1
|
Wang P, Zhang X, Yao M, Li J, Wei X, Qiu Z, Chen L, Zhang L. Targeting high mobility group protein B2 exerts antiproliferative effects in hypoxic pulmonary hypertension by modulating miR-21. Toxicol Appl Pharmacol 2025; 497:117265. [PMID: 39952300 DOI: 10.1016/j.taap.2025.117265] [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: 11/24/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is characterized by excessive vascular cell proliferation, leading to vascular remodeling. In this study, we aimed to investigate the molecular mechanisms underlying the regulation of vascular cell proliferation in the context of HMGB2 and its potential involvement in the pathogenesis of PH. METHODS Animals and pulmonary vascular smooth muscle cells (PASMCs) were exposed to hypoxia. Pathological changes in pulmonary vessels were detected by HE and Masson staining. The effect of HMGB2 on cell proliferation was detected by siRNA transfections and recombinant protein treatment. miR-21 inhibitor and mimics were applied, and TPM1 expression was detected. HMGB2-/- mice were applied to observe the possible preventive effect of HMGB2 in PH development. RESULTS HMGB2 expression was increased in hypoxic rats and PASMCs. Silencing ZDHHC5 reduced HMGB2 expression and cell proliferation. Cell proliferation was inhibited by knocking down HMGB2 and promoted by its over-expression. Hypoxia-induced miR-21 upregulation and TPM1 downregulation were mediated by HMGB2. 8-Br-cGMP suppressed HMGB2-induced PASMC proliferation and increased SOX2 expression by activating the cGMP/PKG signaling pathway. HMGB2-/- attenuated pulmonary vascular remodeling and fibrosis in hypoxia induced PH mice. CONCLUSIONS HMGB2 promotes PASMC proliferation through the cGMP/PKG-SOX2-miR-21-TPM1 pathway, which provides a new theoretical basis and possible targets for the pathogenesis and clinical prevention of PH.
Collapse
MESH Headings
- Animals
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/physiology
- Male
- HMGB2 Protein/genetics
- HMGB2 Protein/metabolism
- Hypoxia/metabolism
- Hypoxia/complications
- Mice
- Rats
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Rats, Sprague-Dawley
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Vascular Remodeling
- Mice, Inbred C57BL
- Mice, Knockout
- Tropomyosin/metabolism
- Tropomyosin/genetics
- Cells, Cultured
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Pan Wang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China; Department of Clinical Laboratory, Xi'an Fifth Hospital, Xi'an, China
| | - Xu Zhang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China
| | - Mengge Yao
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China
| | - Jiakang Li
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China
| | - Xiaozhen Wei
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China
| | - Zhihuang Qiu
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China.
| | - Li Zhang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, China; Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, China.
| |
Collapse
|
2
|
Feng J, Liu Y, Li K, Wu Y. Challenges and opportunities in targeting epigenetic mechanisms for pulmonary arterial hypertension treatment. Int J Pharm 2025; 672:125332. [PMID: 39929327 DOI: 10.1016/j.ijpharm.2025.125332] [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: 11/21/2024] [Revised: 01/16/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disorder characterized by elevated pulmonary vascular resistance and pulmonary artery pressure, resulting from a multitude of etiological factors. If left untreated, PAH progressively leads to right heart failure and is associated with high mortality. The etiology of PAH is multifactorial, encompassing both congenital genetic predispositions and acquired secondary influences. Epigenetics, which refers to the regulation of gene expression through chromosomal alterations that do not involve changes in the DNA sequence, has garnered significant attention in PAH research. This includes mechanisms such as DNA methylation, histone modification, and RNA modification. Aberrant epigenetic modifications have been closely linked to the dysregulated proliferation and apoptosis of pulmonary artery smooth muscle cells and endothelial cells, suggesting that these alterations may serve as pivotal drivers of the pathophysiological changes observed in PAH. This review examines the potential impact of epigenetic alterations on the pathogenesis of PAH, highlighting their promise as therapeutic targets. Furthermore, we explore emerging therapeutic strategies and compounds aimed at modulating these epigenetic markers, and discusses their potential applications in both preclinical models and clinical trials. As our understanding of epigenetics deepens, it holds the potential to unlock novel avenues for the precise, individualized treatment of PAH, offering a new frontier in the fight against this debilitating disease.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yunman Liu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
3
|
Aaronson PI. The Role of Hydrogen Sulfide in the Regulation of the Pulmonary Vasculature in Health and Disease. Antioxidants (Basel) 2025; 14:341. [PMID: 40227402 PMCID: PMC11939758 DOI: 10.3390/antiox14030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The gasotransmitter hydrogen sulfide (H2S; also termed sulfide) generally acts as a vasodilator in the systemic vasculature but causes a paradoxical constriction of pulmonary arteries (PAs). In light of evidence that a fall in the partial pressure in oxygen (pO2) increases cellular sulfide levels, it was proposed that a rise in sulfide in pulmonary artery smooth muscle cells (PASMCs) is responsible for hypoxic pulmonary vasoconstriction, the contraction of PAs which develops rapidly in lung regions undergoing alveolar hypoxia. In contrast, pulmonary hypertension (PH), a sustained elevation of pulmonary artery pressure (PAP) which can develop in the presence of a diverse array of pathological stimuli, including chronic hypoxia, is associated with a decrease in the expression of sulfide -producing enzymes in PASMCs and a corresponding fall in sulfide production by the lung. Evidence that PAP in animal models of PH can be lowered by administration of exogenous sulfide has led to an interest in using sulfide-donating agents for treating this condition in humans. Notably, intracellular H2S exists in equilibrium with other sulfur-containing species such as polysulfides and persulfides, and it is these reactive sulfur species which are thought to mediate most of its effects on cells through persulfidation of cysteine thiols on proteins, leading to changes in function in a manner similar to thiol oxidation by reactive oxygen species. This review sets out what is currently known about the mechanisms by which H2S and related sulfur species exert their actions on pulmonary vascular tone, both acutely and chronically, and discusses the potential of sulfide-releasing drugs as treatments for the different types of PH which arise in humans.
Collapse
Affiliation(s)
- Philip I Aaronson
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK
| |
Collapse
|
4
|
Austin ED, Aldred MA, Alotaibi M, Gräf S, Nichols WC, Trembath RC, Chung WK. Genetics and precision genomics approaches to pulmonary hypertension. Eur Respir J 2024; 64:2401370. [PMID: 39209481 PMCID: PMC11525347 DOI: 10.1183/13993003.01370-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024]
Abstract
Considerable progress has been made in the genomics of pulmonary arterial hypertension (PAH) since the 6th World Symposium on Pulmonary Hypertension, with the identification of rare variants in several novel genes, as well as common variants that confer a modest increase in PAH risk. Gene and variant curation by an expert panel now provides a robust framework for knowing which genes to test and how to interpret variants in clinical practice. We recommend that genetic testing be offered to specific subgroups of symptomatic patients with PAH, and to children with certain types of group 3 pulmonary hypertension (PH). Testing of asymptomatic family members and the use of genetics in reproductive decision-making require the involvement of genetics experts. Large cohorts of PAH patients with biospecimens now exist and extension to non-group 1 PH has begun. However, these cohorts are largely of European origin; greater diversity will be essential to characterise the full extent of genomic variation contributing to PH risk and treatment responses. Other types of omics data are also being incorporated. Furthermore, to advance gene- and pathway-specific care and targeted therapies, gene-specific registries will be essential to support patients and their families and to lay the foundation for genetically informed clinical trials. This will require international outreach and collaboration between patients/families, clinicians and researchers. Ultimately, harmonisation of patient-derived biospecimens, clinical and omic information, and analytic approaches will advance the field.
Collapse
Affiliation(s)
- Eric D. Austin
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mona Alotaibi
- University of California San Diego, San Diego, CA, USA
| | - Stefan Gräf
- Department of Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart and Lung Research Institute, Cambridge, UK
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Richard C. Trembath
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Wendy K. Chung
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Popescu AI, Rață AL, Vlad D, Vlad C, Popescu R, Onofrei RR, Morelli M, Pantea S, Barac S. miRNA in the Diagnosis and Treatment of Critical Limb Ischemia. Biomedicines 2024; 12:2026. [PMID: 39335540 PMCID: PMC11428243 DOI: 10.3390/biomedicines12092026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Chronic threatening limb ischemia of the inferior limbs (CLTI) is the final stage of peripheral arterial disease (PAD) and is one of the most feared atherosclerotic manifestations because if left untreated, in time, it can lead to amputation. Although there are currently numerous treatment techniques, both open and endovascular, it is a pathology that has no underlying treatment. Therefore, current studies are very much focused on new therapeutic possibilities that can be applied in the early stages of the atherosclerotic process. In numerous studies in the literature, miRNAs have been identified as important markers of atherosclerosis. The present study aims to identify the expression of three miRNAs-miR-199a, miR-20a, and miR-30c-in patients with chronic limb-threatening ischemia in the pre- and post-revascularization periods. The aim of the study is to identify whether these three markers play a role in critical ischemia and whether they have the potential for future use in new treatments of this pathology.
Collapse
Affiliation(s)
- Alexandra Ioana Popescu
- Pharmacology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Andreea Luciana Rață
- Surgical Emergencies Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Daliborca Vlad
- Pharmacology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cristian Vlad
- Pharmacology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Roxana Popescu
- Cell and Molecular Biology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Ramona Roxana Onofrei
- Department of Rehabilitation, Physical Medicine and Rheumatology, Research Center for Assessment of Human Motion, Functionality and Disability, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Marialuisa Morelli
- Vascular Surgery Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Stelian Pantea
- Surgical Emergencies Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Sorin Barac
- Vascular Surgery Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
6
|
Ejikeme C, Safdar Z. Exploring the pathogenesis of pulmonary vascular disease. Front Med (Lausanne) 2024; 11:1402639. [PMID: 39050536 PMCID: PMC11267418 DOI: 10.3389/fmed.2024.1402639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a complex cardiopulmonary disorder impacting the lung vasculature, resulting in increased pulmonary vascular resistance that leads to right ventricular dysfunction. Pulmonary hypertension comprises of 5 groups (PH group 1 to 5) where group 1 pulmonary arterial hypertension (PAH), results from alterations that directly affect the pulmonary arteries. Although PAH has a complex pathophysiology that is not completely understood, it is known to be a multifactorial disease that results from a combination of genetic, epigenetic and environmental factors, leading to a varied range of symptoms in PAH patients. PAH does not have a cure, its incidence and prevalence continue to increase every year, resulting in higher morbidity and mortality rates. In this review, we discuss the different pathologic mechanisms with a focus on epigenetic modifications and their roles in the development and progression of PAH. These modifications include DNA methylation, histone modifications, and microRNA dysregulation. Understanding these epigenetic modifications will improve our understanding of PAH and unveil novel therapeutic targets, thus steering research toward innovative treatment strategies.
Collapse
Affiliation(s)
| | - Zeenat Safdar
- Department of Pulmonary-Critical Care Medicine, Houston Methodist Lung Center, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
7
|
Efentakis P, Symeonidi L, Gianniou DD, Mikropoulou EV, Giardoglou P, Valakos D, Vatsellas G, Tsota M, Kostomitsopoulos N, Smyrnioudis I, Trougakos IP, Halabalaki M, Dedoussis GV, Andreadou I. Antihypertensive Potential of Pistacia lentiscus var. Chia: Molecular Insights and Therapeutic Implications. Nutrients 2024; 16:2152. [PMID: 38999899 PMCID: PMC11243328 DOI: 10.3390/nu16132152] [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: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Background: Hypertension poses a significant global health burden and is associated with cardiovascular morbidity. Chios mastic gum (CMG), derived from Pistacia lentiscus var. Chia, shows potential as a phytotherapeutic agent, due to its multifaceted beneficial effects. However, its anti-hypertensive effects and vascular, circulatory, and renal-related dysfunction, have not been thoroughly investigated. Herein, we aimed to explore the antihypertensive potential of CMG, focusing on vascular and renal endothelium, in vivo. Methods: Two models of hypertension in male rats, induced by Angiotensin II and Deoxycorticosterone acetate (DOCA)-high-salt administration, were utilized. CMG was administered at 220 mg/kg daily for four weeks after hypertension onset and blood pressure was measured non-invasively. Whole blood RNA sequencing, metabolomics, real-time PCR, and Western blot analyses of kidney and aorta tissues were additionally performed. Results: CMG significantly lowered systolic, diastolic, and mean blood pressure in both models. RNA sequencing revealed that CMG modulated immunity in the Angiotensin II model and metabolism in the DOCA-HS model. CMG downregulated genes related to oxidative stress and endothelial dysfunction and upregulated endothelial markers such as Vegfa. Metabolomic analysis indicated improved endothelial homeostasis via lysophosphatidylinositol upregulation. Conclusions: CMG emerges as a potent natural antihypertensive therapy, demonstrating beneficial effects on blood pressure and renal endothelial function.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| | - Lydia Symeonidi
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| | - Despoina D. Gianniou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (D.D.G.); (I.P.T.)
| | - Eleni V. Mikropoulou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.V.M.); (M.H.)
| | - Panagiota Giardoglou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Dimitrios Valakos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (D.V.); (N.K.)
| | - Giannis Vatsellas
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Maria Tsota
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Nikolaos Kostomitsopoulos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (D.V.); (N.K.)
| | | | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, 15784 Athens, Greece; (D.D.G.); (I.P.T.)
