1
|
Ribatti D. Two critical events in the development of the vascular system: pruning and remodeling. Clin Exp Med 2025; 25:181. [PMID: 40434576 PMCID: PMC12119727 DOI: 10.1007/s10238-025-01720-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025]
Abstract
The cardiovascular system is the first functional organ system to develop in the vertebrate embryo. The primitive heart and primitive vascular plexus are formed through vasculogenesis and thereafter through angiogenesis. After the establishment of new connections within the vascular network, a pruning and remodeling process occurs after which some branches are stabilized, whereas others regress. The aim of this article is to describe the most common pruning and remodeling forms, which take place during organogenesis in the experimental and human systems.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari, Bari, Italy.
| |
Collapse
|
2
|
Amirrad F, La V, Ohadi S, Albotaif M, Webster S, Pru JK, Shamloo K, Mohieldin AM, Nauli SM. PGRMC2 is a pressure-volume regulator critical for myocardial responses to stress in mice. Nat Commun 2025; 16:2422. [PMID: 40069180 PMCID: PMC11897200 DOI: 10.1038/s41467-025-57707-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 03/01/2025] [Indexed: 03/14/2025] Open
Abstract
Progesterone receptors are classified into nuclear and membrane-bound receptor families. Previous unbiased proteomic studies indicate a potential association between cardiac diseases and the progesterone receptor membrane-bound component-2 (PGRMC2); however, the role of PGRMC2 in the heart remains unknown. In this study, we use a heart-specific knockout (KO) mouse model (MyH6•Pgrmc2flox/flox) in which the Pgrmc2 gene was selectively deleted in cardiomyocytes. Here we show that PGRMC2 serves as a mediator of steroid hormones for rapid calcium signaling in cardiomyocytes to maintain cardiac contraction, sufficient stroke volume, and adequate cardiac output by regulating the cardiac pressure-volume relationship. The KO hearts from male and female mice exhibit an impairment in pressure-volume relationship. Under hypoxic conditions, this pressure-volume dysregulation progresses to congestive left and right ventricular failure in the KO hearts. Overall, we propose that PGRMC2 is a cardiac pressure-volume regulator to maintain normal cardiac physiology, especially during hypoxic stress.
Collapse
Affiliation(s)
- Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
- Department of Pharmaceutical Sciences, Marshall B. Ketchum University, Fullerton, USA
| | - Vivian La
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Sharareh Ohadi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Miram Albotaif
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Sha Webster
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - James K Pru
- Department of Animal Sciences, University of Wyoming, Laramie, WY, USA
| | - Kiumars Shamloo
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Ashraf M Mohieldin
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
- College of Graduate Studies, California Northstate University, Elk Grove, CA, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA.
- Department of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
3
|
Farha S, Madden E, Trotter D, Zlojutro V, Kirkness JP, Fouras A, Erzurum S, Asosingh K. Quantification of the pulmonary vasculature in mice under chronic hypoxia with contrast-free pulmonary angiography in vivo imaging. J Appl Physiol (1985) 2025; 138:318-325. [PMID: 39717944 DOI: 10.1152/japplphysiol.00279.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Innovative advancements in preclinical imaging have led to the development of cone-beam computed tomography (CBCT) combined with contrast-free pulmonary angiography (CFPA), a novel lung scanning technology capable of assessing lung function and pulmonary vascular morphology. This cutting-edge approach integrates CBCT to provide detailed quantification of the pulmonary vascular tree. The application of this technique to image and quantify changes in the pulmonary vascular tree of mice exposed to chronic hypoxia has not been investigated. In this study, we investigated the feasibility of utilizing CFPA for imaging changes in the murine lung vascular bed under chronic hypoxia and assessed whether vascular metrics correlate with hematologic parameters and/or right ventricular pressure and mass. Our results revealed a significant increase in hemoglobin and total pulmonary vascular blood volume, as well as total pulmonary vessel length following exposure to chronic hypoxia. The pulmonary vascular blood volume and total vessel length strongly correlated with hemoglobin. There was also an increase in pulmonary arterial pressure and right ventricular mass under hypoxia that was linked to the hematological response and the changes in the pulmonary vascular bed. These findings highlight the application of preclinical CBCT and CFPA imaging as a valuable tool for visual and quantitative analysis of the pulmonary vasculature in preclinical models of chronic hypoxia and its potential use in investigating other pulmonary vasculopathies.NEW & NOTEWORTHY Cone-beam computed tomography (CBCT) combined with contrast-free pulmonary angiography (CFPA) is a novel lung scanning technology capable of assessing pulmonary vascular morphology in vivo in murine models.
Collapse
Affiliation(s)
- Samar Farha
- Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio, United States
| | - Evan Madden
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Dylan Trotter
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Violetta Zlojutro
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Jason P Kirkness
- Miller School of Medicine, University of Miami, Miami, Florida, United States
| | | | - Serpil Erzurum
- Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio, United States
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Kewal Asosingh
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
4
|
Cui C, Chen S, Mi B, Qi Y, Jiao C, Zhang M, Dai Y, Wang X, Hu J, Shi B, Wang J, Zhao Z, Liu X, Zhang X. Transcriptomic and Metabolomic Insights into Age-Related Changes in Lung Tissue of Yaks Under Highland Stress. Int J Mol Sci 2024; 25:12071. [PMID: 39596139 PMCID: PMC11593661 DOI: 10.3390/ijms252212071] [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: 10/09/2024] [Revised: 11/07/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
As an indigenous species on the Tibetan Plateau, the yak is well adapted to the plateau hypoxic environment. The high-altitude hypoxia adaptation of the yak requires the adaptive reshaping of multiple tissues and organs, especially the lungs. To reveal the adaptive development of yak lungs under hypoxic stress at the tissue and molecular levels, we conducted histomorphological observations as well as transcriptomic and metabolomic studies of yak lungs at three ages (0.5, 2.5, and 4.5 years). The results showed that the lung tissue developed significantly with age. The mean alveolar area was higher (p < 0.01) in 4.5 and 2.5-year-old yaks than in 0.5-year-old yaks. The percentage of elastic fibers, micro-arterial wall thickness, and micro-arterial area showed an increasing trend (p < 0.01) from 0.5-year-old yaks to 2.5-year-old yaks and then to 4.5-year-old yaks. In addition, some critical differentially expressed genes related to angiogenesis (MYC, EPHA2, TNF), fiber formation (EREG), smooth muscle proliferation (HBEGF), erythropoiesis (SOCS3), and hypoxia response (ZFP36) were identified. Some metabolites associated with these genes were also found simultaneously. These findings provide a deeper understanding of the molecular strategies underlying this species' extraordinary ability to survive normally in low-oxygen environments. In conclusion, the lungs of yaks undergo continuous adaptive development under hypoxic stress, and these findings are crucial for understanding the molecular mechanisms by which native species of the Tibetan Plateau survive in harsh environments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (C.C.); (S.C.); (B.M.); (Y.Q.); (C.J.); (M.Z.); (Y.D.); (X.W.); (J.W.); (Z.Z.); (X.L.); (X.Z.)
| | - Bingang Shi
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (C.C.); (S.C.); (B.M.); (Y.Q.); (C.J.); (M.Z.); (Y.D.); (X.W.); (J.W.); (Z.Z.); (X.L.); (X.Z.)
| | | | | | | | | |
Collapse
|
5
|
Balcan B, Akdeniz B, Peker Y, Collaborators TTURCOSACT. Obstructive Sleep Apnea and Pulmonary Hypertension: A Chicken-and-Egg Relationship. J Clin Med 2024; 13:2961. [PMID: 38792502 PMCID: PMC11122166 DOI: 10.3390/jcm13102961] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Obstructive sleep apnea (OSA) is characterized by repeated episodes of upper airway obstruction during sleep, and it is closely linked to several cardiovascular issues due to intermittent hypoxia, nocturnal hypoxemia, and disrupted sleep patterns. Pulmonary hypertension (PH), identified by elevated pulmonary arterial pressure, shares a complex interplay with OSA, contributing to cardiovascular complications and morbidity. The prevalence of OSA is alarmingly high, with studies indicating rates of 20-30% in males and 10-15% in females, escalating significantly with age and obesity. OSA's impact on cardiovascular health is profound, particularly in exacerbating conditions like systemic hypertension and heart failure. The pivotal role of hypoxemia increases intrathoracic pressure, inflammation, and autonomic nervous system dysregulation in this interplay, which all contribute to PH's pathogenesis. The prevalence of PH among OSA patients varies widely, with studies reporting rates from 15% to 80%, highlighting the variability in diagnostic criteria and methodologies. Conversely, OSA prevalence among PH patients also remains high, often exceeding 25%, stressing the need for careful screening and diagnosis. Treatment strategies like continuous positive airway pressure (CPAP) therapy show promise in mitigating PH progression in OSA patients. However, this review underscores the need for further research into long-term outcomes and the efficacy of these treatments. This review provides comprehensive insights into the epidemiology, pathophysiology, and treatment of the intricate interplay between OSA and PH, calling for integrated, personalized approaches in diagnosis and management. The future landscape of OSA and PH management hinges on continued research, technological advancements, and a holistic approach to improving patient outcomes.
Collapse
Affiliation(s)
- Baran Balcan
- Department of Pulmonary Medicine, Koç University School of Medicine, Istanbul 34450, Turkey;
| | - Bahri Akdeniz
- Department of Cardiology, Dokuz Eylül University Faculty of Medicine, Izmir 35340, Turkey;
| | - Yüksel Peker
- Department of Pulmonary Medicine, Koç University School of Medicine, Istanbul 34450, Turkey;
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Clinical Sciences, Respiratory Medicine and Allergology, Faculty of Medicine, Lund University, 22185 Lund, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden
| | | |
Collapse
|
6
|
Kostyunina DS, Pakhomov NV, Jouida A, Dillon E, Baugh JA, McLoughlin P. Transcriptomics and proteomics revealed sex differences in human pulmonary microvascular endothelial cells. Physiol Genomics 2024; 56:194-220. [PMID: 38047313 DOI: 10.1152/physiolgenomics.00051.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
Marked sexual dimorphism is displayed in the onset and progression of pulmonary hypertension (PH). Females more commonly develop pulmonary arterial hypertension, yet females with pulmonary arterial hypertension and other types of PH have better survival than males. Pulmonary microvascular endothelial cells play a crucial role in pulmonary vascular remodeling and increased pulmonary vascular resistance in PH. Given this background, we hypothesized that there are sex differences in the pulmonary microvascular endothelium basally and in response to hypoxia that are independent of the sex hormone environment. Human pulmonary microvascular endothelial cells (HPMECs) from healthy male and female donors, cultured under physiological shear stress, were analyzed using RNA sequencing and label-free quantitative proteomics. Gene set enrichment analysis identified a number of sex-different pathways in both normoxia and hypoxia, including pathways that regulate cell proliferation. In vitro, the rate of proliferation in female HPMECs was lower than in male HPMECs, a finding that supports the omics results. Interestingly, thrombospondin-1, an inhibitor of proliferation, was more highly expressed in female cells than in male cells. These results demonstrate, for the first time, important differences between female and male HPMECs that persist in the absence of sex hormone differences and identify novel pathways for further investigation that may contribute to sexual dimorphism in pulmonary hypertensive diseases.NEW & NOTEWORTHY There is marked sexual dimorphism in the development and progression of pulmonary hypertension. We show differences in RNA and protein expression between female and male human pulmonary microvascular endothelial cells grown under conditions of physiological shear stress, which identify sex-different cellular pathways both in normoxia and hypoxia. Importantly, these differences were detected in the absence of sex hormone differences. The pathways identified may provide novel targets for the development of sex-specific therapies.
