1
|
Lahmouch N, Bouzerda A, Asfalou I, Lakhal Z, Benyass A. Proximal connection anomalies of the coronary arteries to the aorta: retrospective experience in a Moroccan university hospital and literature review. Pan Afr Med J 2024; 48:139. [PMID: 39554262 PMCID: PMC11567904 DOI: 10.11604/pamj.2024.48.139.42943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/24/2024] [Indexed: 11/19/2024] Open
Abstract
Proximal connection anomalies of the coronary arteries to the aorta represent a rare pathology, with an angiographic prevalence close to 1%. The prognosis of this condition is contingent upon its anatomical form. Some instances are linked to sudden deaths, while others may be associated with myocardial ischemia. Utilizing computed tomography (CT) coronary angiography as the optimal imaging tool, one can identify the origin and course of the ectopic artery. Current guidelines suggest surgical correction as the primary intervention for symptomatic abnormalities when a risky form is identified. To enhance the management of these anomalies, the establishment of comprehensive multicenter observational registers is imperative. Our study, a retrospective analysis spanning two years, focuses on 10 cases of proximal connection anomalies of coronary arteries to the aorta diagnosed in the Cardiology department of the Mohammed V Military Hospital of Rabat. By presenting this series and conducting a literature review, we elucidate the anatomical, epidemiological, physiopathological, clinical, angiographic, and CT angiography features of these anomalies, along with insights into therapeutic management.
Collapse
Affiliation(s)
- Nouhaila Lahmouch
- Department of Cardiology, Mohammed V Military Hospital, Faculty of Medicine and Pharmacy of Rabat, Mohammed V University, Rabat, Morocco
| | - Abdelmajid Bouzerda
- Department of Cardiology, Avicenne Military Hospital, Faculty of Medicine and Pharmacy, Cadi Ayad University, Marrakech, Morroco
| | - Ilyasse Asfalou
- Department of Cardiology, Mohammed V Military Hospital, Faculty of Medicine and Pharmacy of Rabat, Mohammed V University, Rabat, Morocco
| | - Zouhair Lakhal
- Department of Cardiology, Mohammed V Military Hospital, Faculty of Medicine and Pharmacy of Rabat, Mohammed V University, Rabat, Morocco
| | - Aatif Benyass
- Department of Cardiology, Mohammed V Military Hospital, Faculty of Medicine and Pharmacy of Rabat, Mohammed V University, Rabat, Morocco
| |
Collapse
|
2
|
Pineda-Castillo SA, Acar H, Detamore MS, Holzapfel GA, Lee CH. Modulation of Smooth Muscle Cell Phenotype for Translation of Tissue-Engineered Vascular Grafts. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:574-588. [PMID: 37166394 PMCID: PMC10618830 DOI: 10.1089/ten.teb.2023.0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Translation of small-diameter tissue-engineered vascular grafts (TEVGs) for the treatment of coronary artery disease (CAD) remains an unfulfilled promise. This is largely due to the limited integration of TEVGs into the native vascular wall-a process hampered by the insufficient smooth muscle cell (SMC) infiltration and extracellular matrix deposition, and low vasoactivity. These processes can be promoted through the judicious modulation of the SMC toward a synthetic phenotype to promote remodeling and vascular integration; however, the expression of synthetic markers is often accompanied by a decrease in the expression of contractile proteins. Therefore, techniques that can precisely modulate the SMC phenotypical behavior could have the potential to advance the translation of TEVGs. In this review, we describe the phenotypic diversity of SMCs and the different environmental cues that allow the modulation of SMC gene expression. Furthermore, we describe the emerging biomaterial approaches to modulate the SMC phenotype in TEVG design and discuss the limitations of current techniques. In addition, we found that current studies in tissue engineering limit the analysis of the SMC phenotype to a few markers, which are often the characteristic of early differentiation only. This limited scope has reduced the potential of tissue engineering to modulate the SMC toward specific behaviors and applications. Therefore, we recommend using the techniques presented in this review, in addition to modern single-cell proteomics analysis techniques to comprehensively characterize the phenotypic modulation of SMCs. Expanding the holistic potential of SMC modulation presents a great opportunity to advance the translation of living conduits for CAD therapeutics.
