1
|
Yarboro MT, Boatwright N, Sekulich DC, Hooper CW, Wong T, Poole SD, Berger CD, Brown AJ, Jetter CS, Sucre JMS, Shelton EL, Reese J. A novel role for PGE 2-EP 4 in the developmental programming of the mouse ductus arteriosus: consequences for vessel maturation and function. Am J Physiol Heart Circ Physiol 2023; 325:H687-H701. [PMID: 37566109 PMCID: PMC10643004 DOI: 10.1152/ajpheart.00294.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023]
Abstract
The ductus arteriosus (DA) is a vascular shunt that allows oxygenated blood to bypass the developing lungs in utero. Fetal DA patency requires vasodilatory signaling via the prostaglandin E2 (PGE2) receptor EP4. However, in humans and mice, disrupted PGE2-EP4 signaling in utero causes unexpected patency of the DA (PDA) after birth, suggesting another role for EP4 during development. We used EP4-knockout (KO) mice and acute versus chronic pharmacological approaches to investigate EP4 signaling in DA development and function. Expression analyses identified EP4 as the primary EP receptor in the DA from midgestation to term; inhibitor studies verified EP4 as the primary dilator during this period. Chronic antagonism recapitulated the EP4 KO phenotype and revealed a narrow developmental window when EP4 stimulation is required for postnatal DA closure. Myography studies indicate that despite reduced contractile properties, the EP4 KO DA maintains an intact oxygen response. In newborns, hyperoxia constricted the EP4 KO DA but survival was not improved, and permanent remodeling was disrupted. Vasomotion and increased nitric oxide (NO) sensitivity in the EP4 KO DA suggest incomplete DA development. Analysis of DA maturity markers confirmed a partially immature EP4 KO DA phenotype. Together, our data suggest that EP4 signaling in late gestation plays a key developmental role in establishing a functional term DA. When disrupted in EP4 KO mice, the postnatal DA exhibits signaling and contractile properties characteristic of an immature DA, including impairments in the first, muscular phase of DA closure, in addition to known abnormalities in the second permanent remodeling phase.NEW & NOTEWORTHY EP4 is the primary EP receptor in the ductus arteriosus (DA) and is critical during late gestation for its development and eventual closure. The "paradoxical" patent DA (PDA) phenotype of EP4-knockout mice arises from a combination of impaired contractile potential, altered signaling properties, and a failure to remodel associated with an underdeveloped immature vessel. These findings provide new mechanistic insights into women who receive NSAIDs to treat preterm labor, whose infants have unexplained PDA.
Collapse
Affiliation(s)
- Michael T Yarboro
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| | - Naoko Boatwright
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Deanna C Sekulich
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Christopher W Hooper
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ting Wong
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Stanley D Poole
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Courtney D Berger
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Alexus J Brown
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Christopher S Jetter
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer M S Sucre
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Elaine L Shelton
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Jeff Reese
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| |
Collapse
|
2
|
Bentley RET, Hindmarch CCT, Archer SL. Using omics to breathe new life into our understanding of the ductus arteriosus oxygen response. Semin Perinatol 2023; 47:151715. [PMID: 36906478 DOI: 10.1016/j.semperi.2023.151715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
Abstract
The ductus arteriosus (DA) connects the aorta to the pulmonary artery (PA), directing placentally oxygenated blood away from the developing lungs. High pulmonary vascular resistance and low systemic vascular resistance facilitate shunting of blood in utero from the pulmonary to the systemic circulation through the widely patent DA, thereby optimizing fetal oxygen (O2) delivery. With the transition from fetal (hypoxia) to neonatal (normoxia) oxygen conditions, the DA constricts while the PA dilates. This process often fails in prematurity, promoting congenital heart disease. Impaired O2-responsivness in the DA promotes persistent ductus arteriosus (PDA), the most common form of congenital heart disease. Knowledge of DA oxygen sensing has greatly advanced in the past few decades, however we still lack a complete understanding of the sensing mechanism. The genomic revolution of the past two decades has facilitated unprecedented discovery in every biological system. This review will demonstrate how multiomic integration of data generated from the DA can breathe new life into our understanding of the DA's oxygen response.
Collapse
Affiliation(s)
| | - Charles C T Hindmarch
- QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada; Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen L Archer
- QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada; Department of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
3
|
Bentley RET, Hindmarch CCT, Dunham-Snary KJ, Snetsinger B, Mewburn JD, Thébaud A, Lima PDA, Thébaud B, Archer SL. The molecular mechanisms of oxygen-sensing in human ductus arteriosus smooth muscle cells: A comprehensive transcriptome profile reveals a central role for mitochondria. Genomics 2021; 113:3128-3140. [PMID: 34245829 PMCID: PMC10659099 DOI: 10.1016/j.ygeno.2021.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/23/2021] [Accepted: 07/04/2021] [Indexed: 01/22/2023]
Abstract
The ductus arteriosus (DA) connects the fetal pulmonary artery and aorta, diverting placentally oxygenated blood from the developing lungs to the systemic circulation. The DA constricts in response to increases in oxygen (O2) with the first breaths, resulting in functional DA closure, with anatomic closure occurring within the first days of life. Failure of DA closure results in persistent patent ductus arteriosus (PDA), a common complication of extreme preterm birth. The DA's response to O2, though modulated by the endothelium, is intrinsic to the DA smooth muscle cells (DASMC). DA constriction is mediated by mitochondrial-derived reactive oxygen species, which increase in proportion to arterial partial pressure of oxygen (PaO2). The resulting redox changes inhibit voltage-gated potassium channels (Kv) leading to cell depolarization, calcium influx and DASMC constriction. To date, there has not been an unbiased assessment of the human DA O2-sensors using transcriptomics, nor are there known molecular mechanisms which characterize DA closure. DASMCs were isolated from DAs obtained from 10 term infants at the time of congenital heart surgery. Cells were purified by flow cytometry, negatively sorting using CD90 and CD31 to eliminate fibroblasts or endothelial cells, respectively. The purity of the DASMC population was confirmed by positive staining for α-smooth muscle actin, smoothelin B and caldesmon. Cells were grown for 96 h in hypoxia (2.5% O2) or normoxia (19% O2) and confocal imaging with Cal-520 was used to determine oxygen responsiveness. An oxygen-induced increase in intracellular calcium of 18.1% ± 4.4% and SMC constriction (-27% ± 1.5% shortening) occurred in all cell lines within five minutes. RNA sequencing of the cells grown in hypoxia and normoxia revealed significant regulation of 1344 genes (corrected p < 0.05). We examined these genes using Gene Ontology (GO). This unbiased assessment of altered gene expression indicated significant enrichment of the following GOterms: mitochondria, cellular respiration and transcription. The top regulated biologic process was generation of precursor metabolites and energy. The top regulated cellular component was mitochondrial matrix. The top regulated molecular function was transcription coactivator activity. Multiple members of the NADH-ubiquinone oxidoreductase (NDUF) family are upregulated in human DASMC (hDASMC) following normoxia. Several of our differentially regulated transcripts are encoded by genes that have been associated with genetic syndromes that have an increased incidence of PDA (Crebb binding protein and Histone Acetyltransferase P300). This first examination of the effects of O2 on human DA transcriptomics supports a putative role for mitochondria as oxygen sensors.