Collapse
Affiliation(s)
- Daria S Kostyunina
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Nikolai V Pakhomov
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Amina Jouida
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Eugene Dillon
- Conway Institute, University College Dublin, Dublin, Ireland
| | - John A Baugh
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Akiyama T, Sadahiro T, Yamada Y, Fujita R, Abe Y, Nakano K, Honda S, Ema M, Kubota Y, Sakai S, Hizawa N, Ieda M. Flk1 Deficiency and Hypoxia Synergistically Promote Endothelial Dysfunction, Vascular Remodeling, and Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2023; 43:1668-1683. [PMID: 37534464 DOI: 10.1161/atvbaha.123.319266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/11/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND The mechanisms underlying pulmonary hypertension (PH) remain largely unknown; further, why advanced vascular remodeling preferentially occurs in arterioles is yet to be answered. VEGF (vascular endothelial growth factor) regulates angiogenesis through Flk1 (fetal liver kinase 1) and Flt1 (fms-like tyrosine kinase 1) on endothelial cells (ECs), which may be related to PH pathogenesis. However, spatiotemporal expression patterns of Flk1 and Flt1 in the pulmonary vascular system and the role of endothelial Flk1 in PH development remain poorly understood. METHODS We analyzed multiple reporter mice, including Flk1-GFP (green fluorescent protein) bacterial artificial chromosome transgenic (Tg), Flt1-DsRed bacterial artificial chromosome Tg, and Flk1-GFP/Flt1-DsRed double Tg mice, to determine the spatiotemporal expression of Flk1 and Flt1 in hypoxia-induced PH. We also used Cdh5CreERT2/Flk1f/f/Tomato (Flk1-KO [knockout]) mice to induce EC-specific Flk1 deletion and lineage tracing in chronic hypoxia. RESULTS Flk1 was specifically expressed in the ECs of small pulmonary vessels, including arterioles. Conversely, Flt1 was more broadly expressed in the ECs of large- to small-sized vessels in adult mouse lungs. Intriguingly, Flk1+ ECs were transiently increased in hypoxia with proliferation, whereas Flt1 expression was unchanged. Flk1-KO mice did not exhibit pulmonary vascular remodeling nor PH in normoxia; however, the arteriolar ECs changed to a cuboidal shape with protrusion. In hypoxia, Flk1 deletion exacerbated EC dysfunction and reduced their number via apoptosis. Additionally, Flk1 deletion promoted medial thickening and neointimal formation in arterioles and worsened PH. Mechanistically, lineage tracing revealed that neointimal cells were derived from Flk1-KO ECs. Moreover, RNA sequencing in pulmonary ECs demonstrated that Flk1 deletion and hypoxia synergistically activated multiple pathways, including cell cycle, senescence/apoptosis, and cytokine/growth factor, concomitant with suppression of cell adhesion and angiogenesis, to promote vascular remodeling. CONCLUSIONS Flk1 and Flt1 were differentially expressed in pulmonary ECs. Flk1 deficiency and hypoxia jointly dysregulated arteriolar ECs to promote vascular remodeling. Thus, dysfunction of Flk1+ ECs may contribute to the pathogenesis of advanced vascular remodeling in pulmonary arterioles.
Collapse
Affiliation(s)
- Tatsuya Akiyama
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Department of Respiratory Medicine (T.A., N.H.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Taketaro Sadahiro
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yu Yamada
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Ryo Fujita
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Division of Regenerative Medicine, Transborder Medical Research Center (R.F.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yuto Abe
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koji Nakano
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Seiichiro Honda
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masatsugu Ema
- Division of Regenerative Medicine, Transborder Medical Research Center (R.F.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshiaki Kubota
- Departments of Anatomy (Y.K.), Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Sakai
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Faculty of Health Science, Tsukuba University of Technology, Japan (S.S.)
| | - Nobuyuki Hizawa
- Department of Respiratory Medicine (T.A., N.H.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masaki Ieda
- Department of Cardiology (T.A., T.S., Y.Y., R.F., Y.A., K.N., S.H., S.S., M.I.), Institute of Medicine, University of Tsukuba, Ibaraki, Japan
- Cardiology (M.I.), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Ross RL, Mavria G, Del Galdo F, Elies J. Downregulation of Vascular Hemeoxygenase-1 Leads to Vasculopathy in Systemic Sclerosis. Front Physiol 2022; 13:900631. [PMID: 35600300 PMCID: PMC9117635 DOI: 10.3389/fphys.2022.900631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a terminal disease characterized by vasculopathy, tissue fibrosis, and autoimmunity. Although the exact etiology of SSc remains unknown, endothelial dysfunction, oxidative stress, and calcium handling dysregulation have been associated with a large number of SSc-related complications such as neointima formation, vasculogenesis, pulmonary arterial hypertension, impaired angiogenesis, and cardiac arrhythmias. Hemeoxygenase-1 (HO-1) is an antioxidant enzyme involved in multiple biological actions in the cardiovascular system including vascular tone, angiogenesis, cellular proliferation, apoptosis, and oxidative stress. The aim of this work was to investigate the physiological role of HO-1 and its relevance in the cardiovascular complications occurring in SSc. We found that, in early phases of SSc, the expression of HO-1 in dermal fibroblast is lower compared to those isolated from healthy control individuals. This is particularly relevant as reduction of the HO-1/CO signaling pathway is associated with endothelial dysfunction and vasculopathy. We show evidence of the role of HO-1/carbon monoxide (CO) signaling pathway in calcium handling. Using an in vitro model of pulmonary arterial hypertension (PAH) we investigated the role of HO-1 in Ca2+ mobilization from intracellular stores. Our results indicate that HO-1 regulates calcium release from intracellular stores of human pulmonary arterial endothelial cells. We interrogated the activity of HO-1 in angiogenesis using an organotypic co-culture of fibroblast-endothelial cell. Inhibition of HO-1 significantly reduced the ability of endothelial cells to form tubules. We further investigated if this could be associated with cell motility or migration of endothelial cells into the extracellular matrix synthesized by fibroblasts. By mean of holographic imaging, we studied the morphological and functional features of endothelial cells in the presence of an HO-1 activator and selective inhibitors. Our results demonstrate that inhibition of HO-1 significantly reduces cell proliferation and cell motility (migration) of cultured endothelial cells, whilst activation of HO-1 does not modify either morphology, proliferation or motility. In addition, we investigated the actions of CO on the Kv7.1 (KCQN1) channel current, an important component of the cardiac action potential repolarization. Using electrophysiology (whole-cell patch-clamp in a recombinant system overexpressing the KCQN1 channel), we assessed the regulation of KCQN1 by CO. CORM-2, a CO donor, significantly reduced the Kv7.1 current, suggesting that HO-1/CO signaling may play a role in the modulation of the cardiac action potential via regulation of this ion channel. In summary, our results indicate a clear link between: 1) downregulation of HO-1/CO signaling; and 2) pathophysiological processes occurring in early phases of SSc, such as calcium homeostasis dysregulation, impaired angiogenesis and cardiac arrhythmias. A better understanding of the canonical actions (mainly due to the biological actions of CO), and non-canonical actions of HO-1, as well as the interaction of HO-1/CO signaling with other gasotransmitters in SSc will contribute to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Rebecca L Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Georgia Mavria
- Signal Transduction and Tumour Microenvironment Group, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
- Scleroderma Programme, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, United Kingdom
| | - Jacobo Elies
- Cardiovascular Research Group, Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
- *Correspondence: Jacobo Elies,
| |
Collapse
|
9
|
Perez C, Felty Q. Molecular basis of the association between transcription regulators nuclear respiratory factor 1 and inhibitor of DNA binding protein 3 and the development of microvascular lesions. Microvasc Res 2022; 141:104337. [PMID: 35143811 PMCID: PMC8923910 DOI: 10.1016/j.mvr.2022.104337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022]
Abstract
The prognosis of patients with microvascular lesions remains poor because vascular remodeling eventually obliterates the lumen. Here we have focused our efforts on vessel dysfunction in two different organs, the lung and brain. Despite tremendous progress in understanding the importance of blood vessel integrity, gaps remain in our knowledge of the underlying molecular factors contributing to vessel injury, including microvascular lesions. Most of the ongoing research on these lesions have focused on oxidative stress but have not found major molecular targets for the discovery of new treatment or early diagnosis. Herein, we have focused on elucidating the molecular mechanism(s) based on two new emerging molecules NRF1 and ID3, and how they may contribute to microvascular lesions in the lung and brain. Redox sensitive transcriptional activation of target genes depends on not only NRF1, but the recruitment of co-activators such as ID3 to the target gene promoter. Our review highlights the fact that targeting NRF1 and ID3 could be a promising therapeutic approach as they are major players in influencing cell growth, cell repair, senescence, and apoptotic cell death which contribute to vascular lesions. Knowledge about the molecular biology of these processes will be relevant for future therapeutic approaches to not only PAH but cerebral angiopathy and other vascular disorders. Therapies targeting transcription regulators NRF1 or ID3 have the potential for vascular disease-modification because they will address the root causes such as genomic instability and epigenetic changes in vascular lesions. We hope that our findings will serve as a stimulus for further research towards an effective treatment of microvascular lesions.
Collapse
Affiliation(s)
- Christian Perez
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA
| | - Quentin Felty
- Department of Environmental Health Sciences, Florida International University, Miami, FL, USA.
| |
Collapse
|
10
|
Mirzaei Bavil F, Karimi-Sales E, Alihemmati A, Alipour MR. Effect of ghrelin on hypoxia-related cardiac angiogenesis: involvement of miR-210 signalling pathway. Arch Physiol Biochem 2022; 128:270-275. [PMID: 31596148 DOI: 10.1080/13813455.2019.1675712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Hypoxia is the main stimulus for angiogenesis. Hypoxia-inducible factor (HIF)-1α, vascular endothelial growth factor (VEGF), and miR-210 are involved in the hypoxia-induced angiogenesis. This study examined the effects of hypoxia and/or ghrelin on miR-210, HIF-1α, and VEGF levels in the heart of rats. METHODS Wistar rats were randomly divided into 4 groups (n = 6): control; ghrelin, received daily intraperitoneal injections of ghrelin; hypoxia, was exposed to hypoxic condition; hypoxia + ghrelin, was exposed to hypoxic condition and received intraperitoneal injections of ghrelin, for 2 weeks. Myocardial angiogenesis, the expression level of miR-210, and protein levels of HIF-1α and VEGF were assayed in the heart samples. RESULTS Hypoxia increased myocardial angiogenesis and cardiac levels of miR-210, HIF-1α, and VEGF. However, ghrelin inhibited these hypoxia-induced changes. Interestingly, ghrelin had no significant effect on miR-210, HIF-1α, and VEGF levels in normoxic condition. CONCLUSION Ghrelin may be useful as an anti-angiogenic factor.
Collapse
Affiliation(s)
- Fariba Mirzaei Bavil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Karimi-Sales
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alipour
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Szymczak RK, Grzywacz T, Ziemann E, Sawicka M, Laskowski R. Prolonged Sojourn at Very High Altitude Decreases Sea-Level Anaerobic Performance, Anaerobic Threshold, and Fat Mass. Front Physiol 2021; 12:743535. [PMID: 34675820 PMCID: PMC8523780 DOI: 10.3389/fphys.2021.743535] [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: 07/18/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022] Open
Abstract
Background: The influence of high altitude on an organism’s physiology depends on the length and the level of hypoxic exposure it experiences. This study aimed to determine the effect of a prolonged sojourn at very high altitudes (above 3,500m) on subsequent sea-level physical performance, body weight, body composition, and hematological parameters. Materials and Methods: Ten alpinists, nine males and one female, with a mean age of 27±4years, participated in the study. All had been on mountaineering expeditions to 7,000m peaks, where they spent 30±1days above 3,500m with their average sojourn at 4,900±60m. Their aerobic and anaerobic performance, body weight, body composition, and hematological parameters were examined at an altitude of 100m within 7days before the expeditions and 7days after they descended below 3,500m. Results: We found a significant (p<0.01) decrease in maximal anaerobic power (MAPWAnT) from 9.9±1.3 to 9.2±1.3W·kg−1, total anaerobic work from 248.1±23.8 to 228.1±20.1J·kg−1, anaerobic threshold from 39.3±8.0 to 27.8±5.6 mlO2·kg−1·min−1, body fat mass from 14.0±3.1 to 11.5±3.3%, and a significant increase (p<0.05) in maximal tidal volume from 3.2 [3.0–3.2] to 3.5 [3.3–3.9] L after their sojourn at very high attitude. We found no significant changes in maximal aerobic power, maximal oxygen uptake, body weight, fat-free mass, total body water, hemoglobin, and hematocrit. Conclusion: A month-long exposure to very high altitude led to impaired sea-level anaerobic performance and anaerobic threshold, increased maximal tidal volume, and depleted body fat mass, but had no effect on maximal aerobic power, maximal oxygen uptake, or hemoglobin and hematocrit levels.