Collapse
Affiliation(s)
- Sergio A. Pineda-Castillo
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Chung-Hao Lee
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
3
|
Narasimhan B, Narasimhan H, Lorente-Ros M, Romeo FJ, Bhatia K, Aronow WS. Therapeutic angiogenesis in coronary artery disease: a review of mechanisms and current approaches. Expert Opin Investig Drugs 2021; 30:947-963. [PMID: 34346802 DOI: 10.1080/13543784.2021.1964471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Despite tremendous advances, the shortcomings of current therapies for coronary disease are evidenced by the fact that it remains the leading cause of death in many parts of the world. There is hence a drive to develop novel therapies to tackle this disease. Therapeutic approaches to coronary angiogenesis have long been an area of interest in lieu of its incredible, albeit unrealized potential. AREAS COVERED This paper offers an overview of mechanisms of native angiogenesis and a description of angiogenic growth factors. It progresses to outline the advances in gene and stem cell therapy and provides a brief description of other investigational approaches to promote angiogenesis. Finally, the hurdles and limitations unique to this particular area of study are discussed. EXPERT OPINION An effective, sustained, and safe therapeutic option for angiogenesis truly could be the paradigm shift for cardiovascular medicine. Unfortunately, clinically meaningful therapeutic options remain elusive because promising animal studies have not been replicated in human trials. The sheer complexity of this process means that numerous major hurdles remain before therapeutic angiogenesis truly makes its way from the bench to the bedside.
Collapse
Affiliation(s)
- Bharat Narasimhan
- Department Of Medicine, Mount Sinai St.Lukes-Roosevelt, Icahn School Of Medicine At Mount Sinai, New York, NY, USA
| | | | - Marta Lorente-Ros
- Department Of Medicine, Mount Sinai St.Lukes-Roosevelt, Icahn School Of Medicine At Mount Sinai, New York, NY, USA
| | - Francisco Jose Romeo
- Department Of Medicine, Mount Sinai St.Lukes-Roosevelt, Icahn School Of Medicine At Mount Sinai, New York, NY, USA
| | - Kirtipal Bhatia
- Department Of Medicine, Mount Sinai St.Lukes-Roosevelt, Icahn School Of Medicine At Mount Sinai, New York, NY, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center/New York Medical College, Valhalla, NY, USA
| |
Collapse
|
4
|
Stone OA, Zhou B, Red-Horse K, Stainier DYR. Endothelial ontogeny and the establishment of vascular heterogeneity. Bioessays 2021; 43:e2100036. [PMID: 34145927 DOI: 10.1002/bies.202100036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
The establishment of distinct cellular identities was pivotal during the evolution of Metazoa, enabling the emergence of an array of specialized tissues with different functions. In most animals including vertebrates, cell specialization occurs in response to a combination of intrinsic (e.g., cellular ontogeny) and extrinsic (e.g., local environment) factors that drive the acquisition of unique characteristics at the single-cell level. The first functional organ system to form in vertebrates is the cardiovascular system, which is lined by a network of endothelial cells whose organ-specific characteristics have long been recognized. Recent genetic analyses at the single-cell level have revealed that heterogeneity exists not only at the organ level but also between neighboring endothelial cells. Thus, how endothelial heterogeneity is established has become a key question in vascular biology. Drawing upon evidence from multiple organ systems, here we will discuss the role that lineage history may play in establishing endothelial heterogeneity.
Collapse
Affiliation(s)
- Oliver A Stone
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kristy Red-Horse
- Department of Biology, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
5
|
Das RN, Yaniv K. Discovering New Progenitor Cell Populations through Lineage Tracing and In Vivo Imaging. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035618. [PMID: 32041709 DOI: 10.1101/cshperspect.a035618] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Identification of progenitor cells that generate differentiated cell types during development, regeneration, and disease states is central to understanding the mechanisms governing such transitions. For more than a century, different lineage-tracing strategies have been developed, which helped disentangle the complex relationship between progenitor cells and their progenies. In this review, we discuss how lineage-tracing analyses have evolved alongside technological advances, and how this approach has contributed to the identification of progenitor cells in different contexts of cell differentiation. We also highlight a few examples in which lineage-tracing experiments have been instrumental for resolving long-standing debates and for identifying unexpected cellular origins. This discussion emphasizes how this century-old quest to delineate cellular lineage relationships is still active, and new discoveries are being made with the development of newer methodologies.
