hypertension is the right heart catheterization. The estimation of systolic pressure in pulmonary artery by means of transthoracic echocardiography is also used for monitoring the course of the disease. At present, there is still a need for non-invasive biomarkers that reflect pathological changes in pulmonary arterial vessels and allow diagnosing of PH. Our review outlines the new data about some biomarkers potentially useful for diagnosis and prognostication of PH. These biomarkers (mid-regional pro-adrenomedullin, carboxyterminal pro-endothelin-1, copeptin, asymmetric dimethylarginine, growth differentiation factor 15, and others) are classified based on their relationship to endothelial cell dysfunction, inflammation, epigenetics, cardiac function, oxidative stress, extracellular matrix. The determination of biomarkers that are of diagnostic value for predicting the severity, progression of PH and response to therapy, in a simple blood test or condensate of exhaled air, can significantly reduce treatment costs and improve PH management.
Collapse
Affiliation(s)
| | - S N Avdeev
- Sechenov First Moscow State Medical University (Sechenov University); Pulmonology Research Institute
| |
Collapse
|
33
|
Nikolic I, Yung LM, Yang P, Malhotra R, Paskin-Flerlage SD, Dinter T, Bocobo GA, Tumelty KE, Faugno AJ, Troncone L, McNeil ME, Huang X, Coser KR, Lai CSC, Upton PD, Goumans MJ, Zamanian RT, Elliott CG, Lee A, Zheng W, Berasi SP, Huard C, Morrell NW, Chung RT, Channick RW, Roberts KE, Yu PB. Bone Morphogenetic Protein 9 Is a Mechanistic Biomarker of Portopulmonary Hypertension. Am J Respir Crit Care Med 2019; 199:891-902. [PMID: 30312106 PMCID: PMC6444661 DOI: 10.1164/rccm.201807-1236oc] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
RATIONALE BMP9 (bone morphogenetic protein 9) is a circulating endothelial quiescence factor with protective effects in pulmonary arterial hypertension (PAH). Loss-of-function mutations in BMP9, its receptors, and downstream effectors have been reported in heritable PAH. OBJECTIVES To determine how an acquired deficiency of BMP9 signaling might contribute to PAH. METHODS Plasma levels of BMP9 and antagonist soluble endoglin were measured in group 1 PAH, group 2 and 3 pulmonary hypertension (PH), and in patients with severe liver disease without PAH. MEASUREMENTS AND MAIN RESULTS BMP9 levels were markedly lower in portopulmonary hypertension (PoPH) versus healthy control subjects, or other etiologies of PAH or PH; distinguished PoPH from patients with liver disease without PAH; and were an independent predictor of transplant-free survival. BMP9 levels were decreased in mice with PH associated with CCl4-induced portal hypertension and liver cirrhosis, but were normal in other rodent models of PH. Administration of ALK1-Fc, a BMP9 ligand trap consisting of the activin receptor-like kinase-1 extracellular domain, exacerbated PH and pulmonary vascular remodeling in mice treated with hypoxia versus hypoxia alone. CONCLUSIONS BMP9 is a sensitive and specific biomarker of PoPH, predicting transplant-free survival and the presence of PAH in liver disease. In rodent models, acquired deficiency of BMP9 signaling can predispose to or exacerbate PH, providing a possible mechanistic link between PoPH and heritable PAH. These findings describe a novel experimental model of severe PH that provides insight into the synergy between pulmonary vascular injury and diminished BMP9 signaling in the pathogenesis of PAH.