Collapse
Affiliation(s)
| | - Charles C T Hindmarch
- Department of Medicine, Queen's University, Kingston, Ontario, Canada; QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada
| | - Kimberly J Dunham-Snary
- Department of Medicine, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Science, Queen's University, Canada
| | - Brooke Snetsinger
- QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada
| | - Jeffrey D Mewburn
- Department of Biomedical and Molecular Science, Queen's University, Canada
| | - Arthur Thébaud
- Department of Kinesiology and Health Studies, Queen's University, Canada
| | - Patricia D A Lima
- QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada
| | - Bernard Thébaud
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, Ontario, Canada; Department of Pediatrics, Division of Neonatology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada; Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada; QCPU, Queen's Cardiopulmonary Unit, Translational Institute of Medicine (TIME), Department of Medicine, Queen's University, Canada.
| |
Collapse
|
4
|
Yanagisawa H, Yokoyama U. Extracellular matrix-mediated remodeling and mechanotransduction in large vessels during development and disease. Cell Signal 2021; 86:110104. [PMID: 34339854 DOI: 10.1016/j.cellsig.2021.110104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 01/08/2023]
Abstract
The vascular extracellular matrix (ECM) is synthesized and secreted during embryogenesis and facilitates the growth and remodeling of large vessels. Proper interactions between the ECM and vascular cells are pivotal for building the vasculature required for postnatal dynamic circulation. The ECM serves as a structural component by maintaining the integrity of the vessel wall while also regulating intercellular signaling, which involves cytokines and growth factors. The major ECM component in large vessels is elastic fibers, which include elastin and microfibrils. Elastin is predominantly synthesized by vascular smooth muscle cells (SMCs) and uses microfibrils as a scaffold to lay down and assemble cross-linked elastin. The absence of elastin causes developmental defects that result in the subendothelial proliferation of SMCs and inward remodeling of the vessel wall. Notably, elastic fiber formation is attenuated in the ductus arteriosus and umbilical arteries. These two vessels function during embryogenesis and close after birth via cellular proliferation, migration, and matrix accumulation. In dynamic postnatal mechano-environments, the elastic fibers in large vessels also serve an essential role in proper signal transduction as a component of elastin-contractile units. Disrupted mechanotransduction in SMCs leads to pathological conditions such as aortic aneurysms that exhibit outward remodeling. This review discusses the importance of the ECM-mainly the elastic fiber matrix-in large vessels during developmental remodeling and under pathological conditions. By dissecting the role of the ECM in large vessels, we aim to provide insights into the role of ECM-mediated signal transduction that can provide a basis for seeking new targets for intervention in vascular diseases.
Collapse
Affiliation(s)
- Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, The University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| |
Collapse
|
5
|
Saito J, Kojima T, Tanifuji S, Kato Y, Oka S, Ichikawa Y, Miyagi E, Tachibana T, Asou T, Yokoyama U. Transcriptome Analysis Reveals Differential Gene Expression between the Closing Ductus Arteriosus and the Patent Ductus Arteriosus in Humans. J Cardiovasc Dev Dis 2021; 8:jcdd8040045. [PMID: 33923468 PMCID: PMC8073410 DOI: 10.3390/jcdd8040045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
The ductus arteriosus (DA) immediately starts closing after birth. This dynamic process involves DA-specific properties, including highly differentiated smooth muscle, sparse elastic fibers, and intimal thickening (IT). Although several studies have demonstrated DA-specific gene expressions using animal tissues and human fetuses, the transcriptional profiles of the closing DA and the patent DA remain largely unknown. We performed transcriptome analysis using four human DA samples. The three closing DA samples exhibited typical DA morphology, but the patent DA exhibited aorta-like elastic lamellae and poorly formed IT. A cluster analysis revealed that samples were clearly divided into two major clusters, the closing DA and patent DA clusters, and showed distinct gene expression profiles in IT and the tunica media of the closing DA samples. Cardiac neural crest-related genes such as JAG1 were highly expressed in the tunica media and IT of the closing DA samples compared to the patent DA sample. Abundant protein expressions of jagged 1 and the differentiated smooth muscle marker calponin were observed in the closing DA samples but not in the patent DA sample. Second heart field-related genes such as ISL1 were enriched in the patent DA sample. These data indicate that the patent DA may have different cell lineages compared to the closing DA.
Collapse
Affiliation(s)
- Junichi Saito
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Tomoyuki Kojima
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Shota Tanifuji
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yuko Kato
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Sayuki Oka
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yasuhiro Ichikawa
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Tsuyoshi Tachibana
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Toshihide Asou
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Correspondence: ; Tel.: +81-3-3351-6141
| |
Collapse
|
6
|
Hamrick SEG, Sallmon H, Rose AT, Porras D, Shelton EL, Reese J, Hansmann G. Patent Ductus Arteriosus of the Preterm Infant. Pediatrics 2020; 146:e20201209. [PMID: 33093140 PMCID: PMC7605084 DOI: 10.1542/peds.2020-1209] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Postnatal ductal closure is stimulated by rising oxygen tension and withdrawal of vasodilatory mediators (prostaglandins, nitric oxide, adenosine) and by vasoconstrictors (endothelin-1, catecholamines, contractile prostanoids), ion channels, calcium flux, platelets, morphologic maturity, and a favorable genetic predisposition. A persistently patent ductus arteriosus (PDA) in preterm infants can have clinical consequences. Decreasing pulmonary vascular resistance, especially in extremely low gestational age newborns, increases left-to-right shunting through the ductus and increases pulmonary blood flow further, leading to interstitial pulmonary edema and volume load to the left heart. Potential consequences of left-to-right shunting via a hemodynamically significant patent ductus arteriosus (hsPDA) include increased risk for prolonged ventilation, bronchopulmonary dysplasia, necrotizing enterocolitis or focal intestinal perforation, intraventricular hemorrhage, and death. In the last decade, there has been a trend toward less aggressive treatment of PDA in preterm infants. However, there is a subgroup of infants who will likely benefit from intervention, be it pharmacologic, interventional, or surgical: (1) prophylactic intravenous indomethacin in highly selected extremely low gestational age newborns with PDA (<26 + 0/7 weeks' gestation, <750 g birth weight), (2) early targeted therapy of PDA in selected preterm infants at particular high risk for PDA-associated complications, and (3) PDA ligation, catheter intervention, or oral paracetamol may be considered as rescue options for hsPDA closure. The impact of catheter-based closure of hsPDA on clinical outcomes should be determined in future prospective studies. Finally, we provide a novel treatment algorithm for PDA in preterm infants that integrates the several treatment modalities in a staged approach.