Collapse
Affiliation(s)
- Rudra Nayan Das
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Karina Yaniv
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
6
|
Jallerat Q, Feinberg AW. Extracellular Matrix Structure and Composition in the Early Four-Chambered Embryonic Heart. Cells 2020; 9:cells9020285. [PMID: 31991580 PMCID: PMC7072588 DOI: 10.3390/cells9020285] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 01/30/2023] Open
Abstract
During embryonic development, the heart undergoes complex morphogenesis from a liner tube into the four chambers consisting of ventricles, atria and valves. At the same time, the cardiomyocytes compact into a dense, aligned, and highly vascularized myocardium. The extracellular matrix (ECM) is known to play an important role in this process but understanding of the expression and organization remains incomplete. Here, we performed 3D confocal imaging of ECM in the left ventricle and whole heart of embryonic chick from stages Hamburger-Hamilton 28-35 (days 5-9) as an accessible model of heart formation. First, we observed the formation of a fibronectin-rich, capillary-like networks in the myocardium between day 5 and day 9 of development. Then, we focused on day 5 prior to vascularization to determine the relative expression of fibronectin, laminin, and collagen type IV. Cardiomyocytes were found to uniaxially align prior to vascularization and, while the epicardium contained all ECM components, laminin was reduced, and collagen type IV was largely absent. Quantification of fibronectin revealed highly aligned fibers with a mean diameter of ~500 nm and interfiber spacing of ~3 µm. These structural parameters (volume, spacing, fiber diameter, length, and orientation) provide a quantitative framework to describe the organization of the embryonic ECM.
Collapse
Affiliation(s)
- Quentin Jallerat
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Adam W. Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
- Department of Materials Science & Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Correspondence: ; Tel.: +1-412-268-4897
| |
Collapse
|
7
|
Schwartz SM, Virmani R, Majesky MW. An update on clonality: what smooth muscle cell type makes up the atherosclerotic plaque? F1000Res 2018; 7:F1000 Faculty Rev-1969. [PMID: 30613386 PMCID: PMC6305222 DOI: 10.12688/f1000research.15994.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
Almost 50 years ago, Earl Benditt and his son John described the clonality of the atherosclerotic plaque. This led Benditt to propose that the atherosclerotic lesion was a smooth muscle neoplasm, similar to the leiomyomata seen in the uterus of most women. Although the observation of clonality has been confirmed many times, interest in the idea that atherosclerosis might be a form of neoplasia waned because of the clinical success of treatments for hyperlipemia and because animal models have made great progress in understanding how lipid accumulates in the plaque and may lead to plaque rupture. Four advances have made it important to reconsider Benditt's observations. First, we now know that clonality is a property of normal tissue development. Second, this is even true in the vessel wall, where we now know that formation of clonal patches in that wall is part of the development of smooth muscle cells that make up the tunica media of arteries. Third, we know that the intima, the "soil" for development of the human atherosclerotic lesion, develops before the fatty lesions appear. Fourth, while the cells comprising this intima have been called "smooth muscle cells", we do not have a clear definition of cell type nor do we know if the initial accumulation is clonal. As a result, Benditt's hypothesis needs to be revisited in terms of changes in how we define smooth muscle cells and the quite distinct developmental origins of the cells that comprise the muscular coats of all arterial walls. Finally, since clonality of the lesions is real, the obvious questions are do these human tumors precede the development of atherosclerosis, how do the clones develop, what cell type gives rise to the clones, and in what ways do the clones provide the soil for development and natural history of atherosclerosis?
Collapse
Affiliation(s)
| | - Renu Virmani
- CV Path Institute, Gaithersberg, Maryland, 20878, USA
| | - Mark W. Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital Research Institute, Seattle, WA, 98112, USA
| |
Collapse
|
8
|
Ikeda N, Nakazawa N, Kurata Y, Yaura H, Taufiq F, Minato H, Yoshida A, Ninomiya H, Nakayama Y, Kuwabara M, Shirayoshi Y, Hisatome I. Tbx18-positive cells differentiated from murine ES cells serve as proepicardial progenitors to give rise to vascular smooth muscle cells and fibroblasts. Biomed Res 2018; 38:229-238. [PMID: 28794400 DOI: 10.2220/biomedres.38.229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Proepicardium (PE) cells generate cardiac fibroblasts, smooth muscle cells (SMCs) and endothelial cells that form coronary arteries. T-box18 (Tbx18) is a well-known marker of PE cells and epicardium. We examined whether Tbx18-positive cells differentiated from murine embryonic stem (ES) cells serve as PE progenitors to give rise to vascular SMCs and fibroblasts. To collect Tbx18-positive cells, we established Tbx18-EGFP knock-in mouse ES cells using the CRISPR/Cas9 system. We harvested the Tbx18-EGFP-positive cells on day 8, 10 and 14 after the initiation of differentiation; Tbx18 mRNA was enriched on day 8 to 14 and Snai2 mRNA was enriched on day 8 and 10, indicating successful collection of Tbx18-positive cells. Tbx18-EGFP-positive cells expressed the PE marker WT1 on day 8 and 10. They also expressed the SMC marker Acta2 and fibroblast markers Thy1 and Fsp1 on day 8 to 14, but did not express the endothelial cell marker PECAM or the cardiac cell marker CD166 or Myh7. In conclusion, Tbx18-positive cells represent a part of PE cells in the initial phase of differentiation and subsequently include SMCs as well as fibroblasts. These results indicate that Tbx18-positive cells serve as a PE progenitor to supply a variety of cells that contribute to the formation of coronary arteries.