Collapse
Affiliation(s)
- Robert K Szymczak
- Department of Emergency Medicine, Faculty of Health Sciences, Medical University of Gdańsk, Gdańsk, Poland
| | - Tomasz Grzywacz
- Department of Sport, Institute of Physical Culture, Kazimierz Wielki University in Bydgoszcz, Bydgoszcz, Poland
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznan, Poland
| | - Magdalena Sawicka
- Department of Neurology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Radosław Laskowski
- Department of Physiology and Biochemistry, Gdańsk University of Physical Education and Sport, Gdańsk, Poland
| |
Collapse
|
12
|
Mühlfeld C. Stereology and three-dimensional reconstructions to analyze the pulmonary vasculature. Histochem Cell Biol 2021; 156:83-93. [PMID: 34272602 PMCID: PMC8397636 DOI: 10.1007/s00418-021-02013-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2021] [Indexed: 02/05/2023]
Abstract
The pulmonary vasculature consists of a large arterial and venous tree with a vast alveolar capillary network (ACN) in between. Both conducting blood vessels and the gas-exchanging capillaries are part of important human lung diseases, including bronchopulmonary dysplasia, pulmonary hypertension and chronic obstructive pulmonary disease. Morphological tools to investigate the different parts of the pulmonary vasculature quantitatively and in three dimensions are crucial for a better understanding of the contribution of the blood vessels to the pathophysiology and effects of lung diseases. In recent years, new stereological methods and imaging techniques have expanded the analytical tool box and therefore the conclusive power of morphological analyses of the pulmonary vasculature. Three of these developments are presented and discussed in this review article, namely (1) stereological quantification of the number of capillary loops, (2) serial block-face scanning electron microscopy of the ACN and (3) labeling of branching generations in light microscopic sections based on arterial tree segmentations of micro-computed tomography data sets of whole lungs. The implementation of these approaches in research work requires expertise in lung preparation, multimodal imaging at different scales, an advanced IT infrastructure and expertise in image analysis. However, they are expected to provide important data that cannot be obtained by previously existing methodology.
Collapse
Affiliation(s)
- Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany. .,Research Core Unit Electron Microscopy, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
13
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Xin Y, Li J, Wu W, Liu X. Mitofusin-2: A New Mediator of Pathological Cell Proliferation. Front Cell Dev Biol 2021; 9:647631. [PMID: 33869201 PMCID: PMC8049505 DOI: 10.3389/fcell.2021.647631] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 02/05/2023] Open
Abstract
Cell proliferation is an important cellular process for physiological tissue homeostasis and remodeling. The mechanisms of cell proliferation in response to pathological stresses are not fully understood. Mitochondria are highly dynamic organelles whose shape, number, and biological functions are modulated by mitochondrial dynamics, including fusion and fission. Mitofusin-2 (Mfn-2) is an essential GTPase-related mitochondrial dynamics protein for maintaining mitochondrial network and bioenergetics. A growing body of evidence indicates that Mfn-2 has a potential role in regulating cell proliferation in various cell types. Here we review these new functions of Mfn-2, highlighting its crucial role in several signaling pathways during the process of pathological cell proliferation. We conclude that Mfn-2 could be a new mediator of pathological cell proliferation and a potential therapeutic target.
Collapse
Affiliation(s)
- Yanguo Xin
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Junli Li
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Li L, Xu M, Rowan SC, Howell K, Russell-Hallinan A, Donnelly SC, McLoughlin P, Baugh JA. The effects of genetic deletion of Macrophage migration inhibitory factor on the chronically hypoxic pulmonary circulation. Pulm Circ 2021; 10:2045894020941352. [PMID: 33447370 PMCID: PMC7780187 DOI: 10.1177/2045894020941352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/18/2020] [Indexed: 11/17/2022] Open
Abstract
While it is well established that the haemodynamic cause of hypoxic pulmonary hypertension is increased pulmonary vascular resistance, the molecular pathogenesis of the increased resistance remains incompletely understood. Macrophage migration inhibitory factor is a pleiotropic cytokine with endogenous tautomerase enzymatic activity as well as both intracellular and extracellular signalling functions. In several diseases, macrophage migration inhibitory factor has pro-inflammatory roles that are dependent upon signalling through the cell surface receptors CD74, CXCR2 and CXCR4. Macrophage migration inhibitory factor expression is increased in animal models of hypoxic pulmonary hypertension and macrophage migration inhibitory factor tautomerase inhibitors, which block some of the functions of macrophage migration inhibitory factor, and have been shown to attenuate hypoxic pulmonary hypertension in mice and monocrotaline-induced pulmonary hypertension in rats. However, because of the multiple pathways through which it acts, the integrated actions of macrophage migration inhibitory factor during the development of hypoxic pulmonary hypertension were unclear. We report here that isolated lungs from adult macrophage migration inhibitory factor knockout (MIF-/- ) mice maintained in normoxic conditions showed greater acute hypoxic vasoconstriction than the lungs of wild type mice (MIF+/+ ). Following exposure to hypoxia for three weeks, isolated lungs from MIF-/- mice had significantly higher pulmonary vascular resistance than those from MIF+/+ mice. The major mechanism underlying the greater increase in pulmonary vascular resistance in the hypoxic MIF-/- mice was reduction of the pulmonary vascular bed due to an impairment of the normal hypoxia-induced expansion of the alveolar capillary network. Taken together, these results demonstrate that macrophage migration inhibitory factor plays a central role in the development of the pulmonary vascular responses to chronic alveolar hypoxia.
Collapse
Affiliation(s)
- Lili Li
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maojia Xu
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Simon C Rowan
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Katherine Howell
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Adam Russell-Hallinan
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - John A Baugh
- UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Hypoxia and its preconditioning on cardiac and vascular remodelling in experimental animals. Respir Physiol Neurobiol 2020; 285:103588. [PMID: 33253893 DOI: 10.1016/j.resp.2020.103588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/28/2020] [Accepted: 11/12/2020] [Indexed: 11/21/2022]
Abstract
Since oxygen (O2) is indispensable for mammalian life, every cell in the body is endowed with mechanisms to detect and to respond to changes in the O2 levels in the microenvironment. The heart and the brain are the two most vital, life-supporting organs requiring a continuous supply of O2 to sustain their high metabolic rate. On being challenged with hypoxia, maintenance of O2 supply to these organs even at the cost of others becomes a priority. This review describes the cardiovascular, skeletal muscle vascular, pulmonary vascular and cerebrovascular remodelling in face of chronic mild hypoxia exposure and the underlying mechanisms, with special reference to the role of oxidative stress, hypoxia signalling, autonomic nervous mechanisms. The significance of the normalized wall index (NWI) in assessing the remodelling of the vessels particularly of the intramyocardial coronary artery has been underscored. The review also highlights the basic concepts of hypoxic preconditioning and the subsequent protection of the brain against an acute ischemic insult in preclinical studies hinting towards its possible therapeutic potential in the management of ischemic stroke.
Collapse
|
17
|
Tuder RM, Cool CD. Pulmonary Arteries and Microcirculation in COPD With Pulmonary Hypertension: Bystander or Culprit? Chest 2020; 156:4-6. [PMID: 31279376 DOI: 10.1016/j.chest.2019.04.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 04/02/2019] [Indexed: 10/26/2022] Open
Affiliation(s)
- Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Carlyne D Cool
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO.
| |
Collapse
|
18
|
Cao YY, Ba HX, Li Y, Tang SY, Luo ZQ, Li XH. Regulatory effects of Prohibitin 1 on proliferation and apoptosis of pulmonary arterial smooth muscle cells in monocrotaline-induced PAH rats. Life Sci 2020; 250:117548. [PMID: 32173312 DOI: 10.1016/j.lfs.2020.117548] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by unbalanced proliferation and apoptosis of pulmonary arterial smooth muscle cells (PASMCs). Prohibitin 1 (PHB1) is known for its significant anti-proliferative activity. However, the role of PHB1 in PASMCs and PAH have not been elucidated. METHODS Monocrotaline (MCT 60 mg/kg) was used to build a PAH model in SD rats. Right ventricular systolic pressure (RVSP) and right ventricle (RV) hypertrophy were measured. Morphology of pulmonary vessels was observed by Hematoxylin-Eosin (HE) staining. Expression of PHB1 in pulmonary arteries and PASMCs was determinated by immunoblot and immunofluorescence. Cell proliferation was detected by CCK8 and EDU when PASMCs were stimulated by PDGF-BB (20 ng/mL). Furthermore, siRNA for PHB1 and Akt inhibitor were conducted to investigate the mechanism behind the role of PHB1 and AKT signaling pathway in PASMCs proliferation and apoptosis. RESULTS The protein expression of PHB1 in PAH rats lung tissue was significantly up-regulated accompanied by elevated RVSP and enhanced RV hypertrophy. Immunohistochemistry showed that PHB1 was mainly localized in the pulmonary vascular smooth muscle layer. PDGF-BB significantly up-regulated the expression of PHB1 in rat primary PASMCs in a time- and dose-dependent manner. After PHB1 knock down, PASMCs proliferation was significantly suppressed while apoptosis was significantly recovered. Meanwhile the level of proliferating cell nuclear antigen (PCNA) and P-Akt were significantly down-regulated. Perifosine (Akt inhibitor) also significantly inhibit proliferation of PASMCs. CONCLUSION PHB1 contributes to pulmonary vascular remodeling by accelerating proliferation of PASMCs which involves AKT phosphorylation.
Collapse
Affiliation(s)
- Yuan-Yuan Cao
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University, Changsha 410013, China
| | - Hui-Xue Ba
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University, Changsha 410013, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Si-Yuan Tang
- Xiangya Nursing School, Central South University, Changsha 410013, China
| | - Zi-Qiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Xiao-Hui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Science, Central South University, Changsha 410013, China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha 411000, China.
| |
Collapse
|
19
|
Kröpfl JM, Kammerer T, Faihs V, Gruber HJ, Stutz J, Rehm M, Stelzer I, Schäfer ST, Spengler CM. Acute Exercise in Hypobaric Hypoxia Attenuates Endothelial Shedding in Subjects Unacclimatized to High Altitudes. Front Physiol 2020; 10:1632. [PMID: 32116736 PMCID: PMC7010936 DOI: 10.3389/fphys.2019.01632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/26/2019] [Indexed: 11/13/2022] Open
Abstract
Travel of unacclimatized subjects to a high altitude has been growing in popularity. Changes in endothelial shedding [circulating endothelial cells (ECs)] and hematopoietic stem and progenitor cells (CPCs) during physical exercise in hypobaric hypoxia, however, are not well understood. We investigated the change in ECs and CPCs when exposed to high altitude, after acute exercise therein, and after an overnight stay in hypobaric hypoxia in 11 healthy unacclimatized subjects. Blood withdrawal was done at baseline (520 m a.s.l.; baseline), after passive ascent to 3,883 m a.s.l. (arrival), after acute physical exercise (±400 m, postexercise) and after an overnight stay at 3,883 m a.s.l. (24 h). Mature blood cells, ECs, and CPCs were assessed by a hematology analyzer and flow cytometry, respectively. The presence of matrix metalloproteinases (MMPs), their activity, and hematopoietic cytokines were assessed in serum and plasma. EC and CPC concentrations significantly decreased after exercise (p = 0.019, p = 0.007, respectively). CPCs remained low until the next morning (24 h, p = 0.002), while EC concentrations returned back to baseline. MMP-9 decreased at arrival (p = 0.021), stayed low postexercise (p = 0.033), and returned to baseline at 24 h (p = 0.035 to postexercise). MMP-activity did not change throughout the study. Circulating MMP-9 concentrations, but not MMP-activity, were associated with EC concentrations (rrm = 0.48, p = 0.010). CPC concentrations were not linked to hematopoietic cytokines. Acute exercise at high altitude attenuated endothelial shedding, but did not enhance regenerative CPCs. Results were not linked to endothelial matrix remodeling or CPC mobilization. These results provide information to better understand the endothelium and immature immune system during an active, short-term sojourn at high altitude.