Collapse
Affiliation(s)
- Shannon E G Hamrick
- Divisions of Neonatology and
- Cardiology, Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Hannes Sallmon
- Department of Pediatric Cardiology, Charité University Medical Center, Berlin, Germany
| | | | - Diego Porras
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Elaine L Shelton
- Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Jeff Reese
- Division of Neonatology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| |
Collapse
|
7
|
Ito S, Yokoyama U, Nakakoji T, Cooley MA, Sasaki T, Hatano S, Kato Y, Saito J, Nicho N, Iwasaki S, Umemura M, Fujita T, Masuda M, Asou T, Ishikawa Y. Fibulin-1 Integrates Subendothelial Extracellular Matrices and Contributes to Anatomical Closure of the Ductus Arteriosus. Arterioscler Thromb Vasc Biol 2020; 40:2212-2226. [PMID: 32640908 PMCID: PMC7447190 DOI: 10.1161/atvbaha.120.314729] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The ductus arteriosus (DA) is a fetal artery connecting the aorta and pulmonary arteries. Progressive matrix remodeling, that is, intimal thickening (IT), occurs in the subendothelial region of DA to bring anatomic DA closure. IT is comprised of multiple ECMs (extracellular matrices) and migrated smooth muscle cells (SMCs). Because glycoprotein fibulin-1 binds to multiple ECMs and regulates morphogenesis during development, we investigated the role of fibulin-1 in DA closure. Approach and Results: Fibulin-1-deficient (Fbln1-/-) mice exhibited patent DA with hypoplastic IT. An unbiased transcriptome analysis revealed that EP4 (prostaglandin E receptor 4) stimulation markedly increased fibulin-1 in DA-SMCs via phospholipase C-NFκB (nuclear factor κB) signaling pathways. Fluorescence-activated cell sorting (FACS) analysis demonstrated that fibulin-1 binding protein versican was derived from DA-endothelial cells (ECs). We examined the effect of fibulin-1 on directional migration toward ECs in association with versican by using cocultured DA-SMCs and ECs. EP4 stimulation promoted directional DA-SMC migration toward ECs, which was attenuated by either silencing fibulin-1 or versican. Immunofluorescence demonstrated that fibulin-1 and versican V0/V1 were coexpressed at the IT of wild-type DA, whereas 30% of versican-deleted mice lacking a hyaluronan binding site displayed patent DA. Fibulin-1 expression was attenuated in the EP4-deficient mouse (Ptger4-/-) DA, which exhibits patent DA with hypoplastic IT, and fibulin-1 protein administration restored IT formation. In human DA, fibulin-1 and versican were abundantly expressed in SMCs and ECs, respectively. CONCLUSIONS Fibulin-1 contributes to DA closure by forming an environment favoring directional SMC migration toward the subendothelial region, at least, in part, in combination with EC-derived versican and its binding partner hyaluronan.
Collapse
Affiliation(s)
- Satoko Ito
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan.,Department of Physiology, Tokyo Medical University, Japan (S.I., U.Y., Y.K., J.S.)
| | - Utako Yokoyama
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan.,Department of Physiology, Tokyo Medical University, Japan (S.I., U.Y., Y.K., J.S.)
| | - Taichi Nakakoji
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Marion A Cooley
- Department of Oral Biology and Diagnostic Sciences, Augusta University, GA (M.A.C.)
| | - Takako Sasaki
- Department of Biochemistry II, Oita University, Japan (T.S.)
| | - Sonoko Hatano
- Institute for Molecular Science of Medicine, Aichi Medical University, Japan (S.H.)
| | - Yuko Kato
- Department of Physiology, Tokyo Medical University, Japan (S.I., U.Y., Y.K., J.S.)
| | - Junichi Saito
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan.,Department of Physiology, Tokyo Medical University, Japan (S.I., U.Y., Y.K., J.S.)
| | - Naoki Nicho
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Shiho Iwasaki
- Department of Pediatrics (S.I.), Yokohama City University, Japan
| | - Masanari Umemura
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Takayuki Fujita
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan
| | - Munetaka Masuda
- Department of Surgery (M.M.), Yokohama City University, Japan
| | - Toshihide Asou
- Department of Cardiovascular Surgery, Kanagawa Children's Medical Center, Yokohama, Japan (T.A.)
| | - Yoshihiro Ishikawa
- From the Cardiovascular Research Institute (S.I., U.Y., T.N., J.S., N.N., M.U., T.F., Y.I.), Yokohama City University, Japan
| |
Collapse
|
8
|
Akaike T, Shinjo S, Ohmori E, Kajimura I, Goda N, Minamisawa S. Transcriptional profiles in the chicken ductus arteriosus during hatching. PLoS One 2019; 14:e0214139. [PMID: 30897181 PMCID: PMC6428269 DOI: 10.1371/journal.pone.0214139] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/07/2019] [Indexed: 02/08/2023] Open
Abstract
The ductus arteriosus, an essential embryonic blood vessel between the pulmonary artery and the descending aorta, constricts after birth or hatching and eventually closes to terminate embryonic circulation. Chicken embryos have two long ductus arteriosi, which anatomically differ from mammal ductus arteriosus. Each long ductus arteriosus is divided into two parts: the pulmonary artery-sided and descending aorta-sided ductus arteriosi. Although the pulmonary artery-sided and descending aorta-sided ductus arteriosi have distinct functional characteristics, such as oxygen responsiveness, the difference in their transcriptional profiles has not been investigated. We performed a DNA microarray analysis (GSE 120116 at NCBI GEO) with pooled tissues from the chicken pulmonary artery-sided ductus arteriosus, descending aorta-sided ductus arteriosus, and aorta at the internal pipping stage. Although several known ductus arteriosus-dominant genes such as tfap2b were highly expressed in the pulmonary artery-sided ductus arteriosus, we newly found genes that were dominantly expressed in the chicken pulmonary artery-sided ductus arteriosus. Interestingly, cluster analysis showed that the expression pattern of the pulmonary artery-sided ductus arteriosus was closer to that of the descending aorta-sided ductus arteriosus than that of the aorta, whereas the morphology of the descending aorta-sided ductus arteriosus was closer to that of the aorta than that of the pulmonary artery-sided ductus arteriosus. Subsequent pathway analysis with DAVID bioinformatics resources revealed that the pulmonary artery-sided ductus arteriosus showed enhanced expression of the genes involved in melanogenesis and tyrosine metabolism compared with the descending aorta-sided ductus arteriosus, suggesting that tyrosinase and the related genes play an important role in the proper differentiation of neural crest-derived cells during vascular remodeling in the ductus arteriosus. In conclusion, the transcription profiles of the chicken ductus arteriosus provide new insights for investigating the mechanism of ductus arteriosus closure.