Collapse
Affiliation(s)
- Nobuhito Ikeda
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Natsumi Nakazawa
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | | | - Hisako Yaura
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Fikri Taufiq
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Hiroyuki Minato
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Akio Yoshida
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Haruaki Ninomiya
- Department of Biological Regulation, Tottori University Faculty of Medicine
| | - Yuji Nakayama
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University
| | - Masanari Kuwabara
- University of Colorado, Denver, School of Medicine, Division of Renal Diseases and Hypertension
| | - Yasuaki Shirayoshi
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| | - Ichiro Hisatome
- Division of Regenerative Medicine and Therapeutics, Department of Genetic Medicine and Regenerative Therapeutics, Institute of Regenerative Medicine and Biofunction, Tottori University Graduate School of Medical Science
| |
Collapse
|
9
|
Ramai D, Lai J, Monzidelis C, Reddy S. Coronary Artery Development: Origin, Malformations, and Translational Vascular Reparative Therapy. J Cardiovasc Pharmacol Ther 2018; 23:292-300. [PMID: 29642708 DOI: 10.1177/1074248418769633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After thickening of the cardiac chamber walls during embryogenesis, oxygen and nutrients can no longer be adequately supplied to cardiac cells via passive diffusion; therefore, a primitive vascular network develops to supply these vital structures. This plexus further matures into coronary arteries and veins, which ensures continued development of the heart. Various models have been proposed to account for the growth of the coronary arteries. However, lineage-tracing studies in the last decade have identified 3 major sources, namely, the proepicardium, the sinus venosus, and endocardium. Although the exact contribution of each source remains unknown, the emerging model depicts alternative pathways and progenitor cells, which ensure successful coronary angiogenesis. We aim to explore the current trends in coronary artery development, the cellular and molecular signals regulating heart vascularization, and its implications for heart disease and vascular regeneration.
Collapse
Affiliation(s)
- Daryl Ramai
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Jonathan Lai
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Constantine Monzidelis
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| | - Sarath Reddy
- Division of Cardiology, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| |
Collapse
|
10
|
Halper J. Basic Components of Vascular Connective Tissue and Extracellular Matrix. ADVANCES IN PHARMACOLOGY 2017; 81:95-127. [PMID: 29310805 DOI: 10.1016/bs.apha.2017.08.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Though the composition of the three layers constituting the blood vessel wall varies among the different types of blood vessels, and some layers may even be missing in capillaries, certain basic components, and properties are shared by all blood vessels, though each histologically distinct layer contains a unique complement of extracellular components, growth factors and cytokines, and cell types as well. The structure and composition of vessel layers informs and is informed by the function of the particular blood vessel. The adaptation of the composition and the resulting function of the extracellular matrix (ECM) to changes in circulation/blood flow and a variety of other extravascular stimuli can be characterized as remodeling spearheaded by vascular cells. There is a surprising amount of cell traffic among the three layers. It starts with endothelial cell mediated transmigration of inflammatory cells from the bloodstream into the subendothelium, and then into tissue adjoining the blood vessel. Smooth muscle cells and a variety of adventitial cells reside in tunica media and tunica externa, respectively. The latter cells are a mixture of progenitor/stem cells, fibroblasts, myofibroblasts, pericytes, macrophages, and dendritic cells and respond to endothelial injury by transdifferentiation as they travel into the two inner layers, intima and media for corrective mission in the ECM composition. This chapter addresses the role of various vascular cell types and ECM components synthesized by them in maintenance of normal structure and in their contribution to major pathological processes, such as atherosclerosis, organ fibrosis, and diabetic retinopathy.