Collapse
Affiliation(s)
- Julia M Kröpfl
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
| | - Tobias Kammerer
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany.,Institute of Anesthesiology, Heart and Diabetes Center NRW, Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Valentina Faihs
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hans-Jürgen Gruber
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Jan Stutz
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland
| | - Markus Rehm
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany
| | - Ingeborg Stelzer
- Institute of Medical and Chemical Laboratory Diagnostics, LKH Hochsteiermark, Leoben, Austria
| | - Simon T Schäfer
- Department of Anaesthesiology, Ludwig Maximilian University of Munich, Munich, Germany.,Walter Brendel Centre of Experimental Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christina M Spengler
- Exercise Physiology Lab, Institute of Human Movement Sciences and Sport, ETH Zürich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Wang S, Zhang C, Zhang X. Downregulation of long non‑coding RNA ANRIL promotes proliferation and migration in hypoxic human pulmonary artery smooth muscle cells. Mol Med Rep 2019; 21:589-596. [PMID: 31974617 PMCID: PMC6947933 DOI: 10.3892/mmr.2019.10887] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/26/2019] [Indexed: 01/03/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive syndrome. When PAH occurs, the circulatory resistance of the pulmonary vasculature will gradually increase, which may lead to right heart failure and death. Pathological features of PAH include abnormal proliferation of pulmonary vascular smooth muscle cells and pulmonary vascular remodeling. Hypoxia is the main cause of PAH, which directly induces the contraction and proliferation of pulmonary artery smooth muscle cells (PASMCs), and eventually leads to pulmonary vascular remodeling. Recent studies have shown that long non-coding RNAs (lncRNAs) play key roles in numerous biological processes, including cell proliferation and the occurrence and development of cardiovascular diseases. Studies have also shown that lncRNA antisense noncoding RNA in the INK4 locus (ANRIL) can promote the proliferation of vascular smooth muscle cells. Therefore, the hypothesis of the present study was that ANRIL may be expressed in PASMCs and play a regulatory role. In this study, the expression of ANRIL was analyzed by quantitative PCR. The effects of ANRIL on human pulmonary artery smooth muscle cells (HPASMCs) were assessed by MTT assay, flow cytometry, bromodeoxyuridine incorporation assay, Transwell assay, scratch-wound assay, immunofluorescence assay and western blotting. These experiments revealed that the expression of ANRIL was significantly downregulated in HPASMCs induced by hypoxia. The downregulation of ANRIL affected the cell cycle, making more HPASMCs move from the G0/G1 phase to the G2/M+S phase and strengthening the cell proliferation. Moreover, downregulated ANRIL increased the migration of HPASMCs under hypoxia. This study identified ANRIL as a critical regulator in HPASMCs induced by hypoxia and demonstrated the potential of gene therapy and drug development for treating PAH.
Collapse
Affiliation(s)
- Siqi Wang
- College of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang 150076, P.R. China
| | - Chen Zhang
- College of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang 150076, P.R. China
| | - Xiaodan Zhang
- College of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang 150076, P.R. China
| |
Collapse
|
21
|
Volumetric optoacoustic tomography enables non-invasive in vivo characterization of impaired heart function in hypoxic conditions. Sci Rep 2019; 9:8369. [PMID: 31182733 PMCID: PMC6557887 DOI: 10.1038/s41598-019-44818-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/21/2019] [Indexed: 12/28/2022] Open
Abstract
Exposure to chronic hypoxia results in pulmonary hypertension characterized by increased vascular resistance and pulmonary vascular remodeling, changes in functional parameters of the pulmonary vasculature, and right ventricular hypertrophy, which can eventually lead to right heart failure. The underlying mechanisms of hypoxia-induced pulmonary hypertension have still not been fully elucidated while no curative treatment is currently available. Commonly employed pre-clinical analytic methods are largely limited to invasive studies interfering with cardiac tissue or otherwise ex vivo functional studies and histopathology. In this work, we suggest volumetric optoacoustic tomography (VOT) for non-invasive assessment of heart function in response to chronic hypoxia. Mice exposed for 3 consecutive weeks to normoxia or chronic hypoxia were imaged in vivo with heart perfusion tracked by VOT using indocyanide green contrast agent at high temporal (100 Hz) and spatial (200 µm) resolutions in 3D. Unequivocal difference in the pulmonary transit time was revealed between the hypoxic and normoxic conditions concomitant with the presence of pulmonary vascular remodeling within hypoxic models. Furthermore, a beat-to-beat analysis of the volumetric image data enabled identifying and characterizing arrhythmic events in mice exposed to chronic hypoxia. The newly introduced non-invasive methodology for analysis of impaired pulmonary vasculature and heart function under chronic hypoxic exposure provides important inputs into development of early diagnosis and treatment strategies in pulmonary hypertension.
Collapse
|
22
|
do Amaral-Silva L, Lambertz M, José Zara F, Klein W, Gargaglioni LH, Bícego KC. Parabronchial remodeling in chicks in response to embryonic hypoxia. ACTA ACUST UNITED AC 2019; 222:jeb.197970. [PMID: 31028104 DOI: 10.1242/jeb.197970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/18/2019] [Indexed: 01/31/2023]
Abstract
The embryonic development of parabronchi occurs mainly during the second half of incubation in precocious birds, which makes this phase sensitive to possible morphological modifications induced by O2 supply limitation. Thus, we hypothesized that hypoxia during the embryonic phase of parabronchial development induces morphological changes that remain after hatching. To test this hypothesis, chicken embryos were incubated entirely (21 days) under normoxia or partially under hypoxia (15% O2 during days 12 to 18). Lung structures, including air capillaries, blood capillaries, infundibula, atria, parabronchial lumen, bronchi, blood vessels larger than capillaries and interparabronchial tissue, in 1- and 10-day-old chicks were analyzed using light microscopy-assisted stereology. Tissue barrier and surface area of air capillaries were measured using electron microscopy-assisted stereology, allowing for calculation of the anatomical diffusion factor. Hypoxia increased the relative volumes of air and blood capillaries, structures directly involved in gas exchange, but decreased the relative volumes of atria in both groups of chicks, and the parabronchial lumen in older chicks. Accordingly, the surface area of the air capillaries and the anatomical diffusion factor were increased under hypoxic incubation. Treatment did not alter total lung volume, relative volumes of infundibula, bronchi, blood vessels larger than capillaries, interparabronchial tissue or the tissue barrier of any group. We conclude that hypoxia during the embryonic phase of parabronchial development leads to a morphological remodeling, characterized by increased volume density and respiratory surface area of structures involved in gas exchange at the expense of structures responsible for air conduction in chicks up to 10 days old.
Collapse
Affiliation(s)
- Lara do Amaral-Silva
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil.,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| | - Markus Lambertz
- Institut für Zoologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany.,Sektion Herpetologie, Zoologisches Forschungsmuseum Alexander Koenig, 53113 Bonn, Germany
| | - Fernando José Zara
- Invertebrate Morphology Lab, Department of Applied Biology, IEAMar and CAUNESP College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil
| | - Wilfried Klein
- National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil.,Department of Biology, School of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, São Paulo 14040-901, Brazil
| | - Luciane Helena Gargaglioni
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil.,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| | - Kênia Cardoso Bícego
- Department of Animal Morphology and Physiology, College of Agricultural and Veterinary Sciences, São Paulo State University, Unesp. Jaboticabal, São Paulo 14884-900, Brazil .,National Institute of Science and Technology - Comparative Physiology (INCT- Fisiologia Comparada), UNESP-Jaboticabal, São Paulo 14884-900, Brazil
| |
Collapse
|
23
|
Liu A, Liu Y, Li B, Yang M, Liu Y, Su J. Role of miR-223-3p in pulmonary arterial hypertension via targeting ITGB3 in the ECM pathway. Cell Prolif 2018; 52:e12550. [PMID: 30507047 PMCID: PMC6496671 DOI: 10.1111/cpr.12550] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/08/2018] [Accepted: 10/10/2018] [Indexed: 12/17/2022] Open
Abstract
Objectives To investigate the functions of miR‐223‐3p and ITGB3 in pulmonary arterial hypertension (PAH). Materials and Methods Microarray analysis was used to detect differentially expressed genes and microRNAs. In in vitro models, the expressions of miR‐223‐3p and ITGB3 were detected by qRT‐PCR and Western blot. α‐SMA expression and cell proliferation were analysed by immunofluorescence and MTT assay, respectively. In in vivo models, PAH progressions were determined by measuring the levels of mPAP and RVSP. Lung and myocardial tissues were subjected to HE staining and Masson and Sirius red‐saturated carbazotic acid staining to investigate the pathological features. Results The microarray analysis revealed that ITGB3 was upregulated, while hsa‐miR‐223‐3p was downregulated in PAH. After the induction of hypoxia, miR‐223‐3p was downregulated and ITGB3 was upregulated in PASMCs. Hypoxia induction promoted cell proliferation and inhibited α‐SMA expression in PASMCs. Both the upregulation of miR‐223‐3p and the downregulation of ITGB3 attenuated the aberrant proliferation induced by hypoxia conditions. After approximately 4 weeks, the mPAP and RVSP levels of rats injected with MCT were decreased by the overexpression of miR‐223‐3p or the silencing of ITGB3. The staining results revealed that both miR‐223‐3p overexpression and ITGB3 knockdown alleviated the pulmonary vascular remodelling and improved the PAH pathological features of rats. Conclusions MiR‐223‐3p alleviated the progression of PAH by suppressing the expression of ITGB3, a finding which provides novel targets for clinical treatment.
Collapse
Affiliation(s)
- Aijun Liu
- Department of Pediatric Cardiac Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yifan Liu
- Weifang Medicial University, Weifang, China
| | - Bin Li
- Department of Pediatric Cardiac Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ming Yang
- Department of Pediatric Cardiac Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Liu
- Department of Pediatric Cardiac Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junwu Su
- Department of Pediatric Cardiac Surgery Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Eldridge L, Wagner EM. Angiogenesis in the lung. J Physiol 2018; 597:1023-1032. [PMID: 30022479 DOI: 10.1113/jp275860] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/21/2018] [Indexed: 12/12/2022] Open
Abstract
Both systemic (tracheal and bronchial) and pulmonary circulations perfuse the lung. However, documentation of angiogenesis of either is complicated by the presence of the other. Well-documented angiogenesis of the systemic circulations have been identified in asthma, cystic fibrosis, chronic thromboembolism and primary carcinomas. Angiogenesis of the vasa vasorum, which are branches of bronchial arteries, is seen in the walls of large pulmonary vessels after a period of chronic hypoxia. Documentation of increased pulmonary capillaries has been shown in models of chronic hypoxia, after pneumonectomy and in some carcinomas. Although endothelial cell proliferation may occur as part of the repair process in several pulmonary diseases, it is separate from the unique establishment of new functional perfusing networks defined as angiogenesis. Identification of the mechanisms driving the expansion of new vascular beds in the adult needs further investigation. Yet the growth factors and molecular mechanisms of lung angiogenesis remain difficult to separate from underlying disease sequelae.