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece; (E.V.M.); (M.H.)
| | - Georgios V. Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, 17671 Athens, Greece; (P.G.); (M.T.); (G.V.D.)
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, 15771 Zografou, Greece; (P.E.); (L.S.)
| |
Collapse
|
8
|
Correale M, Chirivì F, Bevere EML, Tricarico L, D’Alto M, Badagliacca R, Brunetti ND, Vizza CD, Ghio S. Endothelial Function in Pulmonary Arterial Hypertension: From Bench to Bedside. J Clin Med 2024; 13:2444. [PMID: 38673717 PMCID: PMC11051060 DOI: 10.3390/jcm13082444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Pulmonary arterial hypertension is a complex pathology whose etiology is still not completely well clarified. The pathogenesis of pulmonary arterial hypertension involves different molecular mechanisms, with endothelial dysfunction playing a central role in disease progression. Both individual genetic predispositions and environmental factors seem to contribute to its onset. To further understand the complex relationship between endothelial and pulmonary hypertension and try to contribute to the development of future therapies, we report a comprehensive and updated review on endothelial function in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Francesco Chirivì
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Ester Maria Lucia Bevere
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Lucia Tricarico
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Michele D’Alto
- Department of Cardiology, A.O.R.N. dei Colli, Monaldi Hospital, University of Campania L. ‘Vanvitelli’, 80133 Naples, Italy;
| | - Roberto Badagliacca
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Natale D. Brunetti
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Carmine Dario Vizza
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| |
Collapse
|
9
|
Comarița IK, Tanko G, Anghelache IL, Georgescu A. The siRNA-mediated knockdown of AP-1 restores the function of the pulmonary artery and the right ventricle by reducing perivascular and interstitial fibrosis and key molecular players in cardiopulmonary disease. J Transl Med 2024; 22:137. [PMID: 38317144 PMCID: PMC10845748 DOI: 10.1186/s12967-024-04933-1] [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: 11/10/2023] [Accepted: 01/26/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a complex multifactorial vascular pathology characterized by an increased pulmonary arterial pressure, vasoconstriction, remodelling of the pulmonary vasculature, thrombosis in situ and inflammation associated with right-side heart failure. Herein, we explored the potential beneficial effects of treatment with siRNA AP-1 on pulmonary arterial hypertension (PAH), right ventricular dysfunction along with perivascular and interstitial fibrosis in pulmonary artery-PA, right ventricle-RV and lung in an experimental animal model of monocrotaline (MCT)-induced PAH. METHODS Golden Syrian hamsters were divided into: (1) C group-healthy animals taken as control; (2) MCT group obtained by a single subcutaneous injection of 60 mg/kg MCT at the beginning of the experiment; (3) MCT-siRNA AP-1 group received a one-time subcutaneous dose of MCT and subcutaneous injections containing 100 nM siRNA AP-1, every two weeks. All animal groups received water and standard chow ad libitum for 12 weeks. RESULTS In comparison with the MCT group, siRNA AP-1 treatment had significant beneficial effects on investigated tissues contributing to: (1) a reduction in TGF-β1/ET-1/IL-1β/TNF-α plasma concentrations; (2) a reduced level of cytosolic ROS production in PA, RV and lung and notable improvements regarding the ultrastructure of these tissues; a decrease of inflammatory and fibrotic marker expressions in PA (COL1A/Fibronectin/Vimentin/α-SMA/CTGF/Calponin/MMP-9), RV and lung (COL1A/CTGF/Fibronectin/α-SMA/F-actin/OB-cadherin) and an increase of endothelial marker expressions (CD31/VE-cadherin) in PA; (4) structural and functional recoveries of the PA [reduced Vel, restored vascular reactivity (NA contraction, ACh relaxation)] and RV (enlarged internal cavity diameter in diastole, increased TAPSE and PRVOFs) associated with a decrease in systolic and diastolic blood pressure, and heart rate; (5) a reduced protein expression profile of AP-1S3/ pFAK/FAK/pERK/ERK and a significant decrease in the expression levels of miRNA-145, miRNA-210, miRNA-21, and miRNA-214 along with an increase of miRNA-124 and miRNA-204. CONCLUSIONS The siRNA AP-1-based therapy led to an improvement of pulmonary arterial and right ventricular function accompanied by a regression of perivascular and interstitial fibrosis in PA, RV and lung and a down-regulation of key inflammatory and fibrotic markers in MCT-treated hamsters.
Collapse
Affiliation(s)
- Ioana Karla Comarița
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania
| | - Gabriela Tanko
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania
| | | | - Adriana Georgescu
- Institute of Cellular Biology and Pathology 'Nicolae Simionescu' of Romanian Academy, Bucharest, Romania.
| |
Collapse
|
10
|
Chang W, Lee W, Lin Y, Shih J, Hong C, Chen Z, Chu C, Hsu C. Transpulmonary Expression of Exosomal microRNAs in Idiopathic and Congenital Heart Disease-Related Pulmonary Arterial Hypertension. J Am Heart Assoc 2023; 12:e031435. [PMID: 38014665 PMCID: PMC10727351 DOI: 10.1161/jaha.123.031435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Pulmonary artery hypertension (PAH) is a fatal disease characterized by a complex pathogenesis. Exosomes containing microRNAs (miRs) have emerged as a novel biomarker. Transpulmonary exosomal miRs offer valuable insights into pulmonary circulation microenvironments. Hereby, we aimed to explore the potentials of transpulmonary exosomal miRs as differentiating factors between idiopathic PAH and congenital heart disease (CHD)-related PAH. METHODS AND RESULTS During right heart catheterization, we collected exosomes at pulmonary arteries in 25 patients diagnosed with idiopathic PAH and 20 patients with CHD-related PAH. Next-generation sequencing identified several candidate exosomal miRs. Using quantitative polymerase chain reaction, we validated the expressions of these miRs and revealed significantly elevated expressions of miR-21, miR-139-5p, miR-155-5p, let-7f-5p, miR-328-3p, miR-330-3p, and miR-103a-3p in patients with CHD-related PAH, in contrast to patients with idiopathic PAH. Among these miRs, miR-21 exhibited the highest expression in patients with CHD-related PAH. These findings were further corroborated in an external cohort comprising 10 patients with idiopathic PAH and 8 patients with CHD-related PAH. Using an in vitro flow model simulating the shear stress experienced by pulmonary endothelial cells, we observed a significant upregulation of miR-21. Suppressing miR-21 rescued the shear stress-induced downregulation of the RAS/phosphatidylinositol 3-kinase/protein kinase B pathway, leading to a mitigation of apoptosis. CONCLUSIONS Our study identified a pronounced expression of transpulmonary exosomal miR-21, particularly in patients with CHD-related PAH, through next-generation sequencing analysis. Further investigation is warranted to elucidate the regulatory mechanisms involving miR-21 in the pathophysiology of PAH.
Collapse
Affiliation(s)
- Wei‐Ting Chang
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver DiseaseNational Sun Yat‐sen UniversityKaohsiungTaiwan
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
| | - Wei‐Chieh Lee
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- School of Medicine, College of MedicineNational Sun Yat‐sen UniversityKaohsiungTaiwan
| | - Yu‐Wen Lin
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
| | - Jhih‐Yuan Shih
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- School of Medicine, College of MedicineNational Sun Yat‐sen UniversityKaohsiungTaiwan
| | - Chon‐Seng Hong
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- Department of Health and NutritionChia Nan University of Pharmacy and ScienceTainanTaiwan
| | - Zhih‐Cherng Chen
- Division of Cardiology, Department of Internal MedicineChi Mei Medical CenterTainanTaiwan
- School of Medicine, College of MedicineNational Sun Yat‐sen UniversityKaohsiungTaiwan
| | - Chun‐Yuan Chu
- Division of Cardiology, Department of Internal MedicineKaohsiung Medical University HospitalKaohsiungTaiwan
| | - Chih‐Hsin Hsu
- Division of Critical Care, Department of Internal MedicineNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
11
|
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
|
12
|
Dave J, Jagana V, Janostiak R, Bisserier M. Unraveling the epigenetic landscape of pulmonary arterial hypertension: implications for personalized medicine development. J Transl Med 2023; 21:477. [PMID: 37461108 DOI: 10.1186/s12967-023-04339-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease associated with the remodeling of pulmonary blood vessels. If left unaddressed, PAH can lead to right heart failure and even death. Multiple biological processes, such as smooth muscle proliferation, endothelial dysfunction, inflammation, and resistance to apoptosis, are associated with PAH. Increasing evidence suggests that epigenetic factors play an important role in PAH by regulating the chromatin structure and altering the expression of critical genes. For example, aberrant DNA methylation and histone modifications such as histone acetylation and methylation have been observed in patients with PAH and are linked to vascular remodeling and pulmonary vascular dysfunction. In this review article, we provide a comprehensive overview of the role of key epigenetic targets in PAH pathogenesis, including DNA methyltransferase (DNMT), ten-eleven translocation enzymes (TET), switch-independent 3A (SIN3A), enhancer of zeste homolog 2 (EZH2), histone deacetylase (HDAC), and bromodomain-containing protein 4 (BRD4). Finally, we discuss the potential of multi-omics integration to better understand the molecular signature and profile of PAH patients and how this approach can help identify personalized treatment approaches.
Collapse
Affiliation(s)
- Jaydev Dave
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Vineeta Jagana
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Radoslav Janostiak
- First Faculty of Medicine, BIOCEV, Charles University, Vestec, 25250, Czech Republic
| | - Malik Bisserier
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
| |
Collapse
|
13
|
Wang D, Luo MY, Tian Y, Zhang J, Liang N, Li NP, Gong SX, Wang AP. Critical miRNAs in regulating pulmonary hypertension: A focus on Signaling pathways and therapeutic Targets. Anal Biochem 2023:115228. [PMID: 37393975 DOI: 10.1016/j.ab.2023.115228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
Pulmonary hypertension (PH) is complex disease as a result of obstructive pulmonary arterial remodeling, which in turn results in elevated pulmonary arterial pressure (PAP) and subsequent right ventricular heart failure, eventually leading to premature death. However, there is still a lack of a diagnostic blood-based biomarker and therapeutic target for PH. Because of the difficulty of diagnosis, new and more easily accessible prevention and treatment strategy are being explored. New target and diagnosis biomarkers should also allow for early diagnosis. In biology, miRNAs are short endogenous RNA molecules that are not coding. It is known that miRNAs can regulate gene expression and affect a variety of biological processes. Besides, miRNAs have been proven to be a crucial factor in PH pathogenesis. miRNAs have various effects on pulmonary vascular remodeling and are expressed differentially in various pulmonary vascular cells. Nowadays, it has been shown to be critical in the functions of different miRNAs in the pathogenesis of PH. Therefore, clarifying the mechanism of miRNAs regulating pulmonary vascular remodeling is of great importance to explore new therapeutic targets of PH and improve the survival qualify and time of patients. This review is focused on the role, mechanism, and potential therapeutic targets of miRNAs in PH and puts forward possible clinical treatment strategies.