Collapse
Affiliation(s)
- Jaroslava Halper
- College of Veterinary Medicine and AU/UGA Medical Partnership, The University of Georgia, Athens, GA, United States.
| |
Collapse
|
11
|
Mazurek R, Dave JM, Chandran RR, Misra A, Sheikh AQ, Greif DM. Vascular Cells in Blood Vessel Wall Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:323-350. [PMID: 28212800 DOI: 10.1016/bs.apha.2016.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vessel wall is composed of distinct cellular layers, yet communication among individual cells within and between layers results in a dynamic and versatile structure. The morphogenesis of the normal vascular wall involves a highly regulated process of cell proliferation, migration, and differentiation. The use of modern developmental biological and genetic approaches has markedly enriched our understanding of the molecular and cellular mechanisms underlying these developmental events. Additionally, the application of similar approaches to study diverse vascular diseases has resulted in paradigm-shifting insights into pathogenesis. Further investigations into the biology of vascular cells in development and disease promise to have major ramifications on therapeutic strategies to combat pathologies of the vasculature.
Collapse
Affiliation(s)
- R Mazurek
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - J M Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - R R Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Misra
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - D M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
12
|
Raimondi F, Bonnet D. Imaging of congenital anomalies of the coronary arteries. Diagn Interv Imaging 2016; 97:561-9. [PMID: 27132711 DOI: 10.1016/j.diii.2016.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 03/23/2016] [Indexed: 11/28/2022]
Abstract
Congenital abnormalities of the coronary arteries are extremely variable and include anomalies of their origin (atresia, anomalies of origin from the aorta or from the pulmonary artery), the course of the epicardiac coronary branches (intramural, myocardial bridge) and distal connections (coronary-cardiac chamber fistulae). In pediatric practice, the diagnosis relies on ultrasound which should be supplemented by additional cardiac imaging in most cases. Multidetector CT is the most widely used imaging technique to identify abnormal courses and relationships with the greater vessels. In this paper, the important diagnostic and prognostic features in the interpretation of coronary imaging in pediatric practice is discussed.
Collapse
Affiliation(s)
- F Raimondi
- Pediatric cardiology-M3C, hôpital Necker-Enfants-Malades, AP-HP, université Paris-Descartes, Sorbonne Paris Cité, 149, rue de Sèvres, 75743 Paris cedex 15, France.
| | - D Bonnet
- Pediatric cardiology-M3C, hôpital Necker-Enfants-Malades, AP-HP, université Paris-Descartes, Sorbonne Paris Cité, 149, rue de Sèvres, 75743 Paris cedex 15, France
| |
Collapse
|
13
|
Lluri G, Huang V, Touma M, Liu X, Harmon AW, Nakano A. Hematopoietic progenitors are required for proper development of coronary vasculature. J Mol Cell Cardiol 2015; 86:199-207. [PMID: 26241844 DOI: 10.1016/j.yjmcc.2015.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/29/2015] [Accepted: 07/24/2015] [Indexed: 10/23/2022]
Abstract
RATIONALE During embryogenesis, hematopoietic cells appear in the myocardium prior to the initiation of coronary formation. However, their role is unknown. OBJECTIVE Here we investigate whether pre-existing hematopoietic cells are required for the formation of coronary vasculature. METHODS AND RESULTS As a model of for hematopoietic cell deficient animals, we used Runx1 knockout embryos and Vav1-cre; R26-DTA embryos, latter of which genetically ablates 2/3 of CD45(+) hematopoietic cells. Both Runx1 knockout embryos and Vav1-cre; R26-DTA embryos revealed disorganized, hypoplastic microvasculature of coronary vessels on section and whole-mount stainings. Furthermore, coronary explant experiments showed that the mouse heart explants from Runx1 and Vav1-cre; R26-DTA embryos exhibited impaired coronary formation ex vivo. Interestingly, in both models it appears that epicardial to mesenchymal transition is adversely affected in the absence of hematopoietic progenitors. CONCLUSION Hematopoietic cells are not merely passively transported via coronary vessel, but substantially involved in the induction of the coronary growth. Our findings suggest a novel mechanism of coronary growth.