Collapse
Affiliation(s)
- Ivana Nikolic
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lai-Ming Yung
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peiran Yang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Samuel D. Paskin-Flerlage
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Teresa Dinter
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Geoffrey A. Bocobo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Anthony J. Faugno
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Luca Troncone
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Megan E. McNeil
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Xiuli Huang
- Therapy for Rare and Neglected Diseases Program, National Center for Advancing Translational Sciences, Rockville, Maryland
| | - Kathryn R. Coser
- Pfizer Centers for Therapeutic Innovation, Cambridge, Massachusetts
| | - Carol S. C. Lai
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Paul D. Upton
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s and Papworth Hospitals, Cambridge, United Kingdom
| | - Marie Jose Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Centre, Leiden, the Netherlands
| | - Roham T. Zamanian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University Medical Center, Stanford, California; and
| | - C. Gregory Elliott
- Department of Medicine, Intermountain Medical Center and University of Utah, Salt Lake City, Utah
| | - Arthur Lee
- Therapy for Rare and Neglected Diseases Program, National Center for Advancing Translational Sciences, Rockville, Maryland
| | - Wei Zheng
- Therapy for Rare and Neglected Diseases Program, National Center for Advancing Translational Sciences, Rockville, Maryland
| | | | - Christine Huard
- Pfizer Centers for Therapeutic Innovation, Cambridge, Massachusetts
| | - Nicholas W. Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s and Papworth Hospitals, Cambridge, United Kingdom
| | | | - Richard W. Channick
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kari E. Roberts
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Tufts Medical Center, Boston, Massachusetts
| | - Paul B. Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Rice LM, Mantero JC, Stratton EA, Warburton R, Roberts K, Hill N, Simms RW, Domsic R, Farber HW, Layfatis R. Serum biomarker for diagnostic evaluation of pulmonary arterial hypertension in systemic sclerosis. Arthritis Res Ther 2018; 20:185. [PMID: 30115106 PMCID: PMC6097341 DOI: 10.1186/s13075-018-1679-8] [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: 03/01/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023] Open
Abstract
Background Systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) is one of the leading causes of death in SSc. Identification of a serum-based proteomic diagnostic biomarker for SSc-PAH would allow for rapid non-invasive screening and could positively impact patient survival. Identification and validation of novel proteins could potentially facilitate the identification of SSc-PAH, and might also point to important protein mediators in pathogenesis. Methods Thirteen treatment-naïve SSc-PAH patients had serum collected at time of diagnosis and were used as the discovery cohort for the protein-expression biomarker. Two proteins, Midkine and Follistatin-like 3 (FSTL3) were then validated by enzyme-linked immunosorbent assays. Midkine and FSTL3 were tested in combination to identify SSc-PAH and were validated in two independent cohorts of SSc-PAH (n = 23, n = 11). Results Eighty-two proteins were found to be differentially regulated in SSc-PAH sera. Two proteins (Midkine and FSTL3) were also shown to be elevated in publicly available data and their expression was evaluated in independent cohorts. In the validation cohorts, the combination of Midkine and FSTL3 had an area under the receiver operating characteristic curve (AUC) of 0.85 and 0.92 with respective corresponding measures of sensitivity of 76% and 91%, and specificity measures of 76% and 80%. Conclusions These findings indicate that there is a clear delineation between overall protein expression in sera from SSc patients and those with SSc-PAH. The combination of Midkine and FSTL3 can serve as an SSc-PAH biomarker and are potential drug targets for this rare disease population. Electronic supplementary material The online version of this article (10.1186/s13075-018-1679-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lisa M Rice
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA.
| | - Julio C Mantero
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Eric A Stratton
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | | | | | | | - Robert W Simms
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Robyn Domsic
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Harrison W Farber
- Boston University School of Medicine, E5 Arthritis Center, 72 E Concord Street, Boston, MA, 0211, USA
| | - Robert Layfatis
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Kylhammar D, Hesselstrand R, Nielsen S, Scheele C, Rådegran G. Angiogenic and inflammatory biomarkers for screening and follow-up in patients with pulmonary arterial hypertension. Scand J Rheumatol 2018. [DOI: 10.1080/03009742.2017.1378714] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- D Kylhammar
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden
- Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - R Hesselstrand
- Department of Clinical Sciences Lund, Rheumatology, Faculty of Medicine, Lund University, Lund, Sweden
- Section for Rheumatology, VO Gastroenterology, Nephrology and Rheumatology, Skåne University Hospital, Lund, Sweden
| | - S Nielsen
- Centre for Inflammation and Metabolism, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - C Scheele