Collapse
Affiliation(s)
- Di Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China
| | - Meng-Yi Luo
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Ying Tian
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Jing Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China
| | - Na Liang
- Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Nan-Ping Li
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Department of Anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China.
| | - Ai-Ping Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, PR China; Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, PR China.
| |
Collapse
|
14
|
Singh DD, Kim Y, Choi SA, Han I, Yadav DK. Clinical Significance of MicroRNAs, Long Non-Coding RNAs, and CircRNAs in Cardiovascular Diseases. Cells 2023; 12:1629. [PMID: 37371099 DOI: 10.3390/cells12121629] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/17/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Based on recent research, the non-coding genome is essential for controlling genes and genetic programming during development, as well as for health and cardiovascular diseases (CVDs). The microRNAs (miRNAs), lncRNAs (long ncRNAs), and circRNAs (circular RNAs) with significant regulatory and structural roles make up approximately 99% of the human genome, which does not contain proteins. Non-coding RNAs (ncRNA) have been discovered to be essential novel regulators of cardiovascular risk factors and cellular processes, making them significant prospects for advanced diagnostics and prognosis evaluation. Cases of CVDs are rising due to limitations in the current therapeutic approach; most of the treatment options are based on the coding transcripts that encode proteins. Recently, various investigations have shown the role of nc-RNA in the early diagnosis and treatment of CVDs. Furthermore, the development of novel diagnoses and treatments based on miRNAs, lncRNAs, and circRNAs could be more helpful in the clinical management of patients with CVDs. CVDs are classified into various types of heart diseases, including cardiac hypertrophy (CH), heart failure (HF), rheumatic heart disease (RHD), acute coronary syndrome (ACS), myocardial infarction (MI), atherosclerosis (AS), myocardial fibrosis (MF), arrhythmia (ARR), and pulmonary arterial hypertension (PAH). Here, we discuss the biological and clinical importance of miRNAs, lncRNAs, and circRNAs and their expression profiles and manipulation of non-coding transcripts in CVDs, which will deliver an in-depth knowledge of the role of ncRNAs in CVDs for progressing new clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur 303002, India
| | - Youngsun Kim
- Department of Obstetrics and Gynecology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Ah Choi
- Division of Pediatric Neurosurgery, Pediatric Clinical Neuroscience Center, Seoul National University Children's Hospital, Seoul 08826, Republic of Korea
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Plasma Biodisplay, Kwangwoon University, Seoul 01897, Republic of Korea
| | - Dharmendra Kumar Yadav
- Department of Pharmacy, Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Incheon 21924, Republic of Korea
| |
Collapse
|
15
|
Hafeez N, Kirillova A, Yue Y, Rao RJ, Kelly NJ, El Khoury W, Al Aaraj Y, Tai Y, Handen A, Tang Y, Jiang D, Wu T, Zhang Y, McNamara D, Kudryashova TV, Goncharova EA, Goncharov D, Bertero T, Nouraie M, Li G, Sun W, Chan SY. Single Nucleotide Polymorphism rs9277336 Controls the Nuclear Alpha Actinin 4-Human Leukocyte Antigen-DPA1 Axis and Pulmonary Endothelial Pathophenotypes in Pulmonary Arterial Hypertension. J Am Heart Assoc 2023; 12:e027894. [PMID: 36974749 PMCID: PMC10122886 DOI: 10.1161/jaha.122.027894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/06/2023] [Indexed: 03/29/2023]
Abstract
Background Pulmonary arterial hypertension (PAH) is a complex, fatal disease where disease severity has been associated with the single nucleotide polymorphism (SNP) rs2856830, located near the human leukocyte antigen DPA1 (HLA-DPA1) gene. We aimed to define the genetic architecture of functional variants associated with PAH disease severity by identifying allele-specific binding transcription factors and downstream targets that control endothelial pathophenotypes and PAH. Methods and Results Electrophoretic mobility shift assays of oligonucleotides containing SNP rs2856830 and 8 SNPs in linkage disequilibrium revealed functional SNPs via allele-imbalanced binding to human pulmonary arterial endothelial cell nuclear proteins. DNA pulldown proteomics identified SNP-binding proteins. SNP genotyping and clinical correlation analysis were performed in 84 patients with PAH at University of Pittsburgh Medical Center and in 679 patients with PAH in the All of Us database. SNP rs9277336 was identified as a functional SNP in linkage disequilibrium (r2>0.8) defined by rs2856830, and the minor allele was associated with decreased hospitalizations and improved cardiac output in patients with PAH, an index of disease severity. SNP pulldown proteomics showed allele-specific binding of nuclear ACTN4 (alpha actinin 4) protein to rs9277336 minor allele. Both ACTN4 and HLA-DPA1 were downregulated in pulmonary endothelium in human patients and rodent models of PAH. Via transcriptomic and phenotypic analyses, knockdown of HLA-DPA1 phenocopied knockdown of ACTN4, both similarly controlling cell structure pathways, immune pathways, and endothelial dysfunction. Conclusions We defined the pathogenic activity of functional SNP rs9277336, entailing the allele-specific binding of ACTN4 and controlling expression of the neighboring HLA-DPA1 gene. Through inflammatory or genetic means, downregulation of this ACTN4-HLA-DPA1 regulatory axis promotes endothelial pathophenotypes, providing a mechanistic explanation for the association between this SNP and PAH outcomes.
Collapse
Affiliation(s)
- Neha Hafeez
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Anna Kirillova
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yunshan Yue
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
- School of MedicineTsinghua UniversityBeijingChina
| | - Rashmi J. Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Neil J. Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yi‐Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Adam Handen
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Danli Jiang
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Ting Wu
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Yingze Zhang
- Division of Pulmonary Allergy and Critical Care Medicine, Department of MedicineUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Dennis McNamara
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Tatiana V. Kudryashova
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Dmitry Goncharov
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Thomas Bertero
- Université Côte d’Azur, CNRS, UMR7275, IPMCValbonneFrance
| | - Mehdi Nouraie
- Division of Pulmonary Allergy and Critical Care Medicine, Department of MedicineUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Gang Li
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| |
Collapse
|
16
|
Fayyaz AU, Sabbah MS, Dasari S, Griffiths LG, DuBrock HM, Wang Y, Charlesworth MC, Borlaug BA, Jenkins SM, Edwards WD, Redfield MM. Histologic and proteomic remodeling of the pulmonary veins and arteries in a porcine model of chronic pulmonary venous hypertension. Cardiovasc Res 2023; 119:268-282. [PMID: 35022664 PMCID: PMC10233294 DOI: 10.1093/cvr/cvac005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 11/15/2021] [Accepted: 01/10/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS In heart failure (HF), pulmonary venous hypertension (PVH) produces pulmonary hypertension (PH) with remodeling of pulmonary veins (PV) and arteries (PA). In a porcine PVH model, we performed proteomic-based bioinformatics to investigate unique pathophysiologic mechanisms mediating PA and PV remodeling. METHODS AND RESULTS Large PV were banded (PVH, n = 10) or not (Sham, n = 9) in piglets. At sacrifice, PV and PA were perfusion labelled for vessel-specific histology and proteomics. The PA and PV were separately sampled with laser-capture micro-dissection for mass spectrometry. Pulmonary vascular resistance [Wood Units; 8.6 (95% confidence interval: 6.3, 12.3) vs. 2.0 (1.7, 2.3)] and PA [19.9 (standard error of mean, 1.1) vs. 10.3 (1.1)] and PV [14.2 (1.2) vs. 7.6 (1.1)] wall thickness/external diameter (%) were increased in PVH (P < 0.05 for all). Similar numbers of proteins were identified in PA (2093) and PV (2085) with 94% overlap, but biological processes differed. There were more differentially expressed proteins (287 vs. 161), altered canonical pathways (17 vs. 3), and predicted upstream regulators (PUSR; 22 vs. 6) in PV than PA. In PA and PV, bioinformatics indicated activation of the integrated stress response and mammalian target of rapamycin signalling with dysregulated growth. In PV, there was also activation of Rho/Rho-kinase signalling with decreased actin cytoskeletal signalling and altered tight and adherens junctions, ephrin B, and caveolae-mediated endocytosis signalling; all indicating disrupted endothelial barrier function. Indeed, protein biomarkers and the top PUSR in PV (transforming growth factor-beta) suggested endothelial to mesenchymal transition in PV. Findings were similar in human autopsy specimens. CONCLUSION These findings provide new therapeutic targets to oppose pulmonary vascular remodeling in HF-related PH.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Michael S Sabbah
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Surendra Dasari
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Leigh G Griffiths
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Hilary M DuBrock
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - M Cristine Charlesworth
- Molecular Genome Facility Proteomics Core, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Sarah M Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - William D Edwards
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Margaret M Redfield
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
17
|
DÜZGÜN Z, KAYIKÇIOĞLU M, AKTAN Ç, BARA B, EROĞLU Z, YAĞMUR B, BOZOK ÇETİNTAŞ V, BAYINDIR M, NALBANTGİL S, ı TETİK VARDARLI A. Decreased circulating microRNA-21 and microRNA-143 are associated to pulmonary hypertension. Turk J Med Sci 2023; 53:130-141. [PMID: 36945942 PMCID: PMC10388131 DOI: 10.55730/1300-0144.5566] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 02/22/2023] [Accepted: 10/10/2022] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by maladaptation of pulmonary vasculature which is leading to right ventricular hypertrophy and heart failure. miRNAs play a crucial role in the regulation of many diseases such as viral infection, cancer, cardiovascular diseases, and pulmonary hypertension (PH). In this study, we aimed to investigate the expression pattern of eight human plasma miRNAs (hsa-miR-21-3p, hsa-miR-143- 3p, hsa-miR-138-5p, hsa-miR-145-3p, hsa-miR-190a, hsa-miR-204-3p, hsamiR-206, hsa-miR-210-3p) in mild-to-severe PH patients and healthy controls. METHODS : miRNAs were extracted from the peripheral plasma of the PH patients (n: 44) and healthy individuals (n: 30) by using the miRNA Isolation Kit. cDNA was synthesized using All in-One First strand cDNA Synthesis Kit. Expression of the human plasma hsa-miR- 21-3p, hsa-miR-143-3p, hsa-miR-138-5p, hsa-miR-145-3p, hsa-miR-190a, hsa-miR-204- 3p, hsa-miR-206, hsa-miR210-3p, and miRNAs were analyzed by qRT-PCR. RESULTS According to our results, in PH patients hsa-miR-21-3p and hsa-miR-143-3p expression levels were decreased by 4.7 and 2.3 times, respectively. No significant changes were detected in hsa-miR-138-5p, hsa-miR-145-3p, hsa-miR-190a, hsa-miR-204-3p, hsamiR-206, and hsa-miR-210-3p expression levels between PH and control groups. In addition, considering the severity of the disease, it was observed that the decrease in miR-138, miR-143, miR-145, miR-190, mir-204, mir-206 and miR-208 expressions was significant in patients with severe PH. DISCUSSION : In the early diagnosis of PAH, hsa-miR-21-3p and especially hsa-miR-143-3p in peripheral plasma can be considered as potential biomarkers.
Collapse
Affiliation(s)
- Zekeriya DÜZGÜN
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun,
Turkey
| | - Meral KAYIKÇIOĞLU
- Department of Cardiology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | - Çağdaş AKTAN
- Department of Medical Biology, Beykent University School of Medicine, İstanbul,
Turkey
| | - Busra BARA
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | - Zuhal EROĞLU
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | - Burcu YAĞMUR
- Department of Cardiology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | | | - Melike BAYINDIR
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | - Sanem NALBANTGİL
- Department of Cardiology, Faculty of Medicine, Ege University, İzmir,
Turkey
| | - As ı TETİK VARDARLI
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkey
| |
Collapse
|
18
|
Bernardi N, Bianconi E, Vecchi A, Ameri P. Noncoding RNAs in Pulmonary Arterial Hypertension. Heart Fail Clin 2023; 19:137-152. [DOI: 10.1016/j.hfc.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Garcia SM, Matheson B, Morales-Loredo JH, Jernigan NL, Kanagy NL, Resta TC, Clark RM, Shekarriz R, Gonzalez Bosc LV. Hydrogen sulfide and miR21 are suitable biomarkers of hypoxic exposure. Am J Physiol Regul Integr Comp Physiol 2022; 323:R900-R909. [PMID: 36250874 PMCID: PMC9678419 DOI: 10.1152/ajpregu.00199.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/12/2022] [Accepted: 10/13/2022] [Indexed: 11/22/2022]
Abstract
Hypoxia is the reduction of alveolar partial pressure of oxygen ([Formula: see text]). Military members and people who practice recreational activities from moderate to high altitudes are at risk for hypoxic exposure. Hypoxemia's signs and symptoms vary from asymptomatic to severe responses, such as excessive hypoxic ventilatory responses and residual neurobehavioral impairment. Therefore, it is essential to identify hypoxia-induced biomarkers to indicate people with exposure to hypoxia. Advances have been made in understanding physiological responses to hypoxia, including elevations in circulating levels of endothelin 1 (ET-1) and microRNA 21 (miR-21) and reduction in circulating levels of hydrogen sulfide (H2S). Although the levels of these factors change upon exposure to hypoxia, it is unclear if these changes are sustained on return to normoxia. We hypothesize that hypoxia-induced ET-1 and miR-21 remain elevated, whereas hypoxia-reduction in H2S sustains after returning to normoxic conditions. To test this hypothesis, we exposed male rats to 6 h of 12% O2 and measured circulating levels of ET-1 and miR-21, pre, during, and posthypoxia. We found that ET-1 plasma levels increased in response to hypoxia but returned to normal levels within 30 min after the restoration of normoxia. miR-21 plasma levels and transdermal H2S emissions decreased in response to hypoxia, remaining decreased on return to normoxia, thus following the biomarker criteria. Therefore, this study supports a unique role for plasma miR21 and transdermal H2S as hypoxia biomarkers that could be used to identify individuals after exposure to hypoxia.