Collapse
Affiliation(s)
- Gentian Lluri
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, Section of Cardiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vincent Huang
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marlin Touma
- Children's Discovery and Innovation Institute Department of Pediatrics, Department of Molecular Cell and Integrative Physiology, David Geffen School of Medicine, USA
| | - Xiaoqian Liu
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andrew W Harmon
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Nakano
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
14
|
Abstract
Coronary artery disease causes acute myocardial infarction and heart failure. Identifying coronary vascular progenitors and their developmental program could inspire novel regenerative treatments for cardiac diseases. The developmental origins of the coronary vessels have been shrouded in mystery and debated for several decades. Recent identification of progenitors for coronary vessels within the endocardium, epicardium, and sinus venosus provides new insights into this question. In addition, significant progress has been achieved in elucidating the cellular and molecular programs that orchestrate coronary artery development. Establishing adequate vascular supply will be an essential component of cardiac regenerative strategies, and these findings raise exciting new strategies for therapeutic cardiac revascularization.
Collapse
Affiliation(s)
- Xueying Tian
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - William T Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (X.T., B.Z.) and CAS Center for Excellence in Brain Science (B.Z.), Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China; Department of Cardiology, Boston Children's Hospital, MA (W.T.P.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
15
|
Seidelmann SB, Lighthouse JK, Greif DM. Development and pathologies of the arterial wall. Cell Mol Life Sci 2014; 71:1977-99. [PMID: 24071897 PMCID: PMC11113178 DOI: 10.1007/s00018-013-1478-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/13/2023]
Abstract
Arteries consist of an inner single layer of endothelial cells surrounded by layers of smooth muscle and an outer adventitia. The majority of vascular developmental studies focus on the construction of endothelial networks through the process of angiogenesis. Although many devastating vascular diseases involve abnormalities in components of the smooth muscle and adventitia (i.e., the vascular wall), the morphogenesis of these layers has received relatively less attention. Here, we briefly review key elements underlying endothelial layer formation and then focus on vascular wall development, specifically on smooth muscle cell origins and differentiation, patterning of the vascular wall, and the role of extracellular matrix and adventitial progenitor cells. Finally, we discuss select human diseases characterized by marked vascular wall abnormalities. We propose that continuing to apply approaches from developmental biology to the study of vascular disease will stimulate important advancements in elucidating disease mechanism and devising novel therapeutic strategies.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Animals
- Arteries/growth & development
- Arteries/metabolism
- Arteries/pathology
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cell Differentiation
- Cell Lineage/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Developmental
- Humans
- Morphogenesis/genetics
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
Collapse
Affiliation(s)
- Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Janet K. Lighthouse
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Daniel M. Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| |
Collapse
|
16
|
Wu SP, Dong XR, Regan JN, Su C, Majesky MW. Tbx18 regulates development of the epicardium and coronary vessels. Dev Biol 2013; 383:307-20. [PMID: 24016759 DOI: 10.1016/j.ydbio.2013.08.019] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/21/2013] [Accepted: 08/21/2013] [Indexed: 11/16/2022]
Abstract
The epicardium and coronary vessels originate from progenitor cells in the proepicardium. Here we show that Tbx18, a T-box family member highly expressed in the proepicardium, controls critical early steps in coronary development. In Tbx18(-/-) mouse embryos, both the epicardium and coronary vessels exhibit structural and functional defects. At E12.5, the Tbx18-deficient epicardium contains protrusions and cyst-like structures overlying a disorganized coronary vascular plexus that contains ectopic structures resembling blood islands. At E13.5, the left and right coronary stems form correctly in mutant hearts. However, analysis of PECAM-1 whole mount immunostaining, distribution of SM22α(lacZ/+) activity, and analysis of coronary vascular casts suggest that defective vascular plexus remodeling produces a compromised arterial network at birth consisting of fewer distributing conduit arteries with smaller lumens and a reduced capacity to conduct blood flow. Gene expression profiles of Tbx18(-/-) hearts at E12.5 reveal altered expression of 79 genes that are associated with development of the vascular system including sonic hedgehog signaling components patched and smoothened, VEGF-A, angiopoietin-1, endoglin, and Wnt factors compared to wild type hearts. Thus, formation of coronary vasculature is responsive to Tbx18-dependent gene targets in the epicardium, and a poorly structured network of coronary conduit vessels is formed in Tbx18 null hearts due to defects in epicardial cell signaling and fate during heart development. Lastly, we demonstrate that Tbx18 possesses a SRF/CArG box dependent repressor activity capable of inhibiting progenitor cell differentiation into smooth muscle cells, suggesting a potential function of Tbx18 in maintaining the progenitor status of epicardial-derived cells.