- Centre for Inflammation and Metabolism, Faculty of Medicine, University of Copenhagen, Copenhagen, Denmark
| | - G Rådegran
- Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine, Lund University, Lund, Sweden
- Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
36
|
Nadeau V, Potus F, Boucherat O, Paradis R, Tremblay E, Iglarz M, Paulin R, Bonnet S, Provencher S. Dual ET A/ET B blockade with macitentan improves both vascular remodeling and angiogenesis in pulmonary arterial hypertension. Pulm Circ 2017; 8:2045893217741429. [PMID: 29064353 PMCID: PMC5731731 DOI: 10.1177/2045893217741429] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dysregulated metabolism and rarefaction of the capillary network play a critical role in pulmonary arterial hypertension (PAH) etiology. They are associated with a decrease in perfusion of the lungs, skeletal muscles, and right ventricle (RV). Previous studies suggested that endothelin-1 (ET-1) modulates both metabolism and angiogenesis. We hypothesized that dual ETA/ETB receptors blockade improves PAH by improving cell metabolism and promoting angiogenesis. Five weeks after disease induction, Sugen/hypoxic rats presented severe PAH with pulmonary artery (PA) remodeling, RV hypertrophy and capillary rarefaction in the lungs, RV, and skeletal muscles (microCT angiogram, lectin perfusion, CD31 staining). Two-week treatment with dual ETA/ETB receptors antagonist macitentan (30 mg/kg/d) significantly improved pulmonary hemodynamics, PA vascular remodeling, and RV function and hypertrophy compared to vehicle-treated animals (all P = 0.05). Moreover, macitentan markedly increased lung, RV and quadriceps perfusion, and microvascular density (all P = 0.05). In vitro, these effects were associated with increases in oxidative phosphorylation (oxPhox) and markedly reduced cell proliferation of PAH-PA smooth muscle cells (PASMCs) treated with macitentan without affecting apoptosis. While macitentan did not affect oxPhox, proliferation, and apoptosis of PAH-PA endothelial cells (PAECs), it significantly improved their angiogenic capacity (tube formation assay). Exposure of control PASMC and PAEC to ET-1 fully mimicked the PAH cells phenotype, thus confirming that ET-1 is implicated in both metabolism and angiogenesis abnormalities in PAH. Dual ETA/ETB receptor blockade improved the metabolic changes involved in PAH-PASMCs' proliferation and the angiogenic capacity of PAH-PAEC leading to an increased capillary density in lungs, RV, and skeletal muscles.
Collapse
Affiliation(s)
- Valerie Nadeau
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada
| | - Francois Potus
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada
| | - Olivier Boucherat
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada.,3 Department of Medicine, Laval University, Quebec, Canada
| | - Renee Paradis
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada
| | - Eve Tremblay
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada
| | - Marc Iglarz
- 4 Drug Discovery Department, Actelion Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Roxane Paulin
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada.,3 Department of Medicine, Laval University, Quebec, Canada
| | - Sebastien Bonnet
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada.,3 Department of Medicine, Laval University, Quebec, Canada
| | - Steeve Provencher
- 1 Pulmonary Hypertension Research Group ( http://www.hypertensionarteriellepulmonaire.ca ).,2 Institut universitaire de cardiologie et de pneumologie de Québec Research Center, Laval University, Quebec City, Canada.,3 Department of Medicine, Laval University, Quebec, Canada
| |
Collapse
|
37
|
Somashekar ST, Sammour I, Huang J, Dominguez-Bendala J, Pastori R, Alvarez-Cubela S, Torres E, Wu S, Young KC. Intra-Amniotic Soluble Endoglin Impairs Lung Development in Neonatal Rats. Am J Respir Cell Mol Biol 2017; 57:468-476. [PMID: 28590142 DOI: 10.1165/rcmb.2016-0165oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Soluble endoglin (sENG) is increased in the amniotic fluid of women with preeclampsia and chorioamnionitis. Preterm infants born to women with these disorders have an increased risk of aberrant lung development. Whether this increased risk is secondary to elevated sENG levels is unclear. The objective of this study was to determine whether intrauterine exposure to an adenovirus overexpressing sENG impairs neonatal lung angiogenesis by modulating lung endothelial nitric oxide synthase (eNOS) signaling. Pregnant Sprague-Dawley rats were randomly assigned to receive ultrasound-guided intra-amniotic injections of adenovirus overexpressing sENG (Ad-sENG) or control adenovirus (Ad-control) on embryonic day 17. After this exposure, rat pups were maintained in normoxia and evaluated on postnatal day 14. Intra-amniotic Ad-sENG decreased lung vascular endothelial growth factor receptor 2 and eNOS expression in rat pups. This was accompanied by a marked decrease in lung angiogenesis and alveolarization. Ad-sENG-exposed pups also had an increase in right ventricular systolic pressure, weight ratio of right ventricle to left ventricle plus septum, and pulmonary vascular remodeling. In addition, exposure of human pulmonary artery endothelial cells to recombinant sENG reduced endothelial tube formation and protein levels of eNOS, phosphorylated eNOS, and phosphorylated Smad1/5. Together, our findings demonstrate that intrauterine exposure to an adenovirus overexpressing sENG disrupts lung development by impairing Smad1/5-eNOS signaling. We speculate that sENG-mediated dysregulation of Smad1/5-eNOS signaling contributes to impaired lung development and potentially primes the developing lung for further postnatal insults. Further studies exploring the relationship between amniotic fluid sENG levels and preterm respiratory outcomes will be necessary.