Collapse
Affiliation(s)
- Toru Akaike
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Satoko Shinjo
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Eriko Ohmori
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Ichige Kajimura
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Nobuhito Goda
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| |
Collapse
|
9
|
Hou HT, Xi-Zhang, Wang J, Liu LX, Zhang JF, Yang Q, He GW. Altered plasma proteins released from platelets and endothelial cells are associated with human patent ductus arteriosus. J Cell Physiol 2018; 234:6842-6853. [PMID: 30480800 DOI: 10.1002/jcp.27433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/27/2018] [Indexed: 11/09/2022]
Abstract
Patent ductus arteriosus is the third most common congenital heart disease and resulted from the persistence of ductal patency after birth. Ductus arteriosus closure involves functional and structural remodeling, controlled by many factors. The changes in plasma protein levels associated with PDA closure are not known. Here we for the first time demonstrate six key differential plasma proteins in human patent ductus arteriosus patients using proteomic technology and present a model to illustrate the constriction and closure of ductus arteriosus. Differentially expressed proteins were analyzed by using isobaric tags for relative and absolute quantification and validated by enzyme-linked immunosorbent assay in new samples. The proteomic data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD008568. We found 74 upregulated and 98 downregulated proteins in the plasma of patients with PDA. Five decreased proteins (platelet factor 4, fibrinogen, von Willebrand factor, collagen, and mannose binding lectin-associated serine protease-2) and one increased protein (fibronectin) may increase the risk of patent ductus arteriosus. Those proteins are closely related to platelet activation and coagulation cascades, complement mannan-binding-lectin, and other systemic signaling pathways. Our findings for the first time indicate that the differential proteins involved in different pathways may play key roles in the nonclosure of the ductus arteriosus in humans and may be developed as biomarkers for diagnosis. All those findings may be served as the basis of understanding the etiology and pathogenesis of patent ductus arteriosus.
Collapse
Affiliation(s)
- Hai-Tao Hou
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,The Heart Center, The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China
| | - Xi-Zhang
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Wang
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Li-Xin Liu
- Department of Pediatric Cardiothoracic Surgery, Maternal and Child Health Hospital of Tangshan, Hebei, China
| | - Jian-Feng Zhang
- Department of Pediatric Cardiothoracic Surgery, Maternal and Child Health Hospital of Tangshan, Hebei, China
| | - Qin Yang
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Department of Cardiovascular Surgery & Center for Basic Medical Research, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,The Heart Center, The Affiliated Hospital of Hangzhou Normal University & Zhejiang University, Hangzhou, China.,Department of Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
10
|
Lewis TR, Shelton EL, Van Driest SL, Kannankeril PJ, Reese J. Genetics of the patent ductus arteriosus (PDA) and pharmacogenetics of PDA treatment. Semin Fetal Neonatal Med 2018; 23:232-238. [PMID: 29510900 PMCID: PMC6098727 DOI: 10.1016/j.siny.2018.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patent ductus arteriosus (PDA) is a frequent, complex, and difficult to treat clinical syndrome among preterm infants in the neonatal intensive care unit. In addition to known clinical risk factors, there are emerging data about genetic predisposition to PDA in both animal and human models. Clinical response and toxicity from drugs used to treat PDA are highly variable. Developmental and genetic aspects of pharmacokinetics and pharmacodynamics influence exposure and response to pharmacologic therapies. Given the variable efficacy and toxicity of known drug therapies, novel therapeutic targets for PDA treatment offer the promise of precision medicine. This review addresses the known genetic contributions to prolonged ductal patency, variability in response to drug therapy for PDA, and potential novel drug targets for future PDA treatment discovery.
Collapse
Affiliation(s)
- Tamorah R Lewis
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, University of Missouri, Kansas City School of Medicine, Kansas City, MO, USA
| | - Elaine L Shelton
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara L Van Driest
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Prince J Kannankeril
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeff Reese
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
11
|
Yarboro MT, Durbin MD, Herington JL, Shelton EL, Zhang T, Ebby CG, Stoller JZ, Clyman RI, Reese J. Transcriptional profiling of the ductus arteriosus: Comparison of rodent microarrays and human RNA sequencing. Semin Perinatol 2018; 42:212-220. [PMID: 29910032 PMCID: PMC6064668 DOI: 10.1053/j.semperi.2018.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
DA closure is crucial for the transition from fetal to neonatal life. This closure is supported by changes to the DA's signaling and structural properties that distinguish it from neighboring vessels. Examining transcriptional differences between these vessels is key to identifying genes or pathways responsible for DA closure. Several microarray studies have explored the DA transcriptome in animal models but varied experimental designs have led to conflicting results. Thorough transcriptomic analysis of the human DA has yet to be performed. A clear picture of the DA transcriptome is key to guiding future research endeavors, both to allow more targeted treatments in the clinical setting, and to understand the basic biology of DA function. In this review, we use a cross-species cross-platform analysis to consider all available published rodent microarray data and novel human RNAseq data in order to provide high priority candidate genes for consideration in future DA studies.
Collapse
Affiliation(s)
- Michael T. Yarboro
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Matthew D. Durbin
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202
| | - Jennifer L. Herington
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | - Elaine L. Shelton
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232,Department of Pharmacology, Vanderbilt University, Nashville, TN 37232
| | - Tao Zhang
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Cris G. Ebby
- Rutgers New Jersey Medical School, Newark, NJ 08901
| | - Jason Z. Stoller
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Ronald I. Clyman
- Department of Pediatrics, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143
| | - Jeff Reese
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Vanderbilt University, 1125 Light Hall/MRB IV Bldg., 2215 B Garland Ave., Nashville, TN 37232; Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232.
| |
Collapse
|
12
|
Shelton EL, Singh GK, Nichols CG. Novel drug targets for ductus arteriosus manipulation: Looking beyond prostaglandins. Semin Perinatol 2018; 42:221-227. [PMID: 29880312 PMCID: PMC6064654 DOI: 10.1053/j.semperi.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Forty years ago, non-steroidal anti-inflammatory drugs were first reported to decrease systemic prostaglandin levels and promote ductus arteriosus (DA) closure. And yet, prolonged patency of the DA (PDA) remains a significant clinical problem, complicated by imperfect therapies and wide variations in treatment strategy. There are few pharmacology-based tools available for treating PDA (indomethacin, ibuprofen, and acetaminophen), or for maintaining DA patency (PGE1) as is needed to facilitate corrective surgery for ductus-dependent congenital heart defects. Unfortunately, all of these treatments are inefficient and are associated with concerning adverse effects. This review highlights novel potential DA drug targets that may expand our therapeutic repertoire beyond the prostaglandin pathway.