Collapse
Affiliation(s)
- Selina M Garcia
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Benjamin Matheson
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Juan H Morales-Loredo
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Thomas C Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ross M Clark
- Department of Surgery, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | | | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| |
Collapse
|
20
|
Galkin A, Sitapara R, Clemons B, Garcia E, Kennedy M, Guimond D, Carter LL, Douthitt A, Osterhout R, Gandjeva A, Slee D, Salter-Cid L, Tuder RM, Zisman LS. Inhaled seralutinib exhibits potent efficacy in models of pulmonary arterial hypertension. Eur Respir J 2022; 60:2102356. [PMID: 35680144 PMCID: PMC9724289 DOI: 10.1183/13993003.02356-2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 05/20/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Signalling through platelet-derived growth factor receptor (PDGFR), colony-stimulating factor 1 receptor (CSF1R) and mast/stem cell growth factor receptor kit (c-KIT) plays a critical role in pulmonary arterial hypertension (PAH). We examined the preclinical efficacy of inhaled seralutinib, a unique small-molecule PDGFR/CSF1R/c-KIT kinase inhibitor in clinical development for PAH, in comparison to a proof-of-concept kinase inhibitor, imatinib. METHODS Seralutinib and imatinib potency and selectivity were compared. Inhaled seralutinib pharmacokinetics/pharmacodynamics were studied in healthy rats. Efficacy was evaluated in two rat models of PAH: SU5416/Hypoxia (SU5416/H) and monocrotaline pneumonectomy (MCTPN). Effects on inflammatory/cytokine signalling were examined. PDGFR, CSF1R and c-KIT immunohistochemistry in rat and human PAH lung samples and microRNA (miRNA) analysis in the SU5416/H model were performed. RESULTS Seralutinib potently inhibited PDGFRα/β, CSF1R and c-KIT. Inhaled seralutinib demonstrated dose-dependent inhibition of lung PDGFR and c-KIT signalling and increased bone morphogenetic protein receptor type 2 (BMPR2). Seralutinib improved cardiopulmonary haemodynamic parameters and reduced small pulmonary artery muscularisation and right ventricle hypertrophy in both models. In the SU5416/H model, seralutinib improved cardiopulmonary haemodynamic parameters, restored lung BMPR2 protein levels and decreased N-terminal pro-brain natriuretic peptide (NT-proBNP), more than imatinib. Quantitative immunohistochemistry in human lung PAH samples demonstrated increased PDGFR, CSF1R and c-KIT. miRNA analysis revealed candidates that could mediate seralutinib effects on BMPR2. CONCLUSIONS Inhaled seralutinib was an effective treatment of severe PAH in two animal models, with improved cardiopulmonary haemodynamic parameters, a reduction in NT-proBNP, reverse remodelling of pulmonary vascular pathology and improvement in inflammatory biomarkers. Seralutinib showed greater efficacy compared to imatinib in a preclinical study.
Collapse
Affiliation(s)
- Anna Galkin
- Gossamer Bio, Inc., San Diego, CA, USA
- A. Galkin and R. Sitapara contributed equally as first authors
| | - Ravikumar Sitapara
- Gossamer Bio, Inc., San Diego, CA, USA
- The Rensselaer Center for Translational Research Inc., Rensselaer, NY, USA
- A. Galkin and R. Sitapara contributed equally as first authors
| | | | | | | | | | | | | | | | - Aneta Gandjeva
- University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Rubin M Tuder
- University of Colorado School of Medicine, Aurora, CO, USA
| | | |
Collapse
|
21
|
Rogula S, Pomirski B, Czyżak N, Eyileten C, Postuła M, Szarpak Ł, Filipiak KJ, Kurzyna M, Jaguszewski M, Mazurek T, Grabowski M, Gąsecka A. Biomarker-based approach to determine etiology and severity of pulmonary hypertension: Focus on microRNA. Front Cardiovasc Med 2022; 9:980718. [PMID: 36277769 PMCID: PMC9582157 DOI: 10.3389/fcvm.2022.980718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by remodeling of the pulmonary arteries, and defined by elevated pulmonary arterial pressure, measured during right heart catheterization. There are three main challenges to the diagnostic and therapeutic process of patients with PAH. First, it is difficult to differentiate particular PAH etiology. Second, invasive diagnostic is required to precisely determine the severity of PAH, and thus to qualify patients for an appropriate treatment. Third, the results of treatment of PAH are unpredictable and remain unsatisfactory. MicroRNAs (miRNAs) are small non-coding RNAs that regulate post transcriptional gene-expression. Their role as a prognostic, and diagnostic biomarkers in many different diseases have been studied in recent years. MiRNAs are promising novel biomarkers in PAH due to their activity in various molecular pathways and processes underlying PAH. Lack of biomarkers to differentiate between particular PAH etiology and evaluate the severity of PAH, as well as paucity of therapeutic targets in PAH open a new field for the possibility to use miRNAs in these applications. In our article, we discuss the potential of miRNAs use as diagnostic tools, prognostic biomarkers and therapeutic targets in PAH.
Collapse
Affiliation(s)
- Sylwester Rogula
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland,*Correspondence: Sylwester Rogula,
| | - Bartosz Pomirski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Norbert Czyżak
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland,Genomics Core Facility, Center of New Technologies (CeNT), University of Warsaw, Warsaw, Poland
| | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Łukasz Szarpak
- Department of Outcomes Research, Maria Skłodowska-Curie Medical Academy in Warsaw, Warsaw, Poland
| | - Krzysztof J. Filipiak
- Institute of Clinical Sciences, Maria Skłodowska-Curie Medical Academy in Warsaw, Warsaw, Poland
| | - Marcin Kurzyna
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre Otwock, Otwock, Poland
| | - Miłosz Jaguszewski
- 1st Department of Cardiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Grabowski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Hu X, Wang Q, Zhao H, Wu W, Zhao Q, Jiang R, Liu J, Wang L, Yuan P. Role of miR-21-5p/FilGAP axis in estradiol alleviating the progression of monocrotaline-induced pulmonary hypertension. Animal Model Exp Med 2022; 5:217-226. [PMID: 35713208 PMCID: PMC9240735 DOI: 10.1002/ame2.12253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background Aberrant expression of microRNAs (miRNAs) has been associated with the pathogenesis of pulmonary hypertension (PH). It is, however, not clear whether miRNAs are involved in estrogen rescue of PH. Methods Fresh plasma samples were prepared from 12 idiopathic pulmonary arterial hypertension (IPAH) patients and 12 healthy controls undergoing right heart catheterization in Shanghai Pulmonary Hospital. From each sample, 5 μg of total RNA was tagged and hybridized on microRNA microarray chips. Monocrotaline‐induced PH (MCT‐PH) male rats were treated with 17β‐estradiol (E2) or vehicle. Subgroups were cotreated with estrogen receptor (ER) antagonist or with antagonist of miRNA. Results Many circulating miRNAs, including miR‐21‐5p and miR‐574‐5p, were markedly expressed in patients and of interest in predicting mean pulmonary arterial pressure elevation in patients. The expression of miR‐21‐5p in the lungs was significantly upregulated in MCT‐PH rats compared with the controls. However, miR‐574‐5p showed no difference in the lungs of MCT‐PH rats and controls. miR‐21‐5p was selected for further analysis in rats as E2 strongly regulated it. E2 decreased miR‐21‐5p expression in the lungs of MCT‐PH rats by ERβ. E2 reversed miR‐21‐5p target gene FilGAP downregulation in the lungs of MCT‐PH rats. The abnormal expression of RhoA, ROCK2, Rac1 and c‐Jun in the lungs of MCT‐PH rats was inhibited by E2 and miR‐21‐5p antagonist. Conclusions miR‐21‐5p level was remarkably associated with PH severity in patients. Moreover, the miR‐21‐5p/FilGAP signaling pathway modulated the protective effect of E2 on MCT‐PH through ERβ.
Collapse
Affiliation(s)
- Xiaoyi Hu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qian Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.,Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Hui Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.,Institute of Bismuth Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Wenhui Wu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qinhua Zhao
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Rong Jiang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jinming Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| |
Collapse
|
23
|
Li Y, Cai H, Wei J, Zhu L, Yao Y, Xie M, Song L, Zhang C, Huang X, Wang L. Dihydroartemisinin Attenuates Hypoxic Pulmonary Hypertension via the Downregulation of miR-335 Targeting Vangl2. DNA Cell Biol 2022; 41:750-767. [PMID: 35862468 DOI: 10.1089/dna.2021.1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dihydroartemisinin (DHA) is a traditional antimalarial drug. DHA plays a crucial role in preventing pulmonary hypertension (PH); however, its regulatory function on microRNAs (miRNAs) in PH remains unclear. This study aimed to investigate whether DHA exerts its protective functions by regulating miR-335 in PH. Hypoxia-induced PH models were induced both in vitro and in vivo. Mice were treated with various concentrations of DHA, and pulmonary arterial smooth muscle cells (PASMCs) were treated with DHA, miR-335 inhibitor, miR-335 mimic, or Van Gogh-like 2 (Vangl2) plasmid. The expression of miR-335 and Vangl2, pulmonary arterial remodeling index; right ventricular hypertrophy index; and proliferation and migration indexes were measured. DHA improved pulmonary vascular remodeling and alleviated PH in vivo. miRNA sequencing and real-time PCR results further show that the increase in hypoxia-induced miR-335 was avoided by DHA administration, and miR-335 increased the hypoxia-induced PASMC proliferation and migration. MiRNA databases and dual-luciferase reporter assay show that miR-335 directly targets Vangl2, and Vangl2 decreased the hypoxia-induced PASMC proliferation and migration. The miR-335 inhibitor failed to inhibit hypoxia-induced proliferation and migration upregulation in Vangl2 knockdown PASMCs, and the effect of DHA can be blocked by miR-335 upregulation. In hypoxic PH, MiR-335 is increased, whereas Vangl2 is decreased. MiR-335 can significantly promote the hypoxia-induced proliferation and migration of PASMCs by targeting the Vangl2 gene. DHA effectively reverses the hypoxia-induced upregulation of miR-335 expression, avoiding the miR-335-mediated downregulation of Vangl2 and thereby promoting the expression of Vangl2 to prevent PH.
Collapse
Affiliation(s)
- Yaozhe Li
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haijian Cai
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinqiu Wei
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lin Zhu
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yizhu Yao
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengyao Xie
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lanlan Song
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chi Zhang
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liangxing Wang
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Chen Y, He Y, Zhao S, He X, Xue D, Xia Y. Hypoxic/Ischemic Inflammation, MicroRNAs and δ-Opioid Receptors: Hypoxia/Ischemia-Sensitive Versus-Insensitive Organs. Front Aging Neurosci 2022; 14:847374. [PMID: 35615595 PMCID: PMC9124822 DOI: 10.3389/fnagi.2022.847374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/21/2022] [Indexed: 11/15/2022] Open
Abstract
Hypoxia and ischemia cause inflammatory injury and critically participate in the pathogenesis of various diseases in various organs. However, the protective strategies against hypoxic and ischemic insults are very limited in clinical settings up to date. It is of utmost importance to improve our understanding of hypoxic/ischemic (H/I) inflammation and find novel therapies for better prevention/treatment of H/I injury. Recent studies provide strong evidence that the expression of microRNAs (miRNAs), which regulate gene expression and affect H/I inflammation through post-transcriptional mechanisms, are differentially altered in response to H/I stress, while δ-opioid receptors (DOR) play a protective role against H/I insults in different organs, including both H/I-sensitive organs (e.g., brain, kidney, and heart) and H/I-insensitive organs (e.g., liver and muscle). Indeed, many studies have demonstrated the crucial role of the DOR-mediated cyto-protection against H/I injury by several molecular pathways, including NLRP3 inflammasome modulated by miRNAs. In this review, we summarize our recent studies along with those of others worldwide, and compare the effects of DOR on H/I expression of miRNAs in H/I-sensitive and -insensitive organs. The alternation in miRNA expression profiles upon DOR activation and the potential impact on inflammatory injury in different organs under normoxic and hypoxic conditions are discussed at molecular and cellular levels. More in-depth investigations into this field may provide novel clues for new protective strategies against H/I inflammation in different types of organs.