Collapse
Affiliation(s)
- Santhosh T Somashekar
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| | - Ibrahim Sammour
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| | - Jian Huang
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| | - Juan Dominguez-Bendala
- 3 Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Ricardo Pastori
- 3 Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Silvia Alvarez-Cubela
- 3 Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Eneida Torres
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| | - Shu Wu
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| | - Karen C Young
- 1 Division of Neonatology, Department of Pediatrics.,2 Neonatal Developmental Biology Laboratory, Batchelor Children's Research Institute, and
| |
Collapse
|
38
|
Säleby J, Bouzina H, Lundgren J, Rådegran G. Angiogenic and inflammatory biomarkers in the differentiation of pulmonary hypertension. SCAND CARDIOVASC J 2017; 51:261-270. [PMID: 28776404 DOI: 10.1080/14017431.2017.1359419] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Pulmonary hypertension (PH) is a serious condition where diagnosis often is delayed due to unspecific symptoms. New methods to diagnose and differentiate PH earlier would therefore be of great value. The aim of this study was therefore to evaluate the relationship between circulating angiogenic and inflammatory biomarkers and various hemodynamic variables in relation to different causes of PH. DESIGN Plasma samples from 63 patients at diagnosis were extracted from Lund Cardio Pulmonary Register, separated into pulmonary arterial hypertension (PAH, n = 22), chronic thromboembolic pulmonary hypertension (CTEPH, n = 15) and left heart disease (LHD) with (n = 21) and without (n = 5) PH. Blood samples from eight control subjects devoid of PH were additionally evaluated. Plasma concentrations of angiogenic (PlGF, Tie2, VEGF-A, VEGF-D, bFGF, sFlt-1) and inflammatory (IL-6, IL-8, TNF-α) biomarkers were analysed and related to hemodynamic variables. RESULTS SFlt-1 (p < .004) and VEGF-A (p < .035) were higher in all PH groups compared to controls. TNF-α (p < .030) were elevated in PAH patients in relation to the other PH groups as well as controls. Likewise, plasma VEGF-D (p < .008) were elevated in LHD with PH compared to the other groups with PH and controls. In PAH, higher sFlt-1 concentrations correlated to a worse state of hemodynamics. CONCLUSIONS Our findings indicate that sFlt-1 and VEGF-A may be future tools when discriminating PH from non-PH. Moreover, TNF-α may differentiate PAH and VEGF- D may differentiate LHD with PH, from the other groups with PH, as well as controls. SFlt-1 may furthermore play a role as a future marker of disease severity.
Collapse
Affiliation(s)
- Joanna Säleby
- a Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine , Lund University , Lund , Sweden.,b The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine , Skåne University Hospital , Lund , Sweden
| | - Habib Bouzina
- a Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine , Lund University , Lund , Sweden.,b The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine , Skåne University Hospital , Lund , Sweden
| | - Jakob Lundgren
- a Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine , Lund University , Lund , Sweden.,b The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine , Skåne University Hospital , Lund , Sweden
| | - Göran Rådegran
- a Department of Clinical Sciences Lund, Cardiology, Faculty of Medicine , Lund University , Lund , Sweden.,b The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO Heart and Lung Medicine , Skåne University Hospital , Lund , Sweden
| |
Collapse
|
39
|
Abou-Fayçal C, Hatat AS, Gazzeri S, Eymin B. Splice Variants of the RTK Family: Their Role in Tumour Progression and Response to Targeted Therapy. Int J Mol Sci 2017; 18:ijms18020383. [PMID: 28208660 PMCID: PMC5343918 DOI: 10.3390/ijms18020383] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 01/24/2017] [Accepted: 01/30/2017] [Indexed: 12/16/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) belong to a family of transmembrane receptors that display tyrosine kinase activity and trigger the activation of downstream signalling pathways mainly involved in cell proliferation and survival. RTK amplification or somatic mutations leading to their constitutive activation and oncogenic properties have been reported in various tumour types. Numerous RTK-targeted therapies have been developed to counteract this hyperactivation. Alternative splicing of pre-mRNA has recently emerged as an important contributor to cancer development and tumour maintenance. Interestingly, RTKs are alternatively spliced. However, the biological functions of RTK splice variants, as well as the upstream signals that control their expression in tumours, remain to be understood. More importantly, it remains to be determined whether, and how, these splicing events may affect the response of tumour cells to RTK-targeted therapies, and inversely, whether these therapies may impact these splicing events. In this review, we will discuss the role of alternative splicing of RTKs in tumour progression and response to therapies, with a special focus on two major RTKs that control proliferation, survival, and angiogenesis, namely, epidermal growth factor receptor (EGFR) and vascular endothelial growth factor receptor-1 (VEGFR1).