Collapse
Affiliation(s)
- Lindsey Eldridge
- Departments of Medicine and Environmental Health Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Elizabeth M Wagner
- Departments of Medicine and Environmental Health Sciences, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
25
|
Liu Y, Zhang H, Yan L, Du W, Zhang M, Chen H, Zhang L, Li G, Li J, Dong Y, Zhu D. MMP-2 and MMP-9 contribute to the angiogenic effect produced by hypoxia/15-HETE in pulmonary endothelial cells. J Mol Cell Cardiol 2018; 121:36-50. [PMID: 29913136 DOI: 10.1016/j.yjmcc.2018.06.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/06/2018] [Accepted: 06/13/2018] [Indexed: 11/15/2022]
Abstract
Matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9) are the predominant gelatinases in the developing lung. Studies have shown that the expression of MMP-2 and MMP-9 is upregulated in hypoxic fibroblasts, 15-hydroxyeicosatetraenoic acid (15-HETE) regulated fibroblasts migration via modulating MMP-2 or MMP-9, and that hypoxia/15-HETE is a predominant contributor to the development of pulmonary arterial hypertension (PAH) through increased angiogenesis. However, the roles of MMP-2 and MMP-9 in pulmonary arterial endothelial cells (PAECs) angiogenesis as well as the molecular mechanism of hypoxia-regulated MMP-2 and MMP-9 expression have not been identified. The aim of this study was to investigate the role of MMP-2 and MMP-9 in PAEC proliferation and vascular angiogenesis and to determine the effects of hypoxia-induced 15-HETE on the expression of MMP-2 and MMP-9. Western blot, immunofluorescence, and real-time PCR were used to measure the expression of MMP-2 and MMP-9 in hypoxic PAECs. Immunohistochemical staining, flow cytometry, and tube formation as well as cell proliferation, viability, scratch-wound, and Boyden chamber migration assays were used to identify the roles and relationships between MMP-2, MMP-9, and 15-HETE in hypoxic PAECs. We found that hypoxia increased MMP-2 and MMP-9 expression in pulmonary artery endothelium both in vivo and in vitro in a time-dependent pattern. Moreover, administration of the MMP-2 and MMP-9 inhibitor MMI-166 significantly reversed hypoxia-induced increases in right ventricular systemic pressure (RVSP), right ventricular function, and thickening of the tunica media. Furthermore, up-regulation of MMP-2 and MMP-9 expression was induced by 15-HETE, which regulates PAEC proliferation, migration, and cell cycle transition that eventually leads to angiogenesis. Our study demonstrated that hypoxia increases the expression of MMP-2 and MMP-9 through the 15-lipoxygenase/15-HETE pathway, and that MMP-2 and MMP-9 promote PAEC angiogenesis. These findings suggest that MMP-2 and MMP-9 may serve as new potential therapeutic targets for the treatment of PAH.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Lixin Yan
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Wei Du
- School of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang Province, China
| | - Min Zhang
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - He Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lixin Zhang
- Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, Heilongjiang Province, China
| | - Guangqun Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Jijin Li
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Yinchu Dong
- Medical Laboratory Technology, Harbin Medical University, Daqing, Heilongjiang Province, China
| | - Daling Zhu
- Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin 150001, Heilongjiang Province, China; Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang Province, China.
| |
Collapse
|
26
|
Yu X, Chen X, Zheng XD, Zhang J, Zhao X, Liu Y, Zhang H, Zhang L, Yu H, Zhang M, Ma C, Hao X, Zhu D. Growth Differentiation Factor 11 Promotes Abnormal Proliferation and Angiogenesis of Pulmonary Artery Endothelial Cells. Hypertension 2018; 71:729-741. [PMID: 29463625 DOI: 10.1161/hypertensionaha.117.10350] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/09/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Disordered proliferation and angiogenesis of pulmonary artery endothelial cells is an important stage in the development of pulmonary arterial hypertension. Recent studies revealed that GDF11 (growth differentiation factor 11) induces endothelial cells proliferation and migration; however, whether GDF11 is directly involved in the pathogenesis of pulmonary arterial hypertension remains unknown. Here, we found that GDF11 was significantly upregulated and activated in 2 experimental pulmonary arterial hypertension models and cultured pulmonary artery endothelial cells. Genetic ablation of gdf11 in endothelial cells rescued pulmonary arterial hypertension features, as demonstrated by right ventricle hypertrophy, right ventricular systolic pressure, hemodynamics, cardiac function, and vascular remodeling. Moreover, we found that hypoxia significantly increased cell cycle progression, proliferation, migration, adhesion, and tube formation, which were significantly inhibited by GDF11 small interfering RNA. These events could be reproduced using cultured pulmonary artery endothelial cells and were dependent on Smad signaling. Moreover, hypoxia-induced GDF11 expression was regulated by the transcription factor zinc finger protein 740, which assisted RNA polymerase in recognizing and binding to the GDF11 promoter sequence located at a site (-753/-744; CCCCCCCCAC) upstream of the gene. This study identified a novel growth and differentiation factor signaling pathway involved in the zinc finger protein 740/GDF11/transforming growth factor-β receptor I/Smad signaling axis and involved in pulmonary artery endothelial cells proliferation and angiogenesis. These results provide critical insights for the development of novel therapeutic strategies for pulmonary arterial hypertension involving components of the GDF11 signaling system.
Collapse
Affiliation(s)
- Xiufeng Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xinxin Chen
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xiao Dong Zheng
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Junting Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xijuan Zhao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Ying Liu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hongyue Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Lixin Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hao Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Min Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Cui Ma
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xuewei Hao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Daling Zhu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.).
| |
Collapse
|
27
|
Myocardial Fistulisation and Coronary Arterial Ectasia in Children with Univentricular Circulation: An Under-Recognised Problem. Pediatr Cardiol 2018; 39:254-260. [PMID: 29043397 DOI: 10.1007/s00246-017-1749-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 10/06/2017] [Indexed: 10/18/2022]
Abstract
Among 241 children with univentricular circulation who underwent total cavopulmonary connection (Glenn or Fontan) over a 17 years period (January 2000-April 2017), we found two patients who had diffuse coronary fistulisation of their myocardium (0.8%). We also report an additional third case from Oak Lawn, Illinois, USA. One of the children had documented normal coronary arteries and myocardium at catheterisation prior to development of the fistulisation process. The same child also developed extensive veno-venous collaterals. All children had evidence of elevated pulmonary arterial pressures and were treated with a combination of pulmonary vasodilators. The presence of chronic hypoxaemia and elevated filling pressures could potentially have contributed to coronary endothelial dysfunction, thereby giving rise to the coronary vasculopathy and myocardial fistulisation. Alternatively, this myocardial fistulisation process may be present from early in life and not recognised. This may necessitate early referral for orthotopic cardiac transplantation. Diffuse fistulisation of the myocardium in children and young adults with univentricular circulation may be an under-recognised occurrence and coronary angiography should be considered in these patients.
Collapse
|
28
|
Novotný T, Uhlík J, Vajner L. Four-day pulse of sodium cromoglycate modulates pulmonary vessel wall remodeling during 21-day hypoxia in rats. Exp Lung Res 2018; 44:1-12. [PMID: 29324062 DOI: 10.1080/01902148.2017.1393708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
AIM OF THE STUDY Remodeling of pulmonary resistance arteries in rats due to 4-day hypoxia could be successfully suppressed by sodium cromoglycate. In this study, we tested the difference in the suppression between two distinct time patterns of cromoglycate administration during 21-day hypoxia. In the experiment, we focused on some details in both smooth muscle cells and extracellular matrix of pulmonary arterial walls. METHODS During 21-day hypoxia, rats were treated with sodium cromoglycate either in the first four days or in the last four days. The first four days were chosen to test efficiency of an initial pulse of cromoglycate to suppress pulmonary vascular remodeling. The last four-day administration tested possibility to block remodeling post hoc. RESULTS Initial pulse reduced and modified remodeling in all levels of pulmonary arteries, which comprises neomuscularization of prealveolar arteries, asymmetrical hypertrophy of tunica media in muscular pulmonary arteries and hypertrophy of tunica media and tunica adventitia in large conduit arteries. Terminal pulse had only negligible effect. CONCLUSIONS Only the initial cromoglycate therapy led to significant morphological suppression of remodeling. We therefore assume important role of initial remodeling influencing during long time hypoxia experiment.
Collapse
Affiliation(s)
- Tomáš Novotný
- a Department of Histology and Embryology, Second Faculty of Medicine , Charles University in Prague , Plzeňska , Prague , Czech Republic.,b Department of Orthopedics , Municipal Hospital of Litoměřice , Žitenická, Litoměřice , Czech Republic
| | - Jiří Uhlík
- a Department of Histology and Embryology, Second Faculty of Medicine , Charles University in Prague , Plzeňska , Prague , Czech Republic
| | - Luděk Vajner
- a Department of Histology and Embryology, Second Faculty of Medicine , Charles University in Prague , Plzeňska , Prague , Czech Republic
| |
Collapse
|
29
|
Zhang C, Ma C, Yao H, Zhang L, Yu X, Liu Y, Shen T, Zhang L, Zhang F, Chen X, Zhu D. 12-Lipoxygenase and 12-hydroxyeicosatetraenoic acid regulate hypoxic angiogenesis and survival of pulmonary artery endothelial cells via PI3K/Akt pathway. Am J Physiol Lung Cell Mol Physiol 2017; 314:L606-L616. [PMID: 29074487 DOI: 10.1152/ajplung.00049.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Dysfunction and injury of endothelial cells play critical roles in pulmonary arterial hypertension, including aberrant proliferation, suppressed apoptosis, and excessive angiogenesis. The 12-lipoxygenase and 12-hydroxyeicosatetraenoic acid pathway, which has been considered as a crucial mediator, elevates pulmonary vascular resistance and pulmonary arterial pressure. However, the mechanisms underlying the bioactivity of 12-hydroxyeicosatetraenoic acid in pulmonary vasculature, especially in endothelial cells, are still elusive. Thus we aim to determine the key role of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid in angiogenesis and survival of pulmonary artery endothelial cells and ascertain the signaling pathways participating in the pathological process. Here we establish that hypoxia increases the formation of endogenous 12-hydroxyeicosatetraenoic acid through stimulation of 12-lipoxygenase. Furthermore, we put forward new information that 12-hydroxyeicosatetraenoic acid promotes endothelial cell migration and tube formation, whereas it inhibits the serum deprivation-induced apoptotic responses under hypoxia. Particularly, the regulatory effects of 12-lipoxygenase/12-hydroxyeicosatetraenoic acid on pulmonary artery endothelial cells, at least in part, depend on phosphatidylinositol 3-kinase (PI3K)/Akt signaling activation. Taken together, these results may have significant implications for understanding of pulmonary hypertension and offer a potential therapeutic concept focusing on the 12-lipoxygenase/12-hydroxyeicosatetraenoic acid signaling system.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Cui Ma
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Hongmin Yao
- Petit Science Center, Department of Biology, College of Arts and Sciences, Georgia State University , Atlanta, Georgia
| | - Lixin Zhang
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Xiufeng Yu
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Yumei Liu
- Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Tingting Shen
- Department of Pharmacology, Dalian Medical University , Dalian , China
| | - Linlin Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Fengying Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Xinxin Chen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University , Harbin , China.,Central Laboratory, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China.,Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University , Harbin , China
| |
Collapse
|
30
|
Herbert LM, Resta TC, Jernigan NL. RhoA increases ASIC1a plasma membrane localization and calcium influx in pulmonary arterial smooth muscle cells following chronic hypoxia. Am J Physiol Cell Physiol 2017; 314:C166-C176. [PMID: 29070491 DOI: 10.1152/ajpcell.00159.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Increases in pulmonary arterial smooth muscle cell (PASMC) intracellular Ca2+ levels and enhanced RhoA/Rho kinase-dependent Ca2+ sensitization are key determinants of PASMC contraction, migration, and proliferation accompanying the development of hypoxic pulmonary hypertension. We previously showed that acid-sensing ion channel 1a (ASIC1a)-mediated Ca2+ entry in PASMC is an important constituent of the active vasoconstriction, vascular remodeling, and right ventricular hypertrophy associated with hypoxic pulmonary hypertension. However, the enhanced ASIC1a-mediated store-operated Ca2+ entry in PASMC from pulmonary hypertensive animals is not dependent on an increase in ASIC1a protein expression, suggesting that chronic hypoxia (CH) stimulates ASIC1a function through other regulatory mechanism(s). RhoA is involved in ion channel trafficking, and levels of activated RhoA are increased following CH. Therefore, we hypothesize that activation of RhoA following CH increases ASIC1a-mediated Ca2+ entry by promoting ASIC1a plasma membrane localization. Consistent with our hypothesis, we found greater plasma membrane localization of ASIC1a following CH. Inhibition of RhoA decreased ASIC1a plasma membrane expression and largely diminished ASIC1a-mediated Ca2+ influx, whereas activation of RhoA had the opposite effect. A proximity ligation assay revealed that ASIC1a and RhoA colocalize in PASMC and that the activation state of RhoA modulates this interaction. Together, our findings show a novel interaction between RhoA and ASIC1a, such that activation of RhoA in PASMC, both pharmacologically and via CH, promotes ASIC1a plasma membrane localization and Ca2+ entry. In addition to enhanced RhoA-mediated Ca2+ sensitization following CH, RhoA can also activate a Ca2+ signal by facilitating ASIC1a plasma membrane localization and Ca2+ influx in pulmonary hypertension.