Collapse
Affiliation(s)
- Yimeng Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yichen He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Shuchen Zhao
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- *Correspondence: Dong Xue,
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
- Ying Xia,
| |
Collapse
|
25
|
Rhodes CJ, Sweatt AJ, Maron BA. Harnessing Big Data to Advance Treatment and Understanding of Pulmonary Hypertension. Circ Res 2022; 130:1423-1444. [PMID: 35482840 PMCID: PMC9070103 DOI: 10.1161/circresaha.121.319969] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pulmonary hypertension is a complex disease with multiple causes, corresponding to phenotypic heterogeneity and variable therapeutic responses. Advancing understanding of pulmonary hypertension pathogenesis is likely to hinge on integrated methods that leverage data from health records, imaging, novel molecular -omics profiling, and other modalities. In this review, we summarize key data sets generated thus far in the field and describe analytical methods that hold promise for deciphering the molecular mechanisms that underpin pulmonary vascular remodeling, including machine learning, network medicine, and functional genetics. We also detail how genetic and subphenotyping approaches enable earlier diagnosis, refined prognostication, and optimized treatment prediction. We propose strategies that identify functionally important molecular pathways, bolstered by findings across multi-omics platforms, which are well-positioned to individualize drug therapy selection and advance precision medicine in this highly morbid disease.
Collapse
Affiliation(s)
- Christopher J Rhodes
- Department of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (C.J.R.)
| | - Andrew J Sweatt
- Department of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (C.J.R.)
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.A.M.).,Division of Cardiology, VA Boston Healthcare System, West Roxbury, MA (B.A.M.)
| |
Collapse
|
26
|
Lombardo SD, Wangsaputra IF, Menche J, Stevens A. Network Approaches for Charting the Transcriptomic and Epigenetic Landscape of the Developmental Origins of Health and Disease. Genes (Basel) 2022; 13:764. [PMID: 35627149 PMCID: PMC9141211 DOI: 10.3390/genes13050764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
The early developmental phase is of critical importance for human health and disease later in life. To decipher the molecular mechanisms at play, current biomedical research is increasingly relying on large quantities of diverse omics data. The integration and interpretation of the different datasets pose a critical challenge towards the holistic understanding of the complex biological processes that are involved in early development. In this review, we outline the major transcriptomic and epigenetic processes and the respective datasets that are most relevant for studying the periconceptional period. We cover both basic data processing and analysis steps, as well as more advanced data integration methods. A particular focus is given to network-based methods. Finally, we review the medical applications of such integrative analyses.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
| | - Ivan Fernando Wangsaputra
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| | - Jörg Menche
- Max Perutz Labs, Department of Structural and Computational Biology, University of Vienna, 1030 Vienna, Austria;
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1030 Vienna, Austria
- Faculty of Mathematics, University of Vienna, 1030 Vienna, Austria
| | - Adam Stevens
- Maternal and Fetal Health Research Group, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9WL, UK;
| |
Collapse
|
27
|
Song C, Zhang J, Liu Y, Hu Y, Feng C, Shi P, Zhang Y, Wang L, Xie Y, Zhang M, Zhao X, Cao Y, Li C, Sun H. Characterization and Validation of ceRNA-Mediated Pathway–Pathway Crosstalk Networks Across Eight Major Cardiovascular Diseases. Front Cell Dev Biol 2022; 10:762129. [PMID: 35433687 PMCID: PMC9010821 DOI: 10.3389/fcell.2022.762129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 03/01/2022] [Indexed: 01/08/2023] Open
Abstract
Pathway analysis is considered as an important strategy to reveal the underlying mechanisms of diseases. Pathways that are involved in crosstalk can regulate each other and co-regulate downstream biological processes. Furthermore, some genes in the pathways can function with other genes via the relationship of the competing endogenous RNA (ceRNA) mechanism, which has also been demonstrated to play key roles in cellular biology. However, the comprehensive analysis of ceRNA-mediated pathway crosstalk is lacking. Here, we constructed the landscape of the ceRNA-mediated pathway–pathway crosstalk of eight major cardiovascular diseases (CVDs) based on sequencing data from ∼2,800 samples. Some common features shared by numerous CVDs were uncovered. A fraction of the pathway–pathway crosstalk was conserved in multiple CVDs and a core pathway–pathway crosstalk network was identified, suggesting the similarity of pathway–pathway crosstalk among CVDs. Experimental evidence also demonstrated that the pathway crosstalk was functioned in CVDs. We split all hub pathways of each pathway–pathway crosstalk network into three categories, namely, common hubs, differential hubs, and specific hubs, which could highlight the common or specific biological mechanisms. Importantly, after a comparison analysis of the hub pathways of networks, ∼480 hub pathway-induced common modules were identified to exert functions in CVDs broadly. Moreover, we performed a random walk algorithm on the hub pathway-induced sub-network and identified 23 potentially novel CVD-related pathways. In summary, our study revealed the potential molecular regulatory mechanisms of ceRNA crosstalk in pathway–pathway crosstalk levels and provided a novel routine to investigate the pathway–pathway crosstalk in cardiology. All CVD pathway–pathway crosstalks are provided in http://www.licpathway.net/cepathway/index.html.
Collapse
Affiliation(s)
- Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Jian Zhang
- Department of Medical Informatics, Harbin Medical University-Daqing, Daqing, China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Yinling Hu
- Department of Rehabilitation, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, China
| | - Chenchen Feng
- Department of Medical Informatics, Harbin Medical University-Daqing, Daqing, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Yuexin Zhang
- Department of Medical Informatics, Harbin Medical University-Daqing, Daqing, China
| | - Lixin Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Yawen Xie
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Meitian Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Xilong Zhao
- Department of Medical Informatics, Harbin Medical University-Daqing, Daqing, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
| | - Chunquan Li
- Department of Medical Informatics, Harbin Medical University-Daqing, Daqing, China
- *Correspondence: Hongli Sun, ; Chunquan Li,
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, China
- *Correspondence: Hongli Sun, ; Chunquan Li,
| |
Collapse
|
28
|
MicroRNAs in Pulmonary Hypertension, from Pathogenesis to Diagnosis and Treatment. Biomolecules 2022; 12:biom12040496. [PMID: 35454085 PMCID: PMC9031307 DOI: 10.3390/biom12040496] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary hypertension (PH) is a fatal and untreatable disease, ultimately leading to right heart failure and eventually death. microRNAs are small, non-coding endogenous RNA molecules that can regulate gene expression and influence various biological processes. Changes in microRNA expression levels contribute to various cardiovascular disorders, and microRNAs have been shown to play a critical role in PH pathogenesis. In recent years, numerous studies have explored the role of microRNAs in PH, focusing on the expression profiles of microRNAs and their signaling pathways in pulmonary artery smooth muscle cells (PASMCs) or pulmonary artery endothelial cells (PAECs), PH models, and PH patients. Moreover, certain microRNAs, such as miR-150 and miR-26a, have been identified as good candidates of diagnosis biomarkers for PH. However, there are still several challenges for microRNAs as biomarkers, including difficulty in normalization, specificity in PH, and a lack of longitudinal and big sample-sized studies. Furthermore, microRNA target drugs are potential therapeutic agents for PH treatment, which have been demonstrated in PH models and in humans. Nonetheless, synthetic microRNA mimics or antagonists are susceptible to several common defects, such as low drug efficacy, inefficient drug delivery, potential toxicity and especially, off-target effects. Therefore, finding clinically safe and effective microRNA drugs remains a great challenge, and further breakthrough is urgently needed.
Collapse
|
29
|
Dynamic Changes in miR-21 Regulate Right Ventricular Dysfunction in Congenital Heart Disease-Related Pulmonary Arterial Hypertension. Cells 2022; 11:cells11030564. [PMID: 35159373 PMCID: PMC8834169 DOI: 10.3390/cells11030564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
Right ventricular (RV) failure is a major cause of mortality in pulmonary arterial hypertension (PAH), but its mechanism remains largely unknown. MicroRNA-21 (miR-21) is involved in flow-mediated stress in the vasculature, but its effects on RV remodeling require investigations. Herein, we aim to study the mechanism of miR-21 in the early (compensated) and late (decompensated) phases of PAH-induced RV dysfunction. Using aorto-venous fistula (AVS) surgery, we established a rat model of PAH. To mimic the microenvironment of PAH, we treated cardiomyocytes with flow-mediated shear stress in 6 dyne for 3 and 8 h. To evaluate whether miR-21 could be a biomarker, we prospectively collected the sera of patients with congenital heart disease- (CHD) related PAH. Additionally, clinical, echocardiographic and right heart catheterization information was collected. The primary endpoint was hospitalization for decompensated heart failure (HF). It is of note that, despite an initial increase in miR-21 expression in hypertrophic RV post AVS, miR-21 expression decreased with RV dysfunction thereafter. Likewise, the activation of miR-21 in cardiomyocytes under shear stress at 3 h was downregulated at 6 h. The downregulated miR-21 at the late phase was associated with increased apoptosis in cardiomyocytes while miR-21 mimic rescued it. Among 76 CHD-induced PAH patients, 19 who were hospitalized for heart failure represented with a significantly lower expression of circulating miR-21. Collectively, our study revealed that the upregulation of miR-21 in the early phase (RV hypertrophy) and downregulation in the late phase (RV dysfunction) under PAH triggered a biphasic regulation of cardiac remodeling and cardiomyocyte apoptosis.
Collapse
|
30
|
Zhong Y, Zhang Z, Chen X. Inhibition of miR-21 improves pulmonary vascular responses in bronchopulmonary dysplasia by targeting the DDAH1/ADMA/NO pathway. Open Med (Wars) 2022; 17:1949-1964. [PMID: 36561848 PMCID: PMC9743197 DOI: 10.1515/med-2022-0584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 09/07/2022] [Accepted: 10/02/2022] [Indexed: 12/14/2022] Open
Abstract
miR-21 has been confirmed to be overexpressed in neonatal rat lungs with hyperoxia-mediated bronchopulmonary dysplasia (BPD). The specific function of miR-21 in BPD is still unclear. We established the hyperoxia-induced BPD rat model in vivo and the hyperoxia-induced pulmonary microvascular endothelial cells (PMVECs) model in vitro. Transwell assay was utilized to detect the migratory capability of PMVECs. Tube formation assay was utilized to measure angiogenesis ability. ELISA was utilized to test nitric oxide (NO) production and the intracellular and extracellular Asymmetric Dimethylarginine (ADMA) concentration. Furthermore, the interaction between miR-21 and dimethylarginine dimethylaminohydrolase 1 (DDAH1) was evaluated using luciferase reporter assay. We found that miR-21 expression in PMVECs was increased by hyperoxia stimulation. Inhibition of miR-21 improved the migratory and angiogenic activities of PMVECs and overexpression of miR-21 exerted the opposite effects. Furthermore, knockdown of miR-21 increased NO production and decreased intracellular and extracellular ADMA concentration in hyperoxia-treated PMVECs. Next we proved that miR-21 could bind to DDAH1 and negatively regulate its expression. Rescues assays showed that DDAH1 knockdown reversed the effects of miR-21 depletion on hyperoxia-mediated PMVEC functions, NO production, and ADMA concentration. Importantly, miR-21 downregulation restored alveolarization and vascular density in BPD rats. This study demonstrates that inhibition of miR-21 improves pulmonary vascular responses in BPD by targeting the DDAH1/ADMA/NO pathway.
Collapse
Affiliation(s)
- Ying Zhong
- Department of Child Health Care, The First Affiliated Hospital of Nanjing Medical University, 368 Jiangdong North Road, Nanjing 210036, Jiangsu, China
| | - Zhiqun Zhang
- Department of Neonatology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang, China
| | - Xiaoqing Chen
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210036, Jiangsu, China
| |
Collapse
|
31
|
Liu B, Yi D, Pan J, Dai J, Zhu MM, Zhao Y, Oh SP, Fallon MB, Dai Z. Suppression of BMP signaling by PHD2 deficiency in Pulmonary Arterial hypertension. Pulm Circ 2022; 12:e12056. [PMID: 35506101 PMCID: PMC9052986 DOI: 10.1002/pul2.12056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/11/2022] Open
Abstract
BMP signaling deficiency is evident in the lungs of patients with pulmonary arterial hypertension. We demonstrated that PHD2 deficiency suppresses BMP signaling in the lung endothelial cells, suggesting the novel mechanisms of dysregulated BMP signaling in the development of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Bin Liu
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Dan Yi
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Jiakai Pan
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Jingbo Dai
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Maggie M. Zhu
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - You‐Yang Zhao
- Program for Lung and Vascular Biology and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Section for Injury Repair and Regeneration Research, Stanley Manne Children's Research InstituteAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
- Department of Pediatrics, Division of Critical CareNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of PharmacologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Feinberg Cardiovascular and Renal Research InstituteNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - S. Paul Oh
- Department of Neurobiology, Barrow Aneurysm and AVM Research CenterBarrow Neurological InstitutePhoenixArizonaUSA
| | - Michael B. Fallon
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Zhiyu Dai
- Division of Pulmonary, Critical Care and Sleep, Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Department of Internal Medicine, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
- Translational Cardiovascular Research Center, College of Medicine‐PhoenixUniversity of ArizonaPhoenixArizonaUSA
| |
Collapse
|
32
|
Gonzalez-Candia A, Herrera EA. High Altitude Pregnancies and Vascular Dysfunction: Observations From Latin American Studies. Front Physiol 2021; 12:786038. [PMID: 34950057 PMCID: PMC8688922 DOI: 10.3389/fphys.2021.786038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
An estimated human population of 170 million inhabit at high-altitude (HA, above 2,500 m). The potential pathological effects of HA hypobaric hypoxia during gestation have been the focus of several researchers around the world. The studies based on the Himalayan and Central/South American mountains are particularly interesting as these areas account for nearly 70% of the HA world population. At present, studies in human and animal models revealed important alterations in fetal development and growth at HA. Moreover, vascular responses to chronic hypobaria in the pregnant mother and her fetus may induce marked cardiovascular impairments during pregnancy or in the neonatal period. In addition, recent studies have shown potential long-lasting postnatal effects that may increase cardiovascular risk in individuals gestated under chronic hypobaria. Hence, the maternal and fetal adaptive responses to hypoxia, influenced by HA ancestry, are vital for a better developmental and cardiovascular outcome of the offspring. This mini-review exposes and discusses the main determinants of vascular dysfunction due to developmental hypoxia at HA, such as the Andean Mountains, at the maternal and fetal/neonatal levels. Although significant advances have been made from Latin American studies, this area still needs further investigations to reveal the mechanisms involved in vascular dysfunction, to estimate complications of pregnancy and postnatal life adequately, and most importantly, to determine potential treatments to prevent or treat the pathological effects of being developed under chronic hypobaric hypoxia.