Collapse
Affiliation(s)
- Cherine Abou-Fayçal
- Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, University Grenoble Alpes, Grenoble 38702, France.
| | - Anne-Sophie Hatat
- Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, University Grenoble Alpes, Grenoble 38702, France.
| | - Sylvie Gazzeri
- Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, University Grenoble Alpes, Grenoble 38702, France.
| | - Beatrice Eymin
- Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, University Grenoble Alpes, Grenoble 38702, France.
| |
Collapse
|
40
|
DuBrock HM, Rodriguez-Lopez JM, LeVarge BL, Curry MP, VanderLaan PA, Zsengeller ZK, Pernicone E, Preston IR, Yu PB, Nikolic I, Xu D, Thadhani RI, Channick RN, Ananth Karumanchi S. Macrophage migration inhibitory factor as a novel biomarker of portopulmonary hypertension. Pulm Circ 2017; 6:498-507. [PMID: 28090291 DOI: 10.1086/688489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Portopulmonary hypertension (POPH) is a poorly understood complication of liver disease associated with significant morbidity and mortality. We sought to identify novel biomarkers of POPH disease presence and severity. We performed a prospective, multicenter, case-control study involving patients with liver disease undergoing right heart catheterization. POPH cases were defined as a mean pulmonary arterial pressure (mPAP) ≥25 mmHg and pulmonary vascular resistance (PVR) >240 dynes˙s˙cm-5. Plasma samples were collected from the systemic and pulmonary circulation, and antibody microarray was used to identify biomarkers. Characterization and validation of a candidate cytokine, macrophage migration inhibitory factor (MIF), was performed using enzyme-linked immunosorbent assay. Continuous variables were compared using a Mann-Whitney U test and correlated with disease severity using Spearman correlation. MIF levels were elevated in both the systemic and pulmonary circulation in patients with POPH compared with controls (median MIF level [interquartile range] in systemic circulation: 46.68 ng/mL [32.31-76.04] vs. 31.19 ng/mL [26.92-42.17], P = 0.009; in pulmonary circulation: 49.59 ng/mL [35.90-108.80] vs. 37.78 [21.78-45.53], P = 0.002). In patients with POPH, MIF levels were positively correlated with PVR (r = 0.58, P = 0.006) and inversely correlated with cardiac output (r = -0.57, P = 0.007). MIF >60 ng/mL or tricuspid regurgitation gradient >50 mmHg had a 92% sensitivity and specificity for the diagnosis of POPH, with a positive predictive value of 86% and a negative predictive value of 96%. MIF is a promising novel biomarker of POPH disease presence and severity in patients with liver disease and portal hypertension.
Collapse
Affiliation(s)
- Hilary M DuBrock
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | - Michael P Curry
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | - Paul B Yu
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ivana Nikolic
- Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Dihua Xu
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
41
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
42
|
Anwar A, Ruffenach G, Mahajan A, Eghbali M, Umar S. Novel biomarkers for pulmonary arterial hypertension. Respir Res 2016; 17:88. [PMID: 27439993 PMCID: PMC4955255 DOI: 10.1186/s12931-016-0396-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 07/04/2016] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension is a deadly disease characterized by elevated pulmonary arterial pressures leading to right ventricular hypertrophy and failure. The confirmatory gold standard test is the invasive right heart catheterization. The disease course is monitored by pulmonary artery systolic pressure measurement via transthoracic echocardiography. A simple non-invasive test to frequently monitor the patients is much needed. Search for a novel biomarker that can be detected by a simple test is ongoing and many different options are being studied. Here we review some of the new and unique pre-clinical options for potential pulmonary hypertension biomarkers. These biomarkers can be broadly categorized based on their association with endothelial cell dysfunction, inflammation, epigenetics, cardiac function, oxidative stress, metabolism,extracellular matrix, and volatile compounds in exhaled breath condensate. A biomarker that can be detected in blood, urine or breath condensate and correlates with disease severity, progression and response to therapy may result in significant cost reduction and improved patient outcomes.