Collapse
Affiliation(s)
- Elaine L. Shelton
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt and Vanderbilt University
Medical Center, Nashville, Tennessee,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Gautam K. Singh
- Department of Pediatrics, Washington University School of Medicine, Saint Louis Children's Hospital, Saint
Louis, Missouri
| | - Colin G. Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
13
|
Kumar A, D'Souza SS, Moskvin OV, Toh H, Wang B, Zhang J, Swanson S, Guo LW, Thomson JA, Slukvin II. Specification and Diversification of Pericytes and Smooth Muscle Cells from Mesenchymoangioblasts. Cell Rep 2018; 19:1902-1916. [PMID: 28564607 PMCID: PMC6428685 DOI: 10.1016/j.celrep.2017.05.019] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
Elucidating the pathways that lead to vasculogenic cells, and being able to identify their progenitors and lineage-restricted cells, is critical to the establishment of human pluripotent stem cell (hPSC) models for vascular diseases and development of vascular therapies. Here, we find that mesoderm-derived pericytes (PCs) and smooth muscle cells (SMCs) originate from a clonal mesenchymal progenitor mesenchymoangioblast (MB). In clonogenic cultures, MBs differentiate into primitive PDGFRβ+ CD271+CD73− mesenchymal progenitors, which give rise to proliferative PCs, SMCs, and mesenchymal stem/stromal cells. MB-derived PCs can be further specified to CD274+ capillary and DLK1+ arteriolar PCs with a proinflammatory and contractile phenotype, respectively. SMC maturation was induced using a MEK inhibitor. Establishing the vasculogenic lineage tree, along with identification of stage- and lineage-specific markers, provides a platform for interrogating the molecular mechanisms that regulate vasculogenic cell specification and diversification and manufacturing well-defined mural cell populations for vascular engineering and cellular therapies from hPSCs. Kumar et al. find that mesodermal pericytes and smooth muscle cells in human pluripotent stem cell cultures originate from a common endothelial and mesenchymal cell precursor, the mesenchymoangioblast. They show how different lineages of mural cells are specified from mesenchymoangioblasts and define stage- and lineage-specific markers for vasculogenic cells.
Collapse
Affiliation(s)
- Akhilesh Kumar
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Saritha Sandra D'Souza
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Oleg V Moskvin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA; Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI 53703, USA
| | - Huishi Toh
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Bowen Wang
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Jue Zhang
- Morgridge Institute for Research, Madison, WI 53707, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53707, USA
| | - Lian-Wang Guo
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI 53707, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53707, USA; Department of Molecular, Cellular & Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53707, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53792, USA.
| |
Collapse
|
14
|
Yokoyama U, Ichikawa Y, Minamisawa S, Ishikawa Y. Pathology and molecular mechanisms of coarctation of the aorta and its association with the ductus arteriosus. J Physiol Sci 2017; 67:259-270. [PMID: 28000176 PMCID: PMC10717425 DOI: 10.1007/s12576-016-0512-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/06/2016] [Indexed: 01/18/2023]
Abstract
Coarctation of the aorta (CoA) is defined as a congenital stenosis of the thoracic aorta and is one of the most common congenital cardiovascular diseases. Despite successful surgical treatment for CoA, arterial abnormalities, including refractory hypertension, aortic aneurysm, and proatherogenic phenotypic changes, frequently affect patients' quality of life. Emerging evidence from morphological and molecular biological investigations suggest that the area of CoA is characterized by phenotypic modulation of smooth muscle cells, intimal thickening, and impaired elastic fiber formation. These changes extend to the pre-and post-stenotic aorta and impair arterial elasticity. The aim of this review is to present current findings on the pathology and molecular mechanisms of vascular remodeling due to CoA. In particular, we will discuss the association between CoA and the ductus arteriosus since the most common site for the stenosis is in the proximity of the ductus arteriosus.
Collapse
Affiliation(s)
- Utako Yokoyama
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Yasuhiro Ichikawa
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Susumu Minamisawa
- The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato, Tokyo, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
15
|
Goyal R, Goyal D, Longo LD, Clyman RI. Microarray gene expression analysis in ovine ductus arteriosus during fetal development and birth transition. Pediatr Res 2016; 80:610-8. [PMID: 27356085 PMCID: PMC5638653 DOI: 10.1038/pr.2016.123] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Patent ductus arteriosus (PDA) in the newborn is the most common congenital heart anomaly and is significantly more common in preterm infants. Contemporary pharmacological treatment is effective in only 70-80% of the cases. Moreover, indomethacin or ibuprofen, which are used to close a PDA may be accompanied by serious side effects in premature infants. To explore the novel molecular pathways, which may be involved in the maturation and closure of the ductus arteriosus (DA), we used fetal and neonatal sheep to test the hypothesis that maturational development of DA is associated with significant alterations in specific mRNA expression. METHODS We conducted oligonucleotide microarray experiments on the isolated mRNA from DA and ascending aorta from three study groups (premature fetus-97 ± 0 d, near-term fetus-136 ± 0.8 d, and newborn lamb-12 ± 0 h). We compared the alterations in mRNA expression in DA and aorta to identify genes specifically involved in DA maturation. RESULTS Results demonstrate significant changes in wingless-integrin1, thrombospondin 1, receptor activator of nuclear factor-kappa B, nitric oxide synthase, and retinoic acid receptor activation signaling pathways. CONCLUSION We conclude that these pathways may play an important role during both development and postnatal DA closure and warrant further investigation.
Collapse
Affiliation(s)
- Ravi Goyal
- Center for Perinatal Biology, Division of Basic Sciences, Loma Linda University, Loma Linda, California
| | - Dipali Goyal
- Center for Perinatal Biology, Division of Basic Sciences, Loma Linda University, Loma Linda, California
| | - Lawrence D. Longo
- Center for Perinatal Biology, Division of Basic Sciences, Loma Linda University, Loma Linda, California
| | - Ronald I. Clyman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
16
|
Parikh P, Bai H, Swartz MF, Alfieris GM, Dean DA. Identification of differentially regulated genes in human patent ductus arteriosus. Exp Biol Med (Maywood) 2016; 241:2112-2118. [PMID: 27465141 DOI: 10.1177/1535370216661778] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In order to identify differentially expressed genes that are specific to the ductus arteriosus, 18 candidate genes were evaluated in matched ductus arteriosus and aortic samples from infants with coarctation of the aorta. The cell specificity of the gene's promoters was assessed by performing transient transfection studies in primary cells derived from several patients. Segments of ductus arteriosus and aorta were isolated from infants requiring repair for coarctation of the aorta and used for mRNA quantitation and culturing of cells. Differences in expression were determined by quantitative PCR using the ΔΔCt method. Promoter regions of six of these genes were cloned into luciferase reporter plasmids for transient transfection studies in matched human ductus arteriosus and aorta cells. Transcription factor AP-2b and phospholipase A2 were significantly up-regulated in ductus arteriosus compared to aorta in whole tissues and cultured cells, respectively. In transient transfection experiments, Angiotensin II type 1 receptor and Prostaglandin E receptor 4 promoters consistently gave higher expression in matched ductus arteriosus versus aorta cells from multiple patients. Taken together, these results demonstrate that several genes are differentially expressed in ductus arteriosus and that their promoters may be used to drive ductus arteriosus-enriched transgene expression.