Collapse
Affiliation(s)
- Alejandro Gonzalez-Candia
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Emilio A Herrera
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,International Center for Andean Studies (INCAS), Universidad de Chile, Santiago, Chile
| |
Collapse
|
33
|
Zang H, Zhang Q, Li X. Non-Coding RNA Networks in Pulmonary Hypertension. Front Genet 2021; 12:703860. [PMID: 34917122 PMCID: PMC8669616 DOI: 10.3389/fgene.2021.703860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 11/08/2021] [Indexed: 01/12/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are involved in various cellular processes. There are several ncRNA classes, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). The detailed roles of these molecules in pulmonary hypertension (PH) remain unclear. We systematically collected and reviewed reports describing the functions of ncRNAs (miRNAs, lncRNAs, and circRNAs) in PH through database retrieval and manual literature reading. The characteristics of identified articles, especially the experimental methods, were carefully reviewed. Furthermore, regulatory networks were constructed using ncRNAs and their interacting RNAs or genes. These data were extracted from studies on pulmonary arterial smooth muscle cells, pulmonary artery endothelial cells, and pulmonary artery fibroblasts. We included 14 lncRNAs, 1 circRNA, 74 miRNAs, and 110 mRNAs in the constructed networks. Using these networks, herein, we describe the current knowledge on the role of ncRNAs in PH. Moreover, these networks actively provide an improved understanding of the roles of ncRNAs in PH. The results of this study are crucial for the clinical application of ncRNAs.
Collapse
Affiliation(s)
- Hongbin Zang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiongyu Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
34
|
Sánchez-Gloria JL, Carbó R, Buelna-Chontal M, Osorio-Alonso H, Henández-Díazcouder A, de la Fuente-León RL, Sandoval J, Sánchez F, Rubio-Gayosso I, Sánchez-Muñoz F. Cold exposure aggravates pulmonary arterial hypertension through increased miR-146a-5p, miR-155-5p and cytokines TNF-α, IL-1β, and IL-6. Life Sci 2021; 287:120091. [PMID: 34717910 DOI: 10.1016/j.lfs.2021.120091] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cold temperatures can aggravate pulmonary diseases and promote pulmonary arterial hypertension (PAH); however, the underlying mechanism has not been fully explored. AIM To explore the effect of chronic cold exposure on the production of inflammatory cytokines and microRNAs (miRNAs) in a monocrotaline (MCT)-induced PAH model. METHODS Male Sprague Dawley rats were divided into a Control (23.5 ± 2 °C), Cold (5.0 ± 1 °C for ten days), MCT (60 mg/kg body weight i.p.), and MCT + Cold (ten days of cold exposure after 3 weeks of MCT injection). Hemodynamic parameters, right ventricle (RV) hypertrophy, and pulmonary arterial medial wall thickness were determined. IL-1β, IL-6, and TNF-α levels were determined using western blotting. miR-21-5p and -3p, miR-146a-5p and -3p, and miR-155-5p and -3p and plasma extracellular vesicles (EVs) and mRNA expression of Cd68, Cd163, Bmpr2, Smad5, Tgfbr2, and Smad3 were determined using RT-qPCR. RESULTS The MCT + Cold group had aggravated RV hypertrophy hemodynamic parameters, and pulmonary arterial medial wall thickness. In lungs of the MCT + Cold, group the protein levels of TNF-α, IL-1β, and IL-6 were higher than those in the MCT group. The mRNA expression of Cd68 and Cd163 were higher in the MCT + Cold group. miR-146a-5p and miR-155-5p levels were higher in the plasma EVs and lungs of the MCT + Cold group. Cold exposure promoted a greater decrease in miR-21-5p, Bmpr2, Smad5, Tgfbr2, and Smad3 mRNA expression in lungs of the MCT + Cold group. CONCLUSION Cold exposure aggravates MCT-induced PAH with an increase in inflammatory marker and miRNA levels in the plasma EVs and lungs.
Collapse
Affiliation(s)
- José L Sánchez-Gloria
- Sección de Estudios de posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico; Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico
| | - Mabel Buelna-Chontal
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico
| | - Adrián Henández-Díazcouder
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico; Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico city 09340, Mexico
| | | | - Julio Sandoval
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico
| | - Fausto Sánchez
- DPAA, Universidad Autónoma Metropolitana-Xochimilco, Mexico city 04960, Mexico
| | - I Rubio-Gayosso
- Sección de Estudios de posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico
| | - Fausto Sánchez-Muñoz
- Sección de Estudios de posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico city 11340, Mexico; Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico city 14080, Mexico.
| |
Collapse
|
35
|
Li P, Song J, Du H, Lu Y, Dong S, Zhou S, Guo Z, Wu H, Zhao X, Qin Y, Zhu N. MicroRNA-663 prevents monocrotaline-induced pulmonary arterial hypertension by targeting TGF-β1/smad2/3 signaling. J Mol Cell Cardiol 2021; 161:9-22. [PMID: 34339758 DOI: 10.1016/j.yjmcc.2021.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/01/2021] [Accepted: 07/28/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Pulmonary vascular remodeling due to excessive growth factor production and pulmonary artery smooth muscle cells (PASMCs) proliferation is the hallmark feature of pulmonary arterial hypertension (PAH). Recent studies suggest that miR-663 is a potent modulator for tumorigenesis and atherosclerosis. However, whether miR-663 involves in pulmonary vascular remodeling is still unclear. METHODS AND RESULTS By using quantitative RT-PCR, we found that miR-663 was highly expressed in normal human PASMCs. In contrast, circulating level of miR-663 dramatically reduced in PAH patients. In addition, in situ hybridization showed that expression of miR-663 was decreased in pulmonary vasculature of PAH patients. Furthermore, MTT and cell scratch-wound assay showed that transfection of miR-663 mimics significantly inhibited platelet derived growth factor (PDGF)-induced PASMCs proliferation and migration, while knockdown of miR-663 expression enhanced these effects. Mechanistically, dual-luciferase reporter assay revealed that miR-663 directly targets the 3'UTR of TGF-β1. Moreover, western blots and ELISA results showed that miR-663 decreased PDGF-induced TGF-β1 expression and secretion, which in turn suppressed the downstream smad2/3 phosphorylation and collagen I expression. Finally, intratracheal instillation of adeno-miR-663 efficiently inhibited the development of pulmonary vascular remodeling and right ventricular hypertrophy in monocrotaline (MCT)-induced PAH rat models. CONCLUSION These results indicate that miR-663 is a potential biomarker for PAH. MiR-663 decreases PDGF-BB-induced PASMCs proliferation and prevents pulmonary vascular remodeling and right ventricular hypertrophy in MCT-PAH by targeting TGF-β1/smad2/3 signaling. These findings suggest that miR-663 may represent as an attractive approach for the diagnosis and treatment for PAH.
Collapse
Affiliation(s)
- Pan Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jingwen Song
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - He Du
- Department of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Yuwen Lu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Shaohua Dong
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Siwei Zhou
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhifu Guo
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Hong Wu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xianxian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yongwen Qin
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Ni Zhu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
36
|
Xu B, Xu G, Yu Y, Lin J. The role of TGF-β or BMPR2 signaling pathway-related miRNA in pulmonary arterial hypertension and systemic sclerosis. Arthritis Res Ther 2021; 23:288. [PMID: 34819148 PMCID: PMC8613994 DOI: 10.1186/s13075-021-02678-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/07/2021] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe complication of connective tissue disease (CTD), causing death in systemic sclerosis (SSc). The past decade has yielded many scientific insights into microRNA (miRNAs) in PAH and SSc. This growth of knowledge has well-illustrated the complexity of microRNA (miRNA)-based regulation of gene expression in PAH. However, few miRNA-related SSc-PAH were elucidated. This review firstly discusses the role of transforming growth factor-beta (TGF-β) signaling and bone morphogenetic protein receptor type II (BMPR2) in PAH and SSc. Secondly, the miRNAs relating to TGF-β and BMPR2 signaling pathways in PAH and SSc or merely PAH were subsequently summarized. Finally, future studies might develop early diagnostic biomarkers and target-oriented therapeutic strategies for SSc-PAH and PAH treatment.
Collapse
Affiliation(s)
- Bei Xu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Guanhua Xu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Ye Yu
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003
| | - Jin Lin
- Department of Rheumatology, The First Affiliated Hospital, Zhejiang University School of Medicine, #79 Qingchun Road, Hangzhou, Zhejiang Province, People's Republic of China, 310003.
| |
Collapse
|
37
|
An Overview of miRNAs Involved in PASMC Phenotypic Switching in Pulmonary Hypertension. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5765029. [PMID: 34660794 PMCID: PMC8516547 DOI: 10.1155/2021/5765029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/03/2021] [Indexed: 12/31/2022]
Abstract
Pulmonary hypertension (PH) is occult, with no distinctive clinical manifestations and a poor prognosis. Pulmonary vascular remodelling is an important pathological feature in which pulmonary artery smooth muscle cells (PASMCs) phenotypic switching plays a crucial role. MicroRNAs (miRNAs) are a class of evolutionarily highly conserved single-stranded small noncoding RNAs. An increasing number of studies have shown that miRNAs play an important role in the occurrence and development of PH by regulating PASMCs phenotypic switching, which is expected to be a potential target for the prevention and treatment of PH. miRNAs such as miR-221, miR-15b, miR-96, miR-24, miR-23a, miR-9, miR-214, and miR-20a can promote PASMCs phenotypic switching, while such as miR-21, miR-132, miR-449, miR-206, miR-124, miR-30c, miR-140, and the miR-17~92 cluster can inhibit it. The article reviews the research progress on growth factor-related miRNAs and hypoxia-related miRNAs that mediate PASMCs phenotypic switching in PH.
Collapse
|
38
|
Wang R, Loscalzo J. Network module-based drug repositioning for pulmonary arterial hypertension. CPT Pharmacometrics Syst Pharmacol 2021; 10:994-1005. [PMID: 34132494 PMCID: PMC8452304 DOI: 10.1002/psp4.12670] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/12/2021] [Accepted: 05/10/2021] [Indexed: 01/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by pulmonary vascular remodeling leading to increased pulmonary vascular resistance and pulmonary arterial pressure. PAH is a highly morbid cardiopulmonary disease adversely affecting lifespan and quality of life. Despite increased awareness and advances of medical therapies in recent decades, long-term prognosis and survival remain poor for patients with PAH. Novel therapies that can target the underlying pathobiology of PAH and reverse pulmonary vascular remodeling are clearly needed. In this study, we develop a network module-based framework to examine potential drug repositioning for PAH. The rationale for this approach is that in order to have therapeutic effects, the targets of potential drugs must be significantly proximate to the disease module of interest in the human protein-protein interactome. Based on 15 existing drugs for treating PAH, our framework integrates drug-drug interactions, drug-drug chemical similarity, drug targets, and PAH disease proteins into the human interactome, and prioritizes candidate drugs for PAH. We identified 53 drugs that could potentially be repurposed for PAH. Many of these candidates have strong literature support. Compared to black-box-like machine learning models, network module-based drug repositioning can provide mechanistic insights into how repositioned drugs can target the underlying pathobiological mechanisms of PAH.