Collapse
Affiliation(s)
- Anjum Anwar
- Departmentof Anesthesiology, Stanford University, Palo Alto, CA, USA
| | - Gregoire Ruffenach
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Aman Mahajan
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
43
|
Hassoun PM. Inflammation in chronic thromboembolic pulmonary hypertension: accomplice or bystander in altered angiogenesis? Eur Respir J 2016; 46:303-6. [PMID: 26232476 DOI: 10.1183/13993003.00962-2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
Tiede SL, Gall H, Dörr O, Guilherme MDS, Troidl C, Liebetrau C, Voss S, Voswinckel R, Schermuly RT, Seeger W, Grimminger F, Zeiher AM, Dimmeler S, Möllmann H, Hamm CW, Ghofrani HA, Nef HM. New potential diagnostic biomarkers for pulmonary hypertension. Eur Respir J 2015; 46:1390-6. [DOI: 10.1183/13993003.00187-2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 06/08/2015] [Indexed: 01/13/2023]
Abstract
This study aimed to determine whether the vascular endothelial growth factor (VEGF) family members soluble VEGF receptor 1 (also called soluble fms-like tyrosine kinase 1 (sFlt-1)) and placental growth factor (PlGF) could be used as biomarkers for pulmonary hypertension (PH).Consecutive patients undergoing right heart catheterisation were enrolled (those with mean pulmonary arterial pressure ≥25 mmHg were classed as having PH; those with mean pulmonary arterial pressure <25 mmHg acted as non-PH controls). Plasma from the time of PH diagnosis was analysed for PlGF and sFlt-1 using enzyme immunoassays.In total, 247 patients with PH were enrolled: 62 with idiopathic pulmonary arterial hypertension (IPAH), 14 with associated pulmonary arterial hypertension (APAH), 21 with collagen vascular disease (CVD), 26 with pulmonary venous hypertension, 67 with lung disease-associated PH and 57 with chronic thromboembolic PH. The non-PH control group consisted of 40 patients. sFlt-1 plasma levels were significantly higher in patients with IPAH, APAH, CVD and lung disease-associated PH versus controls; PlGF levels were significantly higher in all PH groups versus controls. The combination of sFlt-1 and PlGF resulted in a sensitivity of 83.7% with specificity of 100% for pulmonary arterial hypertension. There was no association between sFlt-1 or PlGF and haemodynamic parameters, 6-min walking distance or survival.In summary, PlGF and sFlt-1 are promising diagnostic biomarkers for PH.
Collapse
|
45
|
McGlinchey N, Johnson MK. Novel serum biomarkers in pulmonary arterial hypertension. Biomark Med 2015; 8:1001-11. [PMID: 25343672 DOI: 10.2217/bmm.14.69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) remains a difficult-to-treat condition with high mortality. Biomarkers are utilized to aid with diagnosis, prognostication and response to treatment. A clinically useful and PAH-specific single biomarker that is easy to measure remains elusive. This is in part due to the heterogeneity of PAH and its complex etiology. Brain natriuretic peptide and its N-terminal fragment are currently the most widely used serum markers; however, several novel serum biomarkers have been investigated recently. Taken individually, the evidence for each of these seems provisionally promising though currently weak overall. It is likely that a multibiomarker panel will be recommended in the future, with the optimal combination yet to be determined.