Collapse
Affiliation(s)
- Lindsay M Herbert
- 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
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center , Albuquerque, New Mexico
| |
Collapse
|
31
|
Kropski JA, Richmond BW, Gaskill CF, Foronjy RF, Majka SM. Deregulated angiogenesis in chronic lung diseases: a possible role for lung mesenchymal progenitor cells (2017 Grover Conference Series). Pulm Circ 2017; 8:2045893217739807. [PMID: 29040010 PMCID: PMC5731726 DOI: 10.1177/2045893217739807] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chronic lung disease (CLD), including pulmonary fibrosis (PF) and chronic obstructive pulmonary disease (COPD), is the fourth leading cause of mortality worldwide. Both are debilitating pathologies that impede overall tissue function. A common co-morbidity in CLD is vasculopathy, characterized by deregulated angiogenesis, remodeling, and loss of microvessels. This substantially worsens prognosis and limits survival, with most current therapeutic strategies being largely palliative. The relevance of angiogenesis, both capillary and lymph, to the pathophysiology of CLD has not been resolved as conflicting evidence depicts angiogenesis as both reparative or pathologic. Therefore, we must begin to understand and model the underlying pathobiology of pulmonary vascular deregulation, alone and in response to injury induced disease, to define cell interactions necessary to maintain normal function and promote repair. Capillary and lymphangiogenesis are deregulated in both PF and COPD, although the mechanisms by which they co-regulate and underlie early pathogenesis of disease are unknown. The cell-specific mechanisms that regulate lung vascular homeostasis, repair, and remodeling represent a significant gap in knowledge, which presents an opportunity to develop targeted therapies. We have shown that that ABCG2pos multipotent adult mesenchymal stem or progenitor cells (MPC) influence the function of the capillary microvasculature as well as lymphangiogenesis. A balance of both is required for normal tissue homeostasis and repair. Our current models suggest that when lymph and capillary angiogenesis are out of balance, the non-equivalence appears to support the progression of disease and tissue remodeling. The angiogenic regulatory mechanisms underlying CLD likely impact other interstitial lung diseases, tuberous sclerosis, and lymphangioleiomyomatosis.
Collapse
Affiliation(s)
- Jonathan A Kropski
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bradley W Richmond
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christa F Gaskill
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert F Foronjy
- 3 5718 Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Susan M Majka
- 1 12328 Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,2 74498 Department of Medicine, Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
32
|
Jernigan NL, Naik JS, Weise-Cross L, Detweiler ND, Herbert LM, Yellowhair TR, Resta TC. Contribution of reactive oxygen species to the pathogenesis of pulmonary arterial hypertension. PLoS One 2017; 12:e0180455. [PMID: 28666030 PMCID: PMC5493402 DOI: 10.1371/journal.pone.0180455] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/15/2017] [Indexed: 11/19/2022] Open
Abstract
Pulmonary arterial hypertension is associated with a decreased antioxidant capacity. However, neither the contribution of reactive oxygen species to pulmonary vasoconstrictor sensitivity, nor the therapeutic efficacy of antioxidant strategies in this setting are known. We hypothesized that reactive oxygen species play a central role in mediating both vasoconstrictor and arterial remodeling components of severe pulmonary arterial hypertension. We examined the effect of the chemical antioxidant, TEMPOL, on right ventricular systolic pressure, vascular remodeling, and enhanced vasoconstrictor reactivity in both chronic hypoxia and hypoxia/SU5416 rat models of pulmonary hypertension. SU5416 is a vascular endothelial growth factor receptor antagonist and the combination of chronic hypoxia/SU5416 produces a model of severe pulmonary arterial hypertension with vascular plexiform lesions/fibrosis that is not present with chronic hypoxia alone. The major findings from this study are: 1) compared to hypoxia alone, hypoxia/SU5416 exposure caused more severe pulmonary hypertension, right ventricular hypertrophy, adventitial lesion formation, and greater vasoconstrictor sensitivity through a superoxide and Rho kinase-dependent Ca2+ sensitization mechanism. 2) Chronic hypoxia increased medial muscularization and superoxide levels, however there was no effect of SU5416 to augment these responses. 3) Treatment with TEMPOL decreased right ventricular systolic pressure in both hypoxia and hypoxia/SU5416 groups. 4) This effect of TEMPOL was associated with normalization of vasoconstrictor responses, but not arterial remodeling. Rather, medial hypertrophy and adventitial fibrotic lesion formation were more pronounced following chronic TEMPOL treatment in hypoxia/SU5416 rats. Our findings support a major role for reactive oxygen species in mediating enhanced vasoconstrictor reactivity and pulmonary hypertension in both chronic hypoxia and hypoxia/SU5416 rat models, despite a paradoxical effect of antioxidant therapy to exacerbate arterial remodeling in animals with severe pulmonary arterial hypertension in the hypoxia/SU5416 model.
Collapse
Affiliation(s)
- Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
- * E-mail:
| | - Jay S. Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Laura Weise-Cross
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Neil D. Detweiler
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Lindsay M. Herbert
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Tracylyn R. Yellowhair
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Thomas C. Resta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| |
Collapse
|
33
|
Beck SC, Feng Y, Sothilingam V, Garcia Garrido M, Tanimoto N, Acar N, Shan S, Seebauer B, Berger W, Hammes HP, Seeliger MW. Long-term consequences of developmental vascular defects on retinal vessel homeostasis and function in a mouse model of Norrie disease. PLoS One 2017; 12:e0178753. [PMID: 28575130 PMCID: PMC5456345 DOI: 10.1371/journal.pone.0178753] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/18/2017] [Indexed: 11/18/2022] Open
Abstract
Loss of Norrin signalling due to mutations in the Norrie disease pseudoglioma gene causes severe vascular defects in the retina, leading to visual impairment and ultimately blindness. While the emphasis of experimental work so far was on the developmental period, we focus here on disease mechanisms that induce progression into severe adult disease. The goal of this study was the comprehensive analysis of the long-term effects of the absence of Norrin on vascular homeostasis and retinal function. In a mouse model of Norrie disease retinal vascular morphology and integrity were studied by means of in vivo angiography; the vascular constituents were assessed in detailed histological analyses using quantitative retinal morphometry. Finally, electroretinographic analyses were performed to assess the retinal function in adult Norrin deficient animals. We could show that the primary developmental defects not only persisted but developed into further vascular abnormalities and microangiopathies. In particular, the overall vessel homeostasis, the vascular integrity, and also the cellular constituents of the vascular wall were affected in the adult Norrin deficient retina. Moreover, functional analyses indicated to persistent hypoxia in the neural retina which was suggested as one of the major driving forces of disease progression. In summary, our data provide evidence that the key to adult Norrie disease are ongoing vascular modifications, driven by the persistent hypoxic conditions, which are ineffective to compensate for the primary Norrin-dependent defects.
Collapse
MESH Headings
- Angiography
- Animals
- Blindness/congenital
- Blindness/diagnostic imaging
- Blindness/genetics
- Blindness/pathology
- Capillaries/pathology
- Cell Hypoxia
- Disease Models, Animal
- Disease Progression
- Electroretinography
- Eye Proteins/genetics
- Eye Proteins/physiology
- Genetic Diseases, X-Linked/diagnostic imaging
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/pathology
- Lasers
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/pathology
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/physiology
- Nervous System Diseases/diagnostic imaging
- Nervous System Diseases/genetics
- Nervous System Diseases/pathology
- Ophthalmoscopy/methods
- Retinal Degeneration
- Retinal Vessels/diagnostic imaging
- Retinal Vessels/pathology
- Spasms, Infantile/diagnostic imaging
- Spasms, Infantile/genetics
- Spasms, Infantile/pathology
Collapse
Affiliation(s)
- Susanne C. Beck
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
- * E-mail:
| | - Yuxi Feng
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Vithiyanjali Sothilingam
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
| | - Marina Garcia Garrido
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
| | - Naoyuki Tanimoto
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
| | - Niyazi Acar
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
| | - Shenliang Shan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Britta Seebauer
- Institute of Medical Molecular Genetics, University of Zurich, Zurich, Switzerland
| | - Wolfgang Berger
- Institute of Medical Molecular Genetics, University of Zurich, Zurich, Switzerland
- Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich (ZNZ), University and ETH Zurich, Zurich, Switzerland
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Mathias W. Seeliger
- Division of Ocular Neurodegeneration, Institute for Ophthalmic Research, Centre for Ophthalmology, Tuebingen, Germany
| |
Collapse
|
34
|
Cryopreserved, Xeno-Free Human Umbilical Cord Mesenchymal Stromal Cells Reduce Lung Injury Severity and Bacterial Burden in Rodent Escherichia coli-Induced Acute Respiratory Distress Syndrome. Crit Care Med 2017; 45:e202-e212. [PMID: 27861182 DOI: 10.1097/ccm.0000000000002073] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Although mesenchymal stem/stromal cells represent a promising therapeutic strategy for acute respiratory distress syndrome, clinical translation faces challenges, including scarcity of bone marrow donors, and reliance on bovine serum during mesenchymal stem/stromal cell proliferation. We wished to compare mesenchymal stem/stromal cells from human umbilical cord, grown in xeno-free conditions, with mesenchymal stem/stromal cells from human bone marrow, in a rat model of Escherichia coli pneumonia. In addition, we wished to determine the potential for umbilical cord-mesenchymal stem/stromal cells to reduce E. coli-induced oxidant injury. DESIGN Randomized animal study. SETTING University research laboratory. SUBJECTS Male Sprague-Dawley rats. INTERVENTIONS Acute respiratory distress syndrome was induced in rats by intratracheal instillation of E. coli (1.5-2 × 10 CFU/kg). "Series 1" compared the effects of freshly thawed cryopreserved umbilical cord-mesenchymal stem/stromal cells with bone marrow-mesenchymal stem/stromal cells on physiologic indices of lung injury, cellular infiltration, and E. coli colony counts in bronchoalveolar lavage. "Series 2" examined the effects of cryopreserved umbilical cord-mesenchymal stem/stromal cells on survival, as well as measures of injury, inflammation and oxidant stress, including production of reactive oxidative species, reactive oxidative species scavenging by superoxide dismutase-1 and superoxide dismutase-2. MEASUREMENTS AND MAIN RESULTS In "Series 1," animals subjected to E. coli pneumonia who received umbilical cord-mesenchymal stem/stromal cells had improvements in oxygenation, respiratory static compliance, and wet-to-dry ratios comparable to bone marrow-mesenchymal stem/stromal cell treatment. E. coli colony-forming units in bronchoalveolar lavage were reduced in both cell therapy groups, despite a reduction in bronchoalveolar lavage neutrophils. In series 2, umbilical cord-mesenchymal stem/stromal cells enhanced animal survival and decreased alveolar protein and proinflammatory cytokine concentrations, whereas increasing interleukin-10 concentrations. Umbilical cord-mesenchymal stem/stromal cell therapy decreased nicotinamide adenine dinucleotide phosphate-oxidase 2 and inducible nitric oxide synthase and enhanced lung concentrations of superoxide dismutase-2, thereby reducing lung tissue reactive oxidative species concentrations. CONCLUSIONS Our results demonstrate that freshly thawed cryopreserved xeno-free human umbilical cord-mesenchymal stem/stromal cells reduce the severity of rodent E. coli-induced acute respiratory distress syndrome. Umbilical cord-mesenchymal stem/stromal cells, therefore, represent an attractive option for future clinical trials in acute respiratory distress syndrome.