Collapse
Affiliation(s)
- San-Pin Wu
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States.
| | | | | | | | | |
Collapse
|
17
|
Zhang Z, Zhou B. Accelerated coronary angiogenesis by vegfr1-knockout endocardial cells. PLoS One 2013; 8:e70570. [PMID: 23894673 DOI: 10.1371/journal.pone.0070570] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/20/2013] [Indexed: 12/21/2022] Open
Abstract
During mouse heart development, ventricular endocardial cells give rise to the coronary arteries by angiogenesis. Myocardially-derived vascular endothelial growth factor-a (Vegfa) regulates embryonic coronary angiogenesis through vascular endothelial growth factor-receptor 2 (Vegfr2) expressed in the endocardium. In this study, we investigated the role of endocardially-produced soluble Vegfr1 (sVegfr1) in the coronary angiogenesis. We deleted sVegfr1 in the endocardium of the developing mouse heart and found that this deletion resulted in a precocious formation of coronary plexuses. Using an ex vivo coronary angiogenesis assay, we showed that the Vegfr1-null ventricular endocardial cells underwent excessive angiogenesis and generated extensive endothelial tubular networks. We also revealed by qPCR analysis that expression of genes involved in the Vegf-Notch pathway was augmented in the Vegfr1-null hearts. We further showed that inhibition of Notch signaling blocked the formation of coronary plexuses by the ventricular endocardial cells. These results establish that Vegfr1 produced in the endocardium negatively regulates embryonic coronary angiogenesis, possibly by limiting the Vegf-Notch signaling.
Collapse
Affiliation(s)
- Zheng Zhang
- The State Key Laboratory of Biotherapy, West China Medical School of Sichuan University, Chengdu, Sichuan, China
| | | |
Collapse
|
18
|
Mending broken hearts: cardiac development as a basis for adult heart regeneration and repair. Nat Rev Mol Cell Biol 2013; 14:529-41. [PMID: 23839576 DOI: 10.1038/nrm3619] [Citation(s) in RCA: 367] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As the adult mammalian heart has limited potential for regeneration and repair, the loss of cardiomyocytes during injury and disease can result in heart failure and death. The cellular processes and regulatory mechanisms involved in heart growth and development can be exploited to repair the injured adult heart through 'reawakening' pathways that are active during embryogenesis. Heart function has been restored in rodents by reprogramming non-myocytes into cardiomyocytes, by expressing transcription factors (GATA4, HAND2, myocyte-specific enhancer factor 2C (MEF2C) and T-box 5 (TBX5)) and microRNAs (miR-1, miR-133, miR-208 and miR-499) that control cardiomyocyte identity. Stimulating cardiomyocyte dedifferentiation and proliferation by activating mitotic signalling pathways involved in embryonic heart growth represents a complementary approach for heart regeneration and repair. Recent advances in understanding the mechanistic basis of heart development offer exciting opportunities for effective therapies for heart failure.
Collapse
|
19
|
Tian X, Hu T, Zhang H, He L, Huang X, Liu Q, Yu W, He L, Yang Z, Zhang Z, Zhong TP, Yang X, Yang Z, Yan Y, Baldini A, Sun Y, Lu J, Schwartz RJ, Evans SM, Gittenberger-de Groot AC, Red-Horse K, Zhou B. Subepicardial endothelial cells invade the embryonic ventricle wall to form coronary arteries. Cell Res 2013; 23:1075-90. [PMID: 23797856 PMCID: PMC3760626 DOI: 10.1038/cr.2013.83] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 04/16/2013] [Accepted: 05/15/2013] [Indexed: 01/03/2023] Open
Abstract
Coronary arteries bring blood flow to the heart muscle. Understanding the developmental program of the coronary arteries provides insights into the treatment of coronary artery diseases. Multiple sources have been described as contributing to coronary arteries including the proepicardium, sinus venosus (SV), and endocardium. However, the developmental origins of coronary vessels are still under intense study. We have produced a new genetic tool for studying coronary development, an AplnCreER mouse line, which expresses an inducible Cre recombinase specifically in developing coronary vessels. Quantitative analysis of coronary development and timed induction of AplnCreER fate tracing showed that the progenies of subepicardial endothelial cells (ECs) both invade the compact myocardium to form coronary arteries and remain on the surface to produce veins. We found that these subepicardial ECs are the major sources of intramyocardial coronary vessels in the developing heart. In vitro explant assays indicate that the majority of these subepicardial ECs arise from endocardium of the SV and atrium, but not from ventricular endocardium. Clonal analysis of Apln-positive cells indicates that a single subepicardial EC contributes equally to both coronary arteries and veins. Collectively, these data suggested that subepicardial ECs are the major source of intramyocardial coronary arteries in the ventricle wall, and that coronary arteries and veins have a common origin in the developing heart.