Collapse
Affiliation(s)
- Neil McGlinchey
- Scottish Pulmonary Vascular Unit, Golden Jubilee National Hospital, Agamemnon Street, Glasgow, UK
| | | |
Collapse
|
46
|
Al-Husseini A, Kraskauskas D, Mezzaroma E, Nordio A, Farkas D, Drake JI, Abbate A, Felty Q, Voelkel NF. Vascular endothelial growth factor receptor 3 signaling contributes to angioobliterative pulmonary hypertension. Pulm Circ 2015; 5:101-16. [PMID: 25992275 DOI: 10.1086/679704] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 10/13/2014] [Indexed: 12/13/2022] Open
Abstract
The mechanisms involved in the development of severe angioobliterative pulmonary arterial hypertension (PAH) are multicellular and complex. Many of the features of human severe PAH, including angioobliteration, lung perivascular inflammation, and right heart failure, are reproduced in the Sugen 5416/chronic hypoxia (SuHx) rat model. Here we address, at first glance, the confusing and paradoxical aspect of the model, namely, that treatment of rats with the antiangiogenic vascular endothelial growth factor (VEGF) receptor 1 and 2 kinase inhibitor, Sugen 5416, when combined with chronic hypoxia, causes angioproliferative pulmonary vascular disease. We postulated that signaling through the unblocked VEGF receptor VEGFR3 (or flt4) could account for some of the pulmonary arteriolar lumen-occluding cell growth. We also considered that Sugen 5416-induced VEGFR1 and VEGFR2 blockade could alter the expression pattern of VEGF isoform proteins. Indeed, in the lungs of SuHx rats we found increased expression of the ligand proteins VEGF-C and VEGF-D as well as enhanced expression of the VEGFR3 protein. In contrast, in the failing right ventricle of SuHx rats there was a profound decrease in the expression of VEGF-B and VEGF-D in addition to the previously described reduction in VEGF-A expression. MAZ51, an inhibitor of VEGFR3 phosphorylation and VEGFR3 signaling, largely prevented the development of angioobliteration in the SuHx model; however, obliterated vessels did not reopen when animals with established PAH were treated with the VEGFR3 inhibitor. Part of the mechanism of vasoobliteration in the SuHx model occurs via VEGFR3. VEGFR1/VEGFR2 inhibition can be initially antiangiogenic by inducing lung vessel endothelial cell apoptosis; however, it can be subsequently angiogenic via VEGF-C and VEGF-D signaling through VEGFR3.
Collapse
Affiliation(s)
- Ayser Al-Husseini
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Donatas Kraskauskas
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Eleanora Mezzaroma
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Andrea Nordio
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Daniela Farkas
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jennifer I Drake
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Quentin Felty
- Department of Environmental and Occupational Health, Florida International University, Miami, Florida, USA
| | - Norbert F Voelkel
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
47
|
Tang H, Chen J, Fraidenburg DR, Song S, Sysol JR, Drennan AR, Offermanns S, Ye RD, Bonini MG, Minshall RD, Garcia JGN, Machado RF, Makino A, Yuan JXJ. Deficiency of Akt1, but not Akt2, attenuates the development of pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2014; 308:L208-20. [PMID: 25416384 DOI: 10.1152/ajplung.00242.2014] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pulmonary vascular remodeling, mainly attributable to enhanced pulmonary arterial smooth muscle cell proliferation and migration, is a major cause for elevated pulmonary vascular resistance and pulmonary arterial pressure in patients with pulmonary hypertension. The signaling cascade through Akt, comprised of three isoforms (Akt1-3) with distinct but overlapping functions, is involved in regulating cell proliferation and migration. This study aims to investigate whether the Akt/mammalian target of rapamycin (mTOR) pathway, and particularly which Akt isoform, contributes to the development and progression of pulmonary vascular remodeling in hypoxia-induced pulmonary hypertension (HPH). Compared with the wild-type littermates, Akt1(-/-) mice were protected against the development and progression of chronic HPH, whereas Akt2(-/-) mice did not demonstrate any significant protection against the development of HPH. Furthermore, pulmonary vascular remodeling was significantly attenuated in the Akt1(-/-) mice, with no significant effect noted in the Akt2(-/-) mice after chronic exposure to normobaric hypoxia (10% O2). Overexpression of the upstream repressor of Akt signaling, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), and conditional and inducible knockout of mTOR in smooth muscle cells were also shown to attenuate the rise in right ventricular systolic pressure and the development of right ventricular hypertrophy. In conclusion, Akt isoforms appear to have a unique function within the pulmonary vasculature, with the Akt1 isoform having a dominant role in pulmonary vascular remodeling associated with HPH. The PTEN/Akt1/mTOR signaling pathway will continue to be a critical area of study in the pathogenesis of pulmonary hypertension, and specific Akt isoforms may help specify therapeutic targets for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Haiyang Tang
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona; Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona; Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jiwang Chen
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Dustin R Fraidenburg
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Shanshan Song