Collapse
|
35
|
Hsia CCW. Comparative analysis of the mechanical signals in lung development and compensatory growth. Cell Tissue Res 2017; 367:687-705. [PMID: 28084523 PMCID: PMC5321790 DOI: 10.1007/s00441-016-2558-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022]
Abstract
This review compares the manner in which physical stress imposed on the parenchyma, vasculature and thorax and the thoraco-pulmonary interactions, drive both developmental and compensatory lung growth. Re-initiation of anatomical lung growth in the mature lung is possible when the loss of functioning lung units renders the existing physiologic-structural reserves insufficient for maintaining adequate function and physical stress on the remaining units exceeds a critical threshold. The appropriate spatial and temporal mechanical interrelationships and the availability of intra-thoracic space, are crucial to growth initiation, follow-on remodeling and physiological outcome. While the endogenous potential for compensatory lung growth is retained and may be pharmacologically augmented, supra-optimal mechanical stimulation, unbalanced structural growth, or inadequate remodeling may limit functional gain. Finding ways to optimize the signal-response relationships and resolve structure-function discrepancies are major challenges that must be overcome before the innate compensatory ability could be fully realized. Partial pneumonectomy reproducibly removes a known fraction of functioning lung units and remains the most robust model for examining the adaptive mechanisms, structure-function consequences and plasticity of the remaining functioning lung units capable of regeneration. Fundamental mechanical stimulus-response relationships established in the pneumonectomy model directly inform the exploration of effective approaches to maximize compensatory growth and function in chronic destructive lung diseases, transplantation and bioengineered lungs.
Collapse
Affiliation(s)
- Connie C W Hsia
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., Dallas, TX, 75390-9034, USA.
| |
Collapse
|
36
|
Cavadas MAS, Cheong A, Taylor CT. The regulation of transcriptional repression in hypoxia. Exp Cell Res 2017; 356:173-181. [PMID: 28219680 DOI: 10.1016/j.yexcr.2017.02.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/20/2022]
Abstract
A sufficient supply molecular oxygen is essential for the maintenance of physiologic metabolism and bioenergetic homeostasis for most metazoans. For this reason, mechanisms have evolved for eukaryotic cells to adapt to conditions where oxygen demand exceeds supply (hypoxia). These mechanisms rely on the modification of pre-existing proteins, translational arrest and transcriptional changes. The hypoxia inducible factor (HIF; a master regulator of gene induction in response to hypoxia) is responsible for the majority of induced gene expression in hypoxia. However, much less is known about the mechanism(s) responsible for gene repression, an essential part of the adaptive transcriptional response. Hypoxia-induced gene repression leads to a reduction in energy demanding processes and the redirection of limited energetic resources to essential housekeeping functions. Recent developments have underscored the importance of transcriptional repressors in cellular adaptation to hypoxia. To date, at least ten distinct transcriptional repressors have been reported to demonstrate sensitivity to hypoxia. Central among these is the Repressor Element-1 Silencing Transcription factor (REST), which regulates over 200 genes. In this review, written to honor the memory and outstanding scientific legacy of Lorenz Poellinger, we provide an overview of our existing knowledge with respect to transcriptional repressors and their target genes in hypoxia.
Collapse
Affiliation(s)
- Miguel A S Cavadas
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156 Oeiras, Portugal
| | - Alex Cheong
- Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | - Cormac T Taylor
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland; Conway Institute of Biomolecular and Biomedical Research, School of Medicine and Medical Sciences and Systems Biology Ireland, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
37
|
Maron BA, Machado RF, Shimoda L. Pulmonary vascular and ventricular dysfunction in the susceptible patient (2015 Grover Conference series). Pulm Circ 2016; 6:426-438. [PMID: 28090285 PMCID: PMC5210067 DOI: 10.1086/688315] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Pulmonary blood vessel structure and tone are maintained by a complex interplay between endogenous vasoactive factors and oxygen-sensing intermediaries. Under physiological conditions, these signaling networks function as an adaptive interface between the pulmonary circulation and environmental or acquired perturbations to preserve oxygenation and maintain systemic delivery of oxygen-rich hemoglobin. Chronic exposure to hypoxia, however, triggers a range of pathogenetic mechanisms that include hypoxia-inducible factor 1α (HIF-1α)-dependent upregulation of the vasoconstrictor peptide endothelin 1 in pulmonary endothelial cells. In pulmonary arterial smooth muscle cells, chronic hypoxia induces HIF-1α-mediated upregulation of canonical transient receptor potential proteins, as well as increased Rho kinase-Ca2+ signaling and pulmonary arteriole synthesis of the profibrotic hormone aldosterone. Collectively, these mechanisms contribute to a contractile or hypertrophic pulmonary vascular phenotype. Genetically inherited disorders in hemoglobin structure are also an important etiology of abnormal pulmonary vasoreactivity. In sickle cell anemia, for example, consumption of the vasodilator and antimitogenic molecule nitric oxide by cell-free hemoglobin is an important mechanism underpinning pulmonary hypertension. Contemporary genomic and transcriptomic analytic methods have also allowed for the discovery of novel risk factors relevant to sickle cell disease, including GALNT13 gene variants. In this report, we review cutting-edge observations characterizing these and other pathobiological mechanisms that contribute to pulmonary vascular and right ventricular vulnerability.
Collapse
Affiliation(s)
- Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA; and Department of Cardiology, Boston Veterans Affairs Healthcare System, Boston, Massachusetts, USA
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Larissa Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
38
|
Huetsch JC, Suresh K, Bernier M, Shimoda LA. Update on novel targets and potential treatment avenues in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2016; 311:L811-L831. [PMID: 27591245 PMCID: PMC5130539 DOI: 10.1152/ajplung.00302.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 08/29/2016] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a condition marked by a combination of constriction and remodeling within the pulmonary vasculature. It remains a disease without a cure, as current treatments were developed with a focus on vasodilatory properties but do not reverse the remodeling component. Numerous recent advances have been made in the understanding of cellular processes that drive pathologic remodeling in each layer of the vessel wall as well as the accompanying maladaptive changes in the right ventricle. In particular, the past few years have yielded much improved insight into the pathways that contribute to altered metabolism, mitochondrial function, and reactive oxygen species signaling and how these pathways promote the proproliferative, promigratory, and antiapoptotic phenotype of the vasculature during PH. Additionally, there have been significant advances in numerous other pathways linked to PH pathogenesis, such as sex hormones and perivascular inflammation. Novel insights into cellular pathology have suggested new avenues for the development of both biomarkers and therapies that will hopefully bring us closer to the elusive goal: a therapy leading to reversal of disease.
Collapse
Affiliation(s)
- John C Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| | - Meghan Bernier
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
39
|
Li L, Watson CJ, Dubourd M, Bruton A, Xu M, Cooke G, Baugh JA. HIF-1-Dependent TGM1 Expression is Associated with Maintenance of Airway Epithelial Junction Proteins. Lung 2016; 194:829-38. [PMID: 27423780 DOI: 10.1007/s00408-016-9918-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 07/02/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Hypoxia has been implicated in the pathogenesis of many inflammatory and fibrotic lung diseases. The effect of hypoxia on epithelial junction protein expression is yet to be fully elucidated but evidence suggests a protective role for the hypoxia-inducible transcription factor HIF-1 in stabilising occludin. Transglutaminase 1 (TGM1) has been shown to stabilise endothelial and keratinocyte cell junctions, and while its expression and function have been mostly studied in the skin, recent studies have reported its expression in the lung. We hypothesised that TGM1 is a hypoxia-induced regulator of pulmonary epithelial junction protein stability, and the aim of this study was to investigate the regulation of TGM1 expression by hypoxia. METHODS Hypoxia-responsive genes were identified in human small airway epithelial cells (SAECs) by DNA microarray. TGM1 mRNA expression in SAECs was measured by quantitative real-time PCR. Protein expression of TGM1 and junction proteins was investigated by western blotting. Hypoxia-induced TGM1 was analysed by immunohistochemistry in vivo. The TGM1 gene promoter was investigated by luciferase assay. RESULTS In vitro exposure of SAECs to hypoxia induced a significant increase in TGM1 expression at both mRNA and protein levels. TGM1 was also significantly upregulated in hypoxic mouse lung epithelium. The hypoxia-responsive region was mapped to a HIF-1-responsive element. Inhibition of HIF-1 expression abolished hypoxia-induced promoter activation. Overexpression of TGM1 in lung epithelial cells or exposure of SAECs to hypoxia led to upregulated expression of junction proteins. CONCLUSION Herein we report that TGM1 is a HIF-1-regulated gene that is associated with the upregulation of airway epithelial junction proteins, supporting a protective role for HIF-1 in the lung. Interventions that augment the expression of TGM1 may provide useful therapeutic strategies for maintaining pulmonary epithelial integrity during lung injury.
Collapse
Affiliation(s)
- Lili Li
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Chris J Watson
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.,Centre for Experimental Medicine, Queen's University Belfast, Wellcome-Wolfson Building, Belfast, Northern Ireland, UK
| | - Mickael Dubourd
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Aine Bruton
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Maojia Xu
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gordon Cooke
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - John A Baugh
- UCD Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
40
|
Geng J, Fan FL, He S, Liu Y, Meng Y, Tian H, Zhang D, Ma Q, Zhang JB, Tian HY. The effects of the 5-HT2Areceptor antagonist sarpogrelate hydrochloride on chronic hypoxic pulmonary hypertension in rats. Exp Lung Res 2016; 42:190-8. [DOI: 10.1080/01902148.2016.1181122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Shen T, Wang N, Yu X, Shi J, Li Q, Zhang C, Fu L, Wang S, Xing Y, Zheng X, Yu L, Zhu D. The Critical Role of Dynamin-Related Protein 1 in Hypoxia-Induced Pulmonary Vascular Angiogenesis. J Cell Biochem 2016; 116:1993-2007. [PMID: 25752284 DOI: 10.1002/jcb.25154] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/03/2015] [Indexed: 11/09/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a lethal disease characterized by pulmonary vascular obstruction due in part to excessive pulmonary artery endothelial cells (PAECs) migration and proliferation. The mitochondrial fission protein dynamin-related protein-1 (DRP1) has important influence on pulmonary vascular remodeling. However, whether DRP1 participates in the development and progression of pulmonary vascular angiogenesis has not been reported previously. To test the hypothesis that DRP1 promotes the angiogenesis via promoting the proliferation, stimulating migration, and inhibiting the apoptosis of PAECs in mitochondrial Ca(2+)-dependent manner, we performed following studies. Using hemodynamic analysis and morphometric assay, we found that DRP1 mediated the elevation of right ventricular systemic pressure (RVSP), right heart hypertrophy, and increase of pulmonary microvessels induced by hypoxia. DRP1 inhibition reversed tube network formation in vitro stimulated by hypoxia. The mitochondrial Ca(2+) inhibited by hypoxia was recovered by DRP1 silencing. Moreover, pulmonary vascular angiogenesis promoted by DRP1 was reversed by the specific mitochondrial Ca(2+) uniporter inhibitor Ru360. In addition, DRP1 promoted the proliferation and migration of PAECs in mitochondrial Ca(2+)-dependent manner. Besides, DRP1 decreased mitochondrial membrane potential, reduced the DNA fragmentation, and inhibited the caspase-3 activation, which were all aggravated by Ru360. Therefore, these results indicate that the mitochondrial fission machinery promotes migration, facilitates proliferation, and prevents from apoptosis via mitochondrial Ca(2+)-dependent pathway in endothelial cells leading to pulmonary angiogenesis.