Collapse
Affiliation(s)
- Xueying Tian
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
The clinical anatomy of the coronary arteries. J Cardiovasc Transl Res 2013; 6:197-207. [PMID: 23423864 DOI: 10.1007/s12265-013-9452-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/01/2013] [Indexed: 01/29/2023]
Abstract
There are a manifold number of variations and anomalies of the origin and course of coronary arteries described in the literature. The incidence of such variations in the general population is reported to range between 0.3 and 1.6 %. Although uncommon, they may be benign or produce symptoms ranging from mild dyspnea to sudden cardiac death, and have been associated with an increased risk of accelerated atherosclerosis and perfusion defects. Thus, in order to effectively utilize the increasing number of therapeutic options available for treating coronary artery diseases, an appreciation of the likely normal and variable arrangements of the coronary arteries is essential. This review will describe the normal anatomy of the coronary arteries as well as the common variations with potential clinical effects.
Collapse
|
21
|
Wu B, Zhang Z, Lui W, Chen X, Wang Y, Chamberlain AA, Moreno-Rodriguez RA, Markwald RR, O'Rourke BP, Sharp DJ, Zheng D, Lenz J, Baldwin HS, Chang CP, Zhou B. Endocardial cells form the coronary arteries by angiogenesis through myocardial-endocardial VEGF signaling. Cell 2013. [PMID: 23178125 DOI: 10.1016/j.cell.2012.10.023] [Citation(s) in RCA: 279] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The origins and developmental mechanisms of coronary arteries are incompletely understood. We show here by fate mapping, clonal analysis, and immunohistochemistry that endocardial cells generate the endothelium of coronary arteries. Dye tracking, live imaging, and tissue transplantation also revealed that ventricular endocardial cells are not terminally differentiated; instead, they are angiogenic and form coronary endothelial networks. Myocardial Vegf-a or endocardial Vegfr-2 deletion inhibited coronary angiogenesis and arterial formation by ventricular endocardial cells. In contrast, lineage and knockout studies showed that endocardial cells make a small contribution to the coronary veins, the formation of which is independent of myocardial-to-endocardial Vegf signaling. Thus, contrary to the current view of a common source for the coronary vessels, our findings indicate that the coronary arteries and veins have distinct origins and are formed by different mechanisms. This information may help develop better cell therapies for coronary artery disease.
Collapse
Affiliation(s)
- Bingruo Wu
- Departments of Genetics, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Coen M, Gabbiani G, Bochaton-Piallat ML. Myofibroblast-mediated adventitial remodeling: an underestimated player in arterial pathology. Arterioscler Thromb Vasc Biol 2012; 31:2391-6. [PMID: 21868702 DOI: 10.1161/atvbaha.111.231548] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The arterial adventitia has been long considered an essentially supportive tissue; however, more and more data suggest that it plays a major role in the modulation of the vascular tone by complex interactions with structures located within intima and media. The purpose of this review is to summarize these data and to describe the mechanisms involved in adventitia/media and adventitia/intima cross-talk. In response to a plethora of stimuli, the adventitia undergoes remodeling processes, resulting in positive (adaptive) remodeling, negative (constrictive) remodeling, or both. The differentiation of the adventitial fibroblast into myofibroblast (MF), a key player of wound healing and fibrosis development, is a hallmark of negative remodeling; this can lead to vessel stenosis and thus contribute to major cardiovascular diseases. The mechanisms of fibroblast-to-MF differentiation and the role of the MF in adventitial remodeling are highlighted herein.
Collapse
Affiliation(s)
- Matteo Coen
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
23
|
Majesky MW, Dong XR, Regan JN, Hoglund VJ. Vascular smooth muscle progenitor cells: building and repairing blood vessels. Circ Res 2011; 108:365-77. [PMID: 21293008 DOI: 