Collapse
Affiliation(s)
- Pratik Parikh
- 1 Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Haiqing Bai
- 1 Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael F Swartz
- 2 Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - George M Alfieris
- 1 Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.,2 Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David A Dean
- 1 Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.,3 Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
17
|
Effects of Advancing Gestation and Non-Caucasian Race on Ductus Arteriosus Gene Expression. J Pediatr 2015; 167:1033-41.e2. [PMID: 26265282 PMCID: PMC4661123 DOI: 10.1016/j.jpeds.2015.07.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/26/2015] [Accepted: 07/08/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To identify genes affected by advancing gestation and racial/ethnic origin in human ductus arteriosus (DA). STUDY DESIGN We collected 3 sets of DA tissue (n = 93, n = 89, n = 91; total = 273 fetuses) from second trimester pregnancies. We examined four genes, with DNA polymorphisms that distribute along racial lines, to identify "Caucasian" and "non-Caucasian" DA. We used real time polymerase chain reaction to measure RNA expression of 48 candidate genes involved in functional closure of the DA, and used multivariable regression analyses to examine the relationships between advancing gestation, "non-Caucasian" race, and gene expression. RESULTS Mature gestation and non-Caucasian race are significant predictors for identifying infants who will close their patent DA when treated with indomethacin. Advancing gestation consistently altered gene expression in pathways involved with oxygen-induced constriction (eg, calcium-channels, potassium-channels, and endothelin signaling), contractile protein maturation, tissue remodeling, and prostaglandin and nitric oxide signaling in all 3 tissue sets. None of the pathways involved with oxygen-induced constriction appeared to be altered in "non-Caucasian" DA. Two genes, SLCO2A1 and NOS3, (involved with prostaglandin reuptake/metabolism and nitric oxide production, respectively) were consistently decreased in "non-Caucasian" DA. CONCLUSIONS Prostaglandins and nitric oxide are the most important vasodilators opposing DA closure. Indomethacin inhibits prostaglandin production, but not nitric oxide production. Because decreased SLCO2A1 and NOS3 expression can lead to increased prostaglandin and decreased nitric oxide concentrations, we speculate that prostaglandin-mediated vasodilation may play a more dominant role in maintaining the "non-Caucasian" patent DA, making it more likely to close when inhibited by indomethacin.
Collapse
|
18
|
Sehgal PB, Yang YM, Miller EJ. Hypothesis: Neuroendocrine Mechanisms (Hypothalamus-Growth Hormone-STAT5 Axis) Contribute to Sex Bias in Pulmonary Hypertension. Mol Med 2015; 21:688-701. [PMID: 26252185 PMCID: PMC4749490 DOI: 10.2119/molmed.2015.00122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/30/2015] [Indexed: 12/12/2022] Open
Abstract
Pulmonary hypertension (PH) is a disease with high morbidity and mortality. The prevalence of idiopathic pulmonary arterial hypertension (IPAH) and hereditary pulmonary arterial hypertension (HPAH) is approximately two- to four-fold higher in women than in men. Paradoxically, there is an opposite male bias in typical rodent models of PH (chronic hypoxia or monocrotaline); in these models, administration of estrogenic compounds (for example, estradiol-17β [E2]) is protective. Further complexities are observed in humans ingesting anorexigens (female bias) and in rodent models, such as after hypoxia plus SU5416/Sugen (little sex bias) or involving serotonin transporter overexpression or dexfenfluramine administration (female bias). These complexities in sex bias in PH remain incompletely understood. We recently discovered that conditional deletion of signal transducer and activator of transcription 5a/b (STAT5a/b) in vascular smooth muscle cells abrogated the male bias in PH in hypoxic mice and that late-stage obliterative lesions in patients of both sexes with IPAH and HPAH showed reduced STAT5a/b, reduced Tyr-P-STAT5 and reduced B-cell lymphoma 6 protein (BCL6). In trying to understand the significance of these observations, we realized that there existed a well-characterized E2-sensitive central neuroendocrine mechanism of sex bias, studied over the last 40 years, that, at its peripheral end, culminated in species-specific male ("pulsatile") versus female ("more continuous") temporal patterns of circulating growth hormone (GH) levels leading to male versus female patterned activation of STAT5a/b in peripheral tissues and thus sex-biased expression of hundreds of genes. In this report, we consider the contribution of this neuroendocrine mechanism (hypothalamus-GH-STAT5) in the generation of sex bias in different PH situations.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, United States of America
- Department of Medicine, New York Medical College, Valhalla, New York, United States of America
| | - Yang-Ming Yang
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, New York, United States of America
| | - Edmund J Miller
- Center for Heart and Lung Research, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| |
Collapse
|
19
|
Sehgal PB, Yang YM, Yuan H, Miller EJ. STAT5a/b contribute to sex bias in vascular disease: A neuroendocrine perspective. JAKSTAT 2015; 4:1-20. [PMID: 27141328 DOI: 10.1080/21623996.2015.1090658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022] Open
Abstract
Previous studies have elucidated a neuroendocrine mechanism consisting of the hypothalamus (growth hormone releasing hormone, GHRH) - pituitary (growth hormone, GH) - STAT5a/b axis that underlies sex-biased gene expression in the liver. It is now established that male vs female patterned secretion of GHRH, and thus of circulating GH levels ("pulsatile" vs "more continuous" respectively), leading to differently patterned activation of PY-STAT5a/b in hepatocytes results in sex-biased gene expression of cohorts of hundreds of downstream genes. This review outlines new data in support of a STAT5a/b-based mechanism of sex bias in the vascular disease pulmonary hypertension (PH). Puzzling observations in PH include its 2-4-fold higher prevalence in women but a male-dominance in many rodent models, and, paradoxically, inhibition of PH development by estrogens in such models. We observed that conditional deletion of STAT5a/b in vascular smooth muscle cells (SMC) in mice converted the male-dominant model of chronic hypoxia-induced PH into a female-dominant phenotype. In human idiopathic PH, there was reduced STAT5a/b and PY-STAT5 in cells in late-stage obliterative pulmonary arterial lesions in both men and women. A juxtaposition of the prior liver data with the newer PH-related data drew attention to the hypothalamus-GH-STAT5 axis, which is the major target of estrogens at the level of the hypothalamus. This hypothesis explains many of the puzzling aspects of sex bias in PH in humans and rodent models. The extension of STAT5-anchored mechanisms of sex bias to vascular disease emphasizes the contribution of central neuroendocrine processes in generating sexual dimorphism in different tissues and cell types.