Collapse
Affiliation(s)
- Tingting Shen
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Na Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Xiufeng Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | | | | | - Chen Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Li Fu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Shuang Wang
- Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, China
| | - Yan Xing
- Department of Pharmacology, College of Basic Medicine, Harbin Medical University, Daqing, China
| | - Xiaodong Zheng
- Department of Pathophysiology, College of Basic Medicine, Harbin Medical University, Daqing, China
| | - Lei Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University (Daqing), Daqing, China.,Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, China
| |
Collapse
|
42
|
Goss KN, Tepper RS, Lahm T, Ahlfeld SK. Increased Cardiac Output and Preserved Gas Exchange Despite Decreased Alveolar Surface Area in Rats Exposed to Neonatal Hyperoxia and Adult Hypoxia. Am J Respir Cell Mol Biol 2016; 53:902-6. [PMID: 26623969 DOI: 10.1165/rcmb.2015-0100le] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kara N Goss
- 1 University of Wisconsin School of Medicine and Public Health Madison, Wisconsin
| | - Robert S Tepper
- 2 Indiana University School of Medicine Indianapolis, Indiana
| | - Tim Lahm
- 2 Indiana University School of Medicine Indianapolis, Indiana.,3 Richard L Roudebush VA Medical Center Indianapolis, Indiana
| | | |
Collapse
|
43
|
Li J, Zhang L, Zhang Y, Liu Y, Zhang H, Wei L, Shen T, Jiang C, Zhu D. A20 deficiency leads to angiogenesis of pulmonary artery endothelial cells through stronger NF-κB activation under hypoxia. J Cell Mol Med 2016; 20:1319-28. [PMID: 26991692 PMCID: PMC4929300 DOI: 10.1111/jcmm.12816] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 01/17/2016] [Indexed: 12/11/2022] Open
Abstract
A20 is a zinc finger protein associated with hypoxia. As chronic hypoxia is responsible for intimal hyperplasia and disordered angiogenesis of pulmonary artery, which are histological hallmarks of pulmonary artery hypertension, we intended to explore the role of A20 in angiogenesis of pulmonary artery endothelial cells (ECs). Here, we found a transient elevation of A20 expression in the lung tissues from hypoxic rats compared with normoxic controls. This rapid enhancement was mainly detected in the endothelium, and similar results were reproduced in vitro. During early hypoxia, genetic inhibition of A20 increased proliferation in pulmonary artery ECs, linking to advanced cell cycle progression as well as microtubule polymerization, and aggravated angiogenic effects including tube formation, cell migration and adhesion molecules expression. In addition, a negative feedback loop between nuclear factor-kappa B and A20 was confirmed. Our findings provide evidence for an adaptive role of A20 against pulmonary artery ECs angiogenesis via nuclear factor-kappa B activation.
Collapse
Affiliation(s)
- Jing Li
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Linlin Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China
| | - Yueming Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Liu
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhang
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liuping Wei
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tingting Shen
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chun Jiang
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Daling Zhu
- Department of Biopharmaceutical Sciences, Harbin Medical University, Daqing, Heilongjiang, China.,Department of Biopharmaceutical Key Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
44
|
Rowan SC, Keane MP, Gaine S, McLoughlin P. Hypoxic pulmonary hypertension in chronic lung diseases: novel vasoconstrictor pathways. THE LANCET RESPIRATORY MEDICINE 2016; 4:225-36. [PMID: 26895650 DOI: 10.1016/s2213-2600(15)00517-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 11/29/2022]
Abstract
Pulmonary hypertension is a well recognised complication of chronic hypoxic lung diseases, which are among the most common causes of death and disability worldwide. Development of pulmonary hypertension independently predicts reduced life expectancy. In chronic obstructive pulmonary disease, long-term oxygen therapy ameliorates pulmonary hypertension and greatly improves survival, although the correction of alveolar hypoxia and pulmonary hypertension is only partial. Advances in understanding of the regulation of vascular smooth muscle tone show that chronic vasoconstriction plays a more important part in the pathogenesis of hypoxic pulmonary hypertension than previously thought, and that structural vascular changes contribute less. Trials of existing vasodilators show that pulmonary hypertension can be ameliorated and systemic oxygen delivery improved in carefully selected patients, although systemic hypotensive effects limit the doses used. Vasoconstrictor pathways that are selective for the pulmonary circulation can be blocked to reduce hypoxic pulmonary hypertension without causing systemic hypotension, and thus provide potential targets for novel therapeutic strategies.
Collapse
Affiliation(s)
- Simon C Rowan
- UCD School of Medicine, Conway Institute, Dublin, Ireland
| | - Michael P Keane
- UCD School of Medicine, Respiratory Medicine, St Vincent's University Hospital, Dublin, Ireland
| | - Seán Gaine
- National Pulmonary Hypertension Unit, Mater Misericordiae University Hospital, Dublin, Ireland
| | | |
Collapse
|
45
|
Mitofusin 2 Downregulation Triggers Pulmonary Artery Smooth Muscle Cell Proliferation and Apoptosis Imbalance in Rats With Hypoxic Pulmonary Hypertension Via the PI3K/Akt and Mitochondrial Apoptosis Pathways. J Cardiovasc Pharmacol 2016; 67:164-74. [DOI: 10.1097/fjc.0000000000000333] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
46
|
REST mediates resolution of HIF-dependent gene expression in prolonged hypoxia. Sci Rep 2015; 5:17851. [PMID: 26647819 PMCID: PMC4673454 DOI: 10.1038/srep17851] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/03/2015] [Indexed: 01/24/2023] Open
Abstract
The hypoxia-inducible factor (HIF) is a key regulator of the cellular response to hypoxia which promotes oxygen delivery and metabolic adaptation to oxygen deprivation. However, the degree and duration of HIF-1α expression in hypoxia must be carefully balanced within cells in order to avoid unwanted side effects associated with excessive activity. The expression of HIF-1α mRNA is suppressed in prolonged hypoxia, suggesting that the control of HIF1A gene transcription is tightly regulated by negative feedback mechanisms. Little is known about the resolution of the HIF-1α protein response and the suppression of HIF-1α mRNA in prolonged hypoxia. Here, we demonstrate that the Repressor Element 1-Silencing Transcription factor (REST) binds to the HIF-1α promoter in a hypoxia-dependent manner. Knockdown of REST using RNAi increases the expression of HIF-1α mRNA, protein and transcriptional activity. Furthermore REST knockdown increases glucose consumption and lactate production in a HIF-1α- (but not HIF-2α-) dependent manner. Finally, REST promotes the resolution of HIF-1α protein expression in prolonged hypoxia. In conclusion, we hypothesize that REST represses transcription of HIF-1α in prolonged hypoxia, thus contributing to the resolution of the HIF-1α response.
Collapse
|
47
|
Veith C, Schermuly RT, Brandes RP, Weissmann N. Molecular mechanisms of hypoxia-inducible factor-induced pulmonary arterial smooth muscle cell alterations in pulmonary hypertension. J Physiol 2015; 594:1167-77. [PMID: 26228924 DOI: 10.1113/jp270689] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/28/2015] [Indexed: 12/18/2022] Open
Abstract
Oxygen (O2) is essential for the viability and function of most metazoan organisms and thus is closely monitored at both the organismal and the cellular levels. However, alveoli often encounter decreased O2 levels (hypoxia), leading to activation of physiological or pathophysiological responses in the pulmonary arteries. Such changes are achieved by activation of transcription factors. The hypoxia-inducible factors (HIFs) are the most prominent hypoxia-regulated transcription factors in this regard. HIFs bind to hypoxia-response elements (HREs) in the promoter region of target genes, whose expression and translation allows the organism, amongst other factors, to cope with decreased environmental O2 partial pressure (pO2). However, prolonged HIF activation can contribute to major structural alterations, especially in the lung, resulting in the development of pulmonary hypertension (PH). PH is characterized by a rise in pulmonary arterial pressure associated with pulmonary arterial remodelling, concomitant with a reduced intravascular lumen area. Patients with PH develop right heart hypertrophy and eventually die from right heart failure. Thus, understanding the molecular mechanisms of HIF regulation in PH is critical for the identification of novel therapeutic strategies. This review addresses the relationship of hypoxia and the HIF system with pulmonary arterial dysfunction in PH. We particularly focus on the cellular and molecular mechanisms underlying the HIF-driven pathophysiological processes.
Collapse
Affiliation(s)
- Christine Veith
- Universities of Giessen and Marburg Lung Centre (UGMLC), member of the German Centre for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary System (ECCPS), 35392, Giessen, Germany
| | - Ralph T Schermuly
- Universities of Giessen and Marburg Lung Centre (UGMLC), member of the German Centre for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary System (ECCPS), 35392, Giessen, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, ECCPS, 60590, Frankfurt, Germany
| | - Norbert Weissmann
- Universities of Giessen and Marburg Lung Centre (UGMLC), member of the German Centre for Lung Research (DZL), Excellence Cluster Cardio-Pulmonary System (ECCPS), 35392, Giessen, Germany
| |
Collapse
|
48
|
Mirzaei Bavil F, Alipour MR, Keyhanmanesh R, Alihemmati A, Ghiyasi R, Mohaddes G. Ghrelin Decreases Angiogenesis, HIF-1α and VEGF Protein Levels in Chronic Hypoxia in Lung Tissue of Male Rats. Adv Pharm Bull 2015; 5:315-20. [PMID: 26504752 DOI: 10.15171/apb.2015.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/14/2014] [Accepted: 10/18/2014] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Hypoxia is a condition of decreased availability of oxygen. When cells are exposed to a low oxygen environment, they impel the hypoxia responses to adapt to new situation. The hypoxia response leads to the activation of various cellular signaling pathways. The aim of this study was to evaluate the effect of ghrelin on angiogenesis, Hypoxia-Inducible-Factor-1α (HIF-1) and Vascular endothelial growth factor (VEGF) levels in normobaric hypoxia situation. METHODS Twenty four animals were divided into 4 groups (n=6): control (C), ghrelin (Gh), hypoxia (H), and hypoxic animals that received ghrelin (H+Gh). Hypoxia (11%) was induced by an Environmental Chamber System GO2 Altitude. Animals in ghrelin groups received a subcutaneous injection of ghrelin (150 μg/kg/day) for 14 days. RESULTS Our results showed that hypoxia significantly (p<0.05) increased angiogenesis without any significant changes on HIF-1 and VEGF levels, whereas ghrelin significantly (p<0.05) decreased angiogenesis, expression of HIF-1 and VEGF in this condition. Ghrelin administration did not show any significant changes in normal conditions. CONCLUSION Ghrelin had no effect on angiogenesis, expression of HIF-1 and VEGF in normal oxygen conditions but it reduced angiogenesis process in lung tissue with reducing the level of HIF and VEGF in hypoxic condition. Therefore, effect of ghrelin on angiogenesis could be related to blood oxygen level.
Collapse
Affiliation(s)
- Fariba Mirzaei Bavil
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alipour
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rafigheh Ghiyasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gisou Mohaddes
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Nishimura R, Nishiwaki T, Kawasaki T, Sekine A, Suda R, Urushibara T, Suzuki T, Takayanagi S, Terada J, Sakao S, Tatsumi K. Hypoxia-induced proliferation of tissue-resident endothelial progenitor cells in the lung. Am J Physiol Lung Cell Mol Physiol 2015; 308:L746-58. [PMID: 25502500 DOI: 10.1152/ajplung.00243.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/05/2014] [Indexed: 11/22/2022] Open
Abstract
Exposure to hypoxia induces changes in the structure and functional phenotypes of the cells composing the pulmonary vascular wall from larger to most peripheral vessels. Endothelial progenitor cells (EPCs) may be involved in vascular endothelial repair. Resident EPCs with a high proliferative potential are found in the pulmonary microcirculation. However, their potential location, identification, and functional role have not been clearly established. We investigated whether resident EPCs or bone marrow (BM)-derived EPCs play a major role in hypoxic response of pulmonary vascular endothelial cells (PVECs). Mice were exposed to hypoxia. The number of PVECs transiently decreased followed by an increase in hypoxic animals. Under hypoxic conditions for 1 wk, prominent bromodeoxyuridine incorporation was detected in PVECs. Some Ki67-positive cells were detected among PVECs after 1 wk under hypoxic conditions, especially in the capillaries. To clarify the origin of proliferating endothelial cells, we used BM chimeric mice expressing green fluorescent protein (GFP). The percentage of GFP-positive PVECs was low and constant during hypoxia in BM-transplanted mice, suggesting little engraftment of BM-derived cells in lungs under hypoxia. Proliferating PVECs in hypoxic animals showed increased expression of CD34, suggesting hypoxia-induced gene expression and cell surface antigen of EPC or stem/progenitor cells markers. Isolated PVECs from hypoxic mice showed colony- and tube-forming capacity. The present study indicated that hypoxia could induce proliferation of PVECs, and the origin of these cells might be tissue-resident EPCs.
Collapse
Affiliation(s)
- Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsu Nishiwaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Urushibara
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin Takayanagi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
50
|
Affiliation(s)
- P. McLoughlin
- University College Dublin School of Medicine and Medical Sciences Conway Institute Dublin Ireland
| |
Collapse
|