Collapse
Affiliation(s)
- Rui‐Sheng Wang
- Department of Medicine, Cardiovascular DivisionBrigham and Women’s HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Joseph Loscalzo
- Department of Medicine, Cardiovascular DivisionBrigham and Women’s HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
39
|
Abstract
Rationale: Sex hormones play a role in pulmonary arterial hypertension (PAH), but the menstrual cycle has never been studied.Objectives: We conducted a prospective observational study of eight women with stable PAH and 20 healthy controls over one cycle.Methods: Participants completed four study visits 1 week apart starting on the first day of menstruation. Relationships between sex hormones, hormone metabolites, and extracellular vesicle microRNA (miRNA) expression and clinical markers were compared with generalized linear mixed modeling.Results: Women with PAH had higher but less variable estradiol (E2) levels (P < 0.001) that tracked with 6-minute walk distance (P < 0.001), N-terminal prohormone of brain natriuretic peptide (P = 0.03) levels, and tricuspid annular plane systolic excursion (P < 0.01); the direction of these associations depended on menstrual phase. Dehydroepiandrosterone sulfate (DHEA-S) levels were lower in women with PAH (all visits, P < 0.001). In PAH, each 100-μg/dl increase in DHEA-S was associated with a 127-m increase in 6-minute walk distance (P < 0.001) and was moderated by the cardioprotective E2 metabolite 2-methoxyestrone (P < 0.001). As DHEA-S increased, N-terminal prohormone of brain natriuretic peptide levels decreased (P = 0.001). Expression of extracellular vesicle miRNAs-21, -29c, and -376a was higher in PAH, moderated by E2 and DHEA-S levels, and tracked with hormone-associated changes in clinical measures.Conclusions: Women with PAH have fluctuations in cardiopulmonary function during menstruation driven by E2 and DHEA-S. These hormones in turn influence transcription of extracellular vesicle miRNAs implicated in the pathobiology of pulmonary vascular disease and cancer.
Collapse
|
40
|
Sánchez-Gloria JL, Martínez-Olivares CE, Rojas-Morales P, Hernández-Pando R, Carbó R, Rubio-Gayosso I, Arellano-Buendía AS, Rada KM, Sánchez-Muñoz F, Osorio-Alonso H. Anti-Inflammatory Effect of Allicin Associated with Fibrosis in Pulmonary Arterial Hypertension. Int J Mol Sci 2021; 22:ijms22168600. [PMID: 34445305 PMCID: PMC8395330 DOI: 10.3390/ijms22168600] [Citation(s) in RCA: 18] [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: 06/30/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/27/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling. Recent evidence supports that inflammation plays a key role in triggering and maintaining pulmonary vascular remodeling. Recent studies have shown that garlic extract has protective effects in PAH, but the precise role of allicin, a compound derived from garlic, is unknown. Thus, we used allicin to evaluate its effects on inflammation and fibrosis in PAH. Male Wistar rats were divided into three groups: control (CON), monocrotaline (60 mg/kg) (MCT), and MCT plus allicin (16 mg/kg/oral gavage) (MCT + A). Right ventricle (RV) hypertrophy and pulmonary arterial medial wall thickness were determined. IL-1β, IL-6, TNF-α, NFκB p65, Iκβ, TGF-β, and α-SMA were determined by Western blot analysis. In addition, TNF-α and TGF-β were determined by immunohistochemistry, and miR-21-5p and mRNA expressions of Cd68, Bmpr2, and Smad5 were determined by RT-qPCR. Results: Allicin prevented increases in vessel wall thickness due to TNF-α, IL-6, IL-1β, and Cd68 in the lung. In addition, TGF-β, α-SMA, and fibrosis were lower in the MCT + A group compared with the MCT group. In the RV, allicin prevented increases in TNF-α, IL-6, and TGF-β. These observations suggest that, through the modulation of proinflammatory and profibrotic markers in the lung and heart, allicin delays the progression of PAH.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Cytokines/genetics
- Cytokines/metabolism
- Disulfides/therapeutic use
- Fibrosis
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Hypertension, Pulmonary/drug therapy
- Male
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Rats
- Rats, Wistar
- Smad5 Protein/genetics
- Smad5 Protein/metabolism
- Sulfinic Acids/therapeutic use
Collapse
Affiliation(s)
- José L. Sánchez-Gloria
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.S.-G.); (I.R.-G.)
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Constanza Estefanía Martínez-Olivares
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Pedro Rojas-Morales
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (P.R.-M.); (A.S.A.-B.)
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, Mexico City 14080, Mexico; (C.E.M.-O.); (R.H.-P.)
| | - Roxana Carbó
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Ivan Rubio-Gayosso
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.S.-G.); (I.R.-G.)
| | - Abraham S. Arellano-Buendía
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (P.R.-M.); (A.S.A.-B.)
| | - Karla M. Rada
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Fausto Sánchez-Muñoz
- Sección de Estudios de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (J.L.S.-G.); (I.R.-G.)
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
- Correspondence: (F.S.-M.); (H.O.-A.)
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (P.R.-M.); (A.S.A.-B.)
- Correspondence: (F.S.-M.); (H.O.-A.)
| |
Collapse
|
41
|
Padmasekar M, Savai R, Seeger W, Pullamsetti SS. Exposomes to Exosomes: Exosomes as Tools to Study Epigenetic Adaptive Mechanisms in High-Altitude Humans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:8280. [PMID: 34444030 PMCID: PMC8392481 DOI: 10.3390/ijerph18168280] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 12/29/2022]
Abstract
Humans on earth inhabit a wide range of environmental conditions and some environments are more challenging for human survival than others. However, many living beings, including humans, have developed adaptive mechanisms to live in such inhospitable, harsh environments. Among different difficult environments, high-altitude living is especially demanding because of diminished partial pressure of oxygen and resulting chronic hypobaric hypoxia. This results in poor blood oxygenation and reduces aerobic oxidative respiration in the mitochondria, leading to increased reactive oxygen species generation and activation of hypoxia-inducible gene expression. Genetic mechanisms in the adaptation to high altitude is well-studied, but there are only limited studies regarding the role of epigenetic mechanisms. The purpose of this review is to understand the epigenetic mechanisms behind high-altitude adaptive and maladaptive phenotypes. Hypobaric hypoxia is a form of cellular hypoxia, which is similar to the one suffered by critically-ill hypoxemia patients. Thus, understanding the adaptive epigenetic signals operating in in high-altitude adjusted indigenous populations may help in therapeutically modulating signaling pathways in hypoxemia patients by copying the most successful epigenotype. In addition, we have summarized the current information about exosomes in hypoxia research and prospects to use them as diagnostic tools to study the epigenome of high-altitude adapted healthy or maladapted individuals.
Collapse
Affiliation(s)
- Manju Padmasekar
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
| | - Rajkumar Savai
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60438 Frankfurt am Main, Germany
| | - Werner Seeger
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
| | - Soni Savai Pullamsetti
- Max-Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany; (M.P.); (R.S.); (W.S.)
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Department of Internal Medicine, Justus-Liebig University Giessen, Member of the DZL, Member of CPI, 35392 Giessen, Germany
| |
Collapse
|
42
|
Lopez-Crisosto C, Arias-Carrasco R, Sepulveda P, Garrido-Olivares L, Maracaja-Coutinho V, Verdejo HE, Castro PF, Lavandero S. Novel molecular insights and public omics data in pulmonary hypertension. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166200. [PMID: 34144090 DOI: 10.1016/j.bbadis.2021.166200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension is a rare disease with high morbidity and mortality which mainly affects women of reproductive age. Despite recent advances in understanding the pathogenesis of pulmonary hypertension, the high heterogeneity in the presentation of the disease among different patients makes it difficult to make an accurate diagnosis and to apply this knowledge to effective treatments. Therefore, new studies are required to focus on translational and personalized medicine to overcome the lack of specificity and efficacy of current management. Here, we review the majority of public databases storing 'omics' data of pulmonary hypertension studies, from animal models to human patients. Moreover, we review some of the new molecular mechanisms involved in the pathogenesis of pulmonary hypertension, including non-coding RNAs and the application of 'omics' data to understand this pathology, hoping that these new approaches will provide insights to guide the way to personalized diagnosis and treatment.
Collapse
Affiliation(s)
- Camila Lopez-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile
| | - Raul Arias-Carrasco
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Pablo Sepulveda
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Garrido-Olivares
- Cardiovascular Surgery, Division of Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
43
|
Benincasa G, DeMeo DL, Glass K, Silverman EK, Napoli C. Epigenetics and pulmonary diseases in the horizon of precision medicine: a review. Eur Respir J 2021; 57:13993003.03406-2020. [PMID: 33214212 DOI: 10.1183/13993003.03406-2020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
Epigenetic mechanisms represent potential molecular routes which could bridge the gap between genetic background and environmental risk factors contributing to the pathogenesis of pulmonary diseases. In patients with COPD, asthma and pulmonary arterial hypertension (PAH), there is emerging evidence of aberrant epigenetic marks, mainly including DNA methylation and histone modifications which directly mediate reversible modifications to the DNA without affecting the genomic sequence. Post-translational events and microRNAs can be also regulated epigenetically and potentially participate in disease pathogenesis. Thus, novel pathogenic mechanisms and putative biomarkers may be detectable in peripheral blood, sputum, nasal and buccal swabs or lung tissue. Besides, DNA methylation plays an important role during the early phases of fetal development and may be impacted by environmental exposures, ultimately influencing an individual's susceptibility to COPD, asthma and PAH later in life. With the advances in omics platforms and the application of computational biology tools, modelling the epigenetic variability in a network framework, rather than as single molecular defects, provides insights into the possible molecular pathways underlying the pathogenesis of COPD, asthma and PAH. Epigenetic modifications may have clinical applications as noninvasive biomarkers of pulmonary diseases. Moreover, combining molecular assays with network analysis of epigenomic data may aid in clarifying the multistage transition from a "pre-disease" to "disease" state, with the goal of improving primary prevention of lung diseases and its subsequent clinical management.We describe epigenetic mechanisms known to be associated with pulmonary diseases and discuss how network analysis could improve our understanding of lung diseases.
Collapse
Affiliation(s)
- Giuditta Benincasa
- Dept of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Dawn L DeMeo
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kimberly Glass
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudio Napoli
- Dept of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy .,Clinical Dept of Internal and Specialty Medicine (DAI), University Hospital (AOU), University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
44
|
Oldham WM, Hemnes AR, Aldred MA, Barnard J, Brittain EL, Chan SY, Cheng F, Cho MH, Desai AA, Garcia JGN, Geraci MW, Ghiassian SD, Hall KT, Horn EM, Jain M, Kelly RS, Leopold JA, Lindstrom S, Modena BD, Nichols WC, Rhodes CJ, Sun W, Sweatt AJ, Vanderpool RR, Wilkins MR, Wilmot B, Zamanian RT, Fessel JP, Aggarwal NR, Loscalzo J, Xiao L. NHLBI-CMREF Workshop Report on Pulmonary Vascular Disease Classification: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 77:2040-2052. [PMID: 33888254 PMCID: PMC8065203 DOI: 10.1016/j.jacc.2021.02.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
The National Heart, Lung, and Blood Institute and the Cardiovascular Medical Research and Education Fund held a workshop on the application of pulmonary vascular disease omics data to the understanding, prevention, and treatment of pulmonary vascular disease. Experts in pulmonary vascular disease, omics, and data analytics met to identify knowledge gaps and formulate ideas for future research priorities in pulmonary vascular disease in line with National Heart, Lung, and Blood Institute Strategic Vision goals. The group identified opportunities to develop analytic approaches to multiomic datasets, to identify molecular pathways in pulmonary vascular disease pathobiology, and to link novel phenotypes to meaningful clinical outcomes. The committee suggested support for interdisciplinary research teams to develop and validate analytic methods, a national effort to coordinate biosamples and data, a consortium of preclinical investigators to expedite target evaluation and drug development, longitudinal assessment of molecular biomarkers in clinical trials, and a task force to develop a master clinical trials protocol for pulmonary vascular disease.