Collapse
Affiliation(s)
- Pravin B Sehgal
- Departments of Cell Biology & Anatomy; New York Medical College; Valhalla, NY USA; Department of Medicine; New York Medical College; Valhalla, NY USA
| | - Yang-Ming Yang
- Departments of Cell Biology & Anatomy; New York Medical College ; Valhalla, NY USA
| | - Huijuan Yuan
- Departments of Cell Biology & Anatomy; New York Medical College ; Valhalla, NY USA
| | - Edmund J Miller
- Center for Heart and Lung Research; The Feinstein Institute for Medical Research ; Manhasset, NY USA
| |
Collapse
|
20
|
Kawakami S, Minamisawa S. Oxygenation decreases elastin secretion from rat ductus arteriosus smooth muscle cells. Pediatr Int 2015; 57:541-5. [PMID: 25970707 DOI: 10.1111/ped.12684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 12/09/2014] [Accepted: 01/23/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND The ductus arteriosus (DA), a fetal arterial connection between the main pulmonary artery and the descending aorta, normally closes immediately after birth. The oxygen concentration in the blood rises after birth, and in the DA this increase in oxygen concentration causes functional closure, which is induced by smooth muscle contraction. Previous studies have demonstrated that hypoxia and/or oxygenation affect vascular remodeling of various vessels. Therefore, we hypothesized that the rise in oxygen concentration would affect the vascular structure of the DA due to production of proteins secreted from DA smooth muscle cells (SMC). METHODS AND RESULTS Liquid chromatography-tandem mass spectrometry was used to comprehensively investigate the secreted proteins in the supernatant of rat DA SMC harvested under hypoxic conditions (1% oxygen) or under normoxic conditions (21% oxygen). We found that the rise in oxygen concentration reduced the secretion of elastin from DA SMC. On reverse transcription-polymerase chain reaction, the expression of elastin mRNA was not significantly changed in DA SMC from hypoxic to normoxic conditions. CONCLUSIONS Given that elastin forms internal elastic lamina and elastic fibers in the vascular muscle layers, and that a rise in oxygen concentration reduced the secretion of elastin, this suggests that the rise in blood oxygen concentration after birth reduces the secretion of elastin, and therefore may play a role in DA structural remodeling after birth.
Collapse
Affiliation(s)
- Shoji Kawakami
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Susumu Minamisawa
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Department of Cell Physiology, Jikei University, Tokyo, Japan
| |
Collapse
|
21
|
Shelton EL, Ector G, Galindo CL, Hooper CW, Brown N, Wilkerson I, Pfaltzgraff ER, Paria BC, Cotton RB, Stoller JZ, Reese J. Transcriptional profiling reveals ductus arteriosus-specific genes that regulate vascular tone. Physiol Genomics 2014; 46:457-66. [PMID: 24790087 DOI: 10.1152/physiolgenomics.00171.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Failure of the ductus arteriosus (DA) to close at birth can lead to serious complications. Conversely, certain profound congenital cardiac malformations require the DA to be patent until corrective surgery can be performed. In each instance, clinicians have a very limited repertoire of therapeutic options at their disposal - indomethacin or ibuprofen to close a patent DA (PDA) and prostaglandin E1 to maintain patency of the DA. Neither treatment is specific to the DA and both may have deleterious off-target effects. Therefore, more therapeutic options specifically targeted to the DA should be considered. We hypothesized the DA possesses a unique genetic signature that would set it apart from other vessels. A microarray was used to compare the genetic profiles of the murine DA and ascending aorta (AO). Over 4,000 genes were differentially expressed between these vessels including a subset of ion channel-related genes. Specifically, the alpha and beta subunits of large-conductance calcium-activated potassium (BKCa) channels are enriched in the DA. Gain- and loss-of-function studies showed inhibition of BKCa channels caused the DA to constrict, while activation caused DA relaxation even in the presence of O2. This study identifies subsets of genes that are enriched in the DA that may be used to develop DA-specific drugs. Ion channels that regulate DA tone, including BKCa channels, are promising targets. Specifically, BKCa channel agonists like NS1619 maintain DA patency even in the presence of O2 and may be clinically useful.
Collapse
Affiliation(s)
- Elaine L Shelton
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee;
| | - Gerren Ector
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Cristi L Galindo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; and
| | - Christopher W Hooper
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Naoko Brown
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Irene Wilkerson
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elise R Pfaltzgraff
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Bibhash C Paria
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Robert B Cotton
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jason Z Stoller
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
22
|
Three-dimensional multilayers of smooth muscle cells as a new experimental model for vascular elastic fiber formation studies. Atherosclerosis 2014; 233:590-600. [DOI: 10.1016/j.atherosclerosis.2014.01.045] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 02/01/2023]
|
23
|
Hsieh YT, Liu NM, Ohmori E, Yokota T, Kajimura I, Akaike T, Ohshima T, Goda N, Minamisawa S. Transcription profiles of the ductus arteriosus in Brown-Norway rats with irregular elastic fiber formation. Circ J 2014; 78:1224-33. [PMID: 24647370 DOI: 10.1253/circj.cj-13-1029] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Patent ductus arteriosus (PDA) is one of the most common congenital cardiovascular defects in children. The Brown-Norway (BN) inbred rat presents a higher frequency of PDA. A previous study reported that 2 different quantitative trait loci on chromosomes 8 and 9 were significantly linked to PDA in this strain. Nevertheless, the genetic or molecular mechanisms underlying PDA phenotypes in BN rats have not been fully investigated yet. METHODS AND RESULTS It was found that the elastic fibers were abundant in the subendothelial area but scarce in the media even in the closed ductus arteriosus (DA) of full-term BN neonates. DNA microarray analysis identified 52 upregulated genes (fold difference >2.5) and 23 downregulated genes (fold difference <0.4) when compared with those of F344 control neonates. Among these genes, 8 (Tbx20, Scn3b, Stac, Sphkap, Trpm8, Rup2, Slc37a2, and RGD1561216) are located in chromosomes 8 and 9. Interestingly, it was also suggested that the significant decrease in the expression levels of the PGE2-specfic receptor, EP4, plays a critical role in elastogenesis in the DA. CONCLUSIONS BN rats exhibited dysregulation of elastogenesis in the DA. DNA microarray analysis identified the candidate genes including EP4 involved in the DNA phenotype. Further investigation of these newly identified genes will hopefully clarify the molecular mechanisms underlying the irregular formation of elastic fibers in PDA.