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona; Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona; Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Justin R Sysol
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Abigail R Drennan
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Richard D Ye
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Marcelo G Bonini
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Richard D Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and
| | - Joe G N Garcia
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Roberto F Machado
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ayako Makino
- Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jason X-J Yuan
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona; Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona; Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
48
|
Voelkel NF, Gomez-Arroyo J. The Role of Vascular Endothelial Growth Factor in Pulmonary Arterial Hypertension. The Angiogenesis Paradox. Am J Respir Cell Mol Biol 2014; 51:474-84. [DOI: 10.1165/rcmb.2014-0045tr] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
49
|
Kapur NK, Qiao X, Paruchuri V, Mackey EE, Daly GH, Ughreja K, Morine KJ, Levine J, Aronovitz MJ, Hill NS, Jaffe IZ, Letarte M, Karas RH. Reducing endoglin activity limits calcineurin and TRPC-6 expression and improves survival in a mouse model of right ventricular pressure overload. J Am Heart Assoc 2014; 3:jah3612. [PMID: 25015075 PMCID: PMC4310384 DOI: 10.1161/jaha.114.000965] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Right ventricular (RV) failure is a major cause of mortality worldwide and is often a consequence of RV pressure overload (RVPO). Endoglin is a coreceptor for the profibrogenic cytokine, transforming growth factor beta 1 (TGF-β1). TGF-β1 signaling by the canonical transient receptor protein channel 6 (TRPC-6) was recently reported to stimulate calcineurin-mediated myofibroblast transformation, a critical component of cardiac fibrosis. We hypothesized that reduced activity of the TGF-β1 coreceptor, endoglin, limits RV calcineurin expression and improves survival in RVPO. METHODS AND RESULTS We first demonstrate that endoglin is required for TGF-β1-mediated calcineurin/TRPC-6 expression and up-regulation of alpha-smooth muscle antigen (α-SMA), a marker of myofibroblast transformation, in human RV fibroblasts. Using endoglin haploinsufficient mice (Eng(+/-)) we show that reduced endoglin activity preserves RV function, limits RV fibrosis, and attenuates activation of the calcineurin/TRPC-6/α-SMA pathway in a model of angio-obliterative pulmonary hypertension. Next, using Eng(+/-) mice or a neutralizing antibody (Ab) against endoglin (N-Eng) in wild-type mice, we show that reduced endoglin activity improves survival and attenuates RV fibrosis in models of RVPO induced by pulmonary artery constriction. To explore the utility of targeting endoglin, we observed a reversal of RV fibrosis and calcineurin levels in wild-type mice treated with a N-Eng Ab, compared to an immunoglobulin G control. CONCLUSION These data establish endoglin as a regulator of TGF-β1 signaling by calcineurin and TRPC-6 in the RV and identify it as a potential therapeutic target to limit RV fibrosis and improve survival in RVPO, a common cause of death in cardiac and pulmonary disease.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Calcineurin/genetics
- Calcineurin/metabolism
- Disease Models, Animal
- Endoglin
- Fibroblasts/metabolism
- Heart Ventricles/cytology
- Heart Ventricles/metabolism
- Humans
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice
- Mice, Knockout
- Myofibroblasts/metabolism
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction
- Survival Rate
- TRPC Cation Channels/genetics
- TRPC Cation Channels/metabolism
- TRPC6 Cation Channel
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Ventricular Dysfunction, Right/genetics
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/physiopathology
Collapse
Affiliation(s)
- Navin K. Kapur
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Xiaoying Qiao
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Vikram Paruchuri
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Emily E. Mackey
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Gerard H. Daly
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Keshan Ughreja
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Kevin J. Morine
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Jonathan Levine
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Mark J. Aronovitz
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Nicholas S. Hill
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Iris Z. Jaffe
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| | - Michelle Letarte
- Molecular Structure and Function Program, Hospital for
Sick Children, and The Heart and Stroke Foundation Richard Lewar Center of Excellence, University of
Toronto, Toronto, Ontario, Canada (M.L.)
| | - Richard H. Karas
- The Molecular Cardiology Research Institute and Surgical
Research Laboratories, Tufts Medical Center and Tufts University School of Medicine,
Boston, MA (N.K.K., X.Q., V.P., E.E.M., G.H.D., K.U., K.J.M., J.L., M.J.A.,
N.S.H., I.Z.J., R.H.K.)
| |
Collapse
|
50
|
Voelkel NF. Pulmonary vascular diseases: in search of a hub among the spokes-an exercise in hypothesis generation. Pulm Circ 2014; 3:723-7. [PMID: 25006390 DOI: 10.1086/674771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Norbert F Voelkel
- Victoria Johnson Laboratory for Lung Research, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|