Collapse
Affiliation(s)
- William M Oldham
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA.
| | - Anna R Hemnes
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - John Barnard
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Evan L Brittain
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Feixiong Cheng
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michael H Cho
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ankit A Desai
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Mark W Geraci
- Department of Medicine, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | - Kathryn T Hall
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Evelyn M Horn
- Weill Cornell Medical Center, New York, New York, USA
| | - Mohit Jain
- University of California at San Diego, San Diego, California, USA
| | - Rachel S Kelly
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jane A Leopold
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - William C Nichols
- Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Andrew J Sweatt
- Stanford University School of Medicine, Stanford, California, USA
| | - Rebecca R Vanderpool
- Department of Medicine, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | - Beth Wilmot
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging and the School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Roham T Zamanian
- Stanford University School of Medicine, Stanford, California, USA
| | - Joshua P Fessel
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Neil R Aggarwal
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lei Xiao
- Division of Lung Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
45
|
Tan C, Jia F, Zhang P, Sun X, Qiao Y, Chen X, Wang Y, Chen J, Lei Y. A miRNA stabilizing polydopamine nano-platform for intraocular delivery of miR-21-5p in glaucoma therapy. J Mater Chem B 2021; 9:3335-3345. [PMID: 33881417 DOI: 10.1039/d0tb02881a] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The elevation of intraocular pressure (IOP) is an important risk factor in the development of primary open angle glaucoma (POAG), which is the main cause of irreversible vision loss. miRNAs are promising new anti-glaucoma therapeutic agents. However, the low stability and cellular transfection of miRNA in vivo hinder its further application. This study aims to investigate the use of polydopamine-polyethylenimine nanoparticles (PDA/PEI NPs) as miRNA carriers in the treatment of ocular hypertension and glaucoma. The in vitro study proves that the carrier preserves the activity of nucleic acid for a long period. Besides, it has comparable transfection efficiency with commercially available vehicles, while having lower cytotoxicity. It has been demonstrated in the animal model that PDA/PEI NPs successfully reach the target tissues without an obvious inflammatory response. PDA/PEI NPs/miR-21-5p increases the permeability of porcine angular aqueous plexus cells, thereby reducing IOP by facilitating the conventional outflow pathway at least partially through the pathway involving endothelial nitric oxide synthase. Our results indicate that PDA/PEI NPs/miR-21-5p is a promising anti-glaucoma drug for treating POAG. And the delivery strategy may be extended to other gene therapy in treating intraocular diseases.
Collapse
Affiliation(s)
- Chen Tan
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wuttge DM, Carlsen AL, Teku G, Wildt M, Rådegran G, Vihinen M, Heegaard NHH, Hesselstrand R. Circulating Plasma microRNAs In Systemic Sclerosis-Associated Pulmonary Arterial Hypertension. Rheumatology (Oxford) 2021; 61:309-318. [PMID: 33784391 PMCID: PMC8742821 DOI: 10.1093/rheumatology/keab300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Objectives SSc-associated pulmonary arterial hypertension (SSc-APAH) is a late but devastating complication of SSc. Early identification of SSc-APAH may improve survival. We examined the role of circulating miRNAs in SSc-APAH. Methods Using quantitative RT-PCR the abundance of mature miRNAs in plasma was determined in 85 female patients with ACA-positive lcSSc. Twenty-two of the patients had SSc-APAH. Sixty-three SSc controls without PAH were matched for disease duration. Forty-six selected miRNA plasma levels were correlated with clinical data. Longitudinal samples were analysed from 14 SSc-APAH and 27 SSc patients. Results The disease duration was 12 years for the SSc-APAH patients and 12.7 years for the SSc controls. Plasma expression levels of 11 miRNAs were lower in patients with SSc-APAH. Four miRNAs displayed higher plasma levels in SSc-APAH patients compared with SSc controls. There was significant difference between groups for miR-20a-5p and miR-203a-3p when correcting for multiple comparisons (P = 0.002 for both). Receiver operating characteristics curve showed AUC = 0.69–0.83 for miR-21-5p and miR-20a-5p or their combination. miR-20a-5p and miR-203a-3p correlated inversely with NT-pro-Brain Natriuretic Protein levels (r = −0.42 and −0.47). Mixed effect model analysis could not identify any miRNAs as predictor of PAH development. However, miR-20a-5p plasma levels were lower in the longitudinal samples of SSc-APAH patients than in the SSc controls. Conclusions Our study links expression levels of the circulating plasma miRNAs, especially miR-20a-5p and miR-203a-3p, to the occurrence of SSc-APAH in female patients with ACA-positive lcSSc.
Collapse
Affiliation(s)
- Dirk M Wuttge
- Department of Clinical Sciences Lund, Rheumatology, Lund University and Skåne University Hospital, SE-22185, Sweden, Lund
| | - Anting L Carlsen
- Department of Autoimmunology & Biomarkers, Statens Serum Institut, Copenhagen S, DK-2300, Denmark
| | - Gabriel Teku
- Department of Experimental Medical Science, Protein Structure Bioinformatics, Lund University, Lund, SE-22185, Sweden
| | - Marie Wildt
- Department of Clinical Sciences Lund, Rheumatology, Lund University and Skåne University Hospital, SE-22185, Sweden, Lund
| | - Göran Rådegran
- Department of Clinical Sciences Lund, Cardiology, Lund University and the Hemodynamic Lab, the Section for Heart failure and Valvular Disease, VO Heart and Lung Medicine, Skåne University Hospital, Lund, SE-22185, Sweden
| | - Mauno Vihinen
- Department of Experimental Medical Science, Protein Structure Bioinformatics, Lund University, Lund, SE-22185, Sweden
| | - Niels H H Heegaard
- Department of Autoimmunology & Biomarkers, Statens Serum Institut, Copenhagen S, DK-2300, Denmark.,Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, DK-5000, Denmark.,Institute of Clinical Research, Clinical Biochemistry, University of Southern Denmark, DK-5000, Odense, Denmark
| | - Roger Hesselstrand
- Department of Clinical Sciences Lund, Rheumatology, Lund University and Skåne University Hospital, SE-22185, Sweden, Lund
| |
Collapse
|
47
|
Chang WT, Fisch S, Dangwal S, Mohebali J, Fiedler AG, Chen M, Hsu CH, Yang Y, Qiu Y, Alexander KM, Chen FY, Liao R. MicroRNA-21 regulates right ventricular remodeling secondary to pulmonary arterial pressure overload. J Mol Cell Cardiol 2021; 154:106-114. [PMID: 33548242 DOI: 10.1016/j.yjmcc.2021.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 12/25/2022]
Abstract
Right ventricular (RV) function is a critical determinant of survival in patients with pulmonary arterial hypertension (PAH). While miR-21 is known to associate with vascular remodeling in small animal models of PAH, its role in RV remodeling in large animal models has not been characterized. Herein, we investigated the role of miR-21 in RV dysfunction using a sheep model of PAH secondary to pulmonary arterial constriction (PAC). RV structural and functional remodeling were examined using ultrasound imaging. Our results showed that post PAC, RV strain significantly decreased at the basal region compared with t the control. Moreover, such dysfunction was accompanied by increases in miR-21 levels. To determine the role of miR-21 in RV remodeling secondary to PAC, we investigated the molecular alteration secondary to phenylephrine induced hypertrophy and miR21 overexpression in vitro using neonatal rat ventricular myocytes (NRVMs). We found that overexpression of miR-21 in the setting of hypertrophic stimulation augmented only the expression of proteins critical for mitosis but not cytokinesis. Strikingly, this molecular alteration was associated with an eccentric cellular hypertrophic phenotype similar to what we observed in vivo PAC animal model in sheep. Importantly, this hypertrophic change was diminished upon suppressing miR-21 in NRVMs. Collectively, our in vitro and in vivo data demonstrate that miR-21 is a critical contributor in the development of RV dysfunction and could represent a novel therapeutic target for PAH associated RV dysfunction.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan; Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| | - Sudeshna Fisch
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Seema Dangwal
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA, United States of America
| | - Jahan Mohebali
- Division of Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, United States of America; Division of Vascular and Endovascular Surgery, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Amy G Fiedler
- Division of Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Michael Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Hsin Hsu
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA, United States of America; Department of Intensive Care Medicine, Cheng Kung University Hospital, Tainan, Taiwan
| | - Yanfei Yang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Yiling Qiu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Kevin M Alexander
- Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA, United States of America
| | - Frederick Y Chen
- Division of Cardiac Surgery, Cardiovascular Center, Tufts Medical Center, Boston, MA, United States of America
| | - Ronglih Liao
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America; Stanford Cardiovascular Institute, Department of Medicine, Stanford University School of Medicine, CA, United States of America.
| |
Collapse
|
48
|
Abstract
Advances in high-throughput biotechnologies have facilitated omics profiling, a key component of precision phenotyping, in patients with pulmonary vascular disease. Omics provides comprehensive information pertaining to genes, transcripts, proteins, and metabolites. The resulting omics big datasets may be integrated for more robust results and are amenable to analysis using machine learning or newer analytical methodologies, such as network analysis. Results from fully integrated multi-omics datasets combined with clinical data are poised to provide novel insight into pulmonary vascular disease as well as diagnose the presence of disease and prognosticate outcomes.
Collapse
Affiliation(s)
- Jane A Leopold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB0630K, Boston, MA 02115, USA.
| | - Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232, USA
| |
Collapse
|
49
|
Dannewitz Prosseda S, Ali MK, Spiekerkoetter E. Novel Advances in Modifying BMPR2 Signaling in PAH. Genes (Basel) 2020; 12:genes12010008. [PMID: 33374819 PMCID: PMC7824173 DOI: 10.3390/genes12010008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is a disease of the pulmonary arteries, that is characterized by progressive narrowing of the pulmonary arterial lumen and increased pulmonary vascular resistance, ultimately leading to right ventricular dysfunction, heart failure and premature death. Current treatments mainly target pulmonary vasodilation and leave the progressive vascular remodeling unchecked resulting in persistent high morbidity and mortality in PAH even with treatment. Therefore, novel therapeutic strategies are urgently needed. Loss of function mutations of the Bone Morphogenetic Protein Receptor 2 (BMPR2) are the most common genetic factor in hereditary forms of PAH, suggesting that the BMPR2 pathway is fundamentally important in the pathogenesis. Dysfunctional BMPR2 signaling recapitulates the cellular abnormalities in PAH as well as the pathobiology in experimental pulmonary hypertension (PH). Approaches to restore BMPR2 signaling by increasing the expression of BMPR2 or its downstream signaling targets are currently actively explored as novel ways to prevent and improve experimental PH as well as PAH in patients. Here, we summarize existing as well as novel potential treatment strategies for PAH that activate the BMPR2 receptor pharmaceutically or genetically, increase the receptor availability at the cell surface, or reconstitute downstream BMPR2 signaling.
Collapse
Affiliation(s)
- Svenja Dannewitz Prosseda
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (S.D.P.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford, CA 94305, USA
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs University Freiburg, 79104 Freiburg, Germany
| | - Md Khadem Ali
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (S.D.P.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford, CA 94305, USA
| | - Edda Spiekerkoetter
- Division Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; (S.D.P.); (M.K.A.)
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford, CA 94305, USA
- Correspondence:
| |
Collapse
|
50
|
Yun E, Kook Y, Yoo KH, Kim KI, Lee MS, Kim J, Lee A. Endothelial to Mesenchymal Transition in Pulmonary Vascular Diseases. Biomedicines 2020; 8:biomedicines8120639. [PMID: 33371458 PMCID: PMC7767472 DOI: 10.3390/biomedicines8120639] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Lung diseases, such as pulmonary hypertension and pulmonary fibrosis, are life-threatening diseases and have common features of vascular remodeling. During progression, extracellular matrix protein deposition and dysregulation of proteolytic enzymes occurs, which results in vascular stiffness and dysfunction. Although vasodilators or anti-fibrotic therapy have been mainly used as therapy owing to these characteristics, their effectiveness does not meet expectations. Therefore, a better understanding of the etiology and new therapeutic approaches are needed. Endothelial cells (ECs) line the inner walls of blood vessels and maintain vascular homeostasis by protecting vascular cells from pathological stimuli. Chronic stimulation of ECs by various factors, including pro-inflammatory cytokines and hypoxia, leads to ECs undergoing an imbalance of endothelial homeostasis, which results in endothelial dysfunction and is closely associated with vascular diseases. Emerging studies suggest that endothelial to mesenchymal transition (EndMT) contributes to endothelial dysfunction and plays a key role in the pathogenesis of vascular diseases. EndMT is a process by which ECs lose their markers and show mesenchymal-like morphological changes, and gain mesenchymal cell markers. Despite the efforts to elucidate these molecular mechanisms, the role of EndMT in the pathogenesis of lung disease still requires further investigation. Here, we review the importance of EndMT in the pathogenesis of pulmonary vascular diseases and discuss various signaling pathways and mediators involved in the EndMT process. Furthermore, we will provide insight into the therapeutic potential of targeting EndMT.
Collapse
Affiliation(s)
- Eunsik Yun
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
| | - Yunjin Kook
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
| | - Kyung Hyun Yoo
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Keun Il Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Myeong-Sok Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul 04310, Korea
- Correspondence: (J.K.); (A.L.); Tel.: +82-2-710-9553 (J.K. & A.L.); Fax: +82-2-2077-7322 (J.K. & A.L.)
| | - Aram Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea; (E.Y.); (Y.K.); (K.H.Y.); (K.I.K.); (M.-S.L.)
- Correspondence: (J.K.); (A.L.); Tel.: +82-2-710-9553 (J.K. & A.L.); Fax: +82-2-2077-7322 (J.K. & A.L.)
| |
Collapse
|