Collapse
Affiliation(s)
- Yi-Ting Hsieh
- Department of Life Science and Medical Bioscience, Waseda University
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Bökenkamp R, van Brempt R, van Munsteren JC, van den Wijngaert I, de Hoogt R, Finos L, Goeman J, Groot ACGD, Poelmann RE, Blom NA, DeRuiter MC. Dlx1 and Rgs5 in the ductus arteriosus: vessel-specific genes identified by transcriptional profiling of laser-capture microdissected endothelial and smooth muscle cells. PLoS One 2014; 9:e86892. [PMID: 24489801 PMCID: PMC3904938 DOI: 10.1371/journal.pone.0086892] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022] Open
Abstract
Closure of the ductus arteriosus (DA) is a crucial step in the transition from fetal to postnatal life. Patent DA is one of the most common cardiovascular anomalies in children with significant clinical consequences especially in premature infants. We aimed to identify genes that specify the DA in the fetus and differentiate it from the aorta. Comparative microarray analysis of laser-captured microdissected endothelial (ECs) and vascular smooth muscle cells (SMCs) from the DA and aorta of fetal rats (embryonic day 18 and 21) identified vessel-specific transcriptional profiles. We found a strong age-dependency of gene expression. Among the genes that were upregulated in the DA the regulator of the G-protein coupled receptor 5 (Rgs5) and the transcription factor distal-less homeobox 1 (Dlx1) exhibited the highest and most significant level of differential expression. The aorta showed a significant preferential expression of the Purkinje cell protein 4 (Pcp4) gene. The results of the microarray analysis were validated by real-time quantitative PCR and immunohistochemistry. Our study confirms vessel-specific transcriptional profiles in ECs and SMCs of rat DA and aorta. Rgs5 and Dlx1 represent novel molecular targets for the regulation of DA maturation and closure.
Collapse
Affiliation(s)
- Regina Bökenkamp
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | - Ronald van Brempt
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
- Johnson and Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | | | | | - Ronald de Hoogt
- Johnson and Johnson Pharmaceutical Research and Development, Beerse, Belgium
| | - Livio Finos
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jelle Goeman
- Biostatistics, Department for Health Evidence, Radboud University Medical Center, Nimegen, The Netherlands
| | - Adriana Cornelia Gittenberger-de Groot
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Eugen Poelmann
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nicolaas Andreas Blom
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
25
|
Transcription profiles of endothelial cells in the rat ductus arteriosus during a perinatal period. PLoS One 2013; 8:e73685. [PMID: 24086288 PMCID: PMC3785468 DOI: 10.1371/journal.pone.0073685] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/19/2013] [Indexed: 11/19/2022] Open
Abstract
Endothelial cells (ECs) lining the blood vessels serve a variety of functions and play a central role in the homeostasis of the circulatory system. Since the ductus arteriosus (DA) has different arterial characteristics from its connecting vessels, we hypothesized that ECs of the DA exhibited a unique gene profile involved in the regulation of DA-specific morphology and function. Using a fluorescence-activated cell sorter, we isolated ECs from pooled tissues from the DA or the descending aorta of Wistar rat fetuses at full-term of gestation (F group) or neonates 30 minutes after birth (N group). Using anti-CD31 and anti-CD45 antibodies as cell surface markers for ECs and hematopoietic derived cells, respectively, cDNAs from the CD31-positive and CD45-negative cells were hybridized to the Affymetrix GeneChip® Rat Gene 1.0 ST Array. Among 26,469 gene-level probe sets, 82 genes in the F group and 81 genes in the N group were expressed at higher levels in DA ECs than in aortic ECs (p<0.05, fold change>2.0). In addition to well-known endothelium-enriched genes such as Tgfb2 and Vegfa, novel DA endothelium-dominant genes including Slc38a1, Capn6, and Lrat were discovered. Enrichment analysis using GeneGo MetaCore software showed that DA endothelium-related biological processes were involved in morphogenesis and development. We identified many overlapping genes in each process including neural crest-related genes (Hoxa1, Hoxa4, and Hand2, etc) and the second heart field-related genes (Tbx1, Isl1, and Fgf10, etc). Moreover, we found that regulation of epithelial-to-mesenchymal transition, cell adhesion, and retinol metabolism are the active pathways involved in the network via potential interactions with many of the identified genes to form DA-specific endothelia. In conclusion, the present study uncovered several significant differences of the transcriptional profile between the DA and aortic ECs. Newly identified DA endothelium-dominant genes may play an important role in DA-specific functional and morphologic characteristics.
Collapse
|
26
|
Abstract
Closure of the ductus arteriosus at birth is a complex phenomenon being conditioned by antenatal events and progressing in preprogrammed steps. Functional at first, narrowing of the vessel is determined by 2 overlapping processes--removal of the prostaglandin E(2)-based relaxation sustaining prenatal patency and activation of a constrictor mechanism by the natural rise in blood oxygen tension. Two schemes have been proposed for oxygen action--one involving a cytochrome P450 hemoprotein (sensor)/endothelin-1 (effector) complex and the other a set of voltage-gated K(+) channels. These proposals, however, are not mutually exclusive. Structural closure follows the constriction through a remodeling process initiated antenatally with the development of intimal cushions and completed postnatally by a host of humoral and mechanical stimuli. Research in this area has already provided clinical applications. Nevertheless, management of premature infants with persistent ductus remains troublesome and calls for an alternative approach to the prostaglandin E(2) inhibitors now in use. Studies in progress on the oxygen-sensing system may lead to a definitive solution for this problem.
Collapse
Affiliation(s)
- Flavio Coceani
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| | | |
Collapse
|
27
|
Stoller JZ, Demauro SB, Dagle JM, Reese J. Current Perspectives on Pathobiology of the Ductus Arteriosus. ACTA ACUST UNITED AC 2012; 8. [PMID: 23519783 DOI: 10.4172/2155-9880.s8-001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The ductus arteriosus (DA) shunts blood away from the lungs during fetal life, but at birth this shunt is no longer needed and the vessel rapidly constricts. Postnatal persistence of the DA, patent ductus arteriosus (PDA), is predominantly a detrimental condition for preterm infants but is simultaneously a condition required to maintain systemic blood flow for infants born with certain severe congenital heart defects. Although PDA in preterm infants is associated with significant morbidities, there is controversy regarding whether PDA is truly causative. Despite advances in our understanding of the pathobiology of PDA, the optimal treatment strategy for PDA in preterm infants is unclear. Here we review recent studies that have continued to elucidate the fundamental mechanisms of DA development and pathogenesis.
Collapse
Affiliation(s)
- Jason Z Stoller
- Department of Pediatrics, University of Pennsylvania School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
28
|
Coceani F, Scebba F, Angeloni D. Gene profiling in ductus arteriosus and aorta: a question of consistency. J Physiol Sci 2011; 61:443-4. [PMID: 21744167 PMCID: PMC10717251 DOI: 10.1007/s12576-011-0161-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 06/18/2011] [Indexed: 10/24/2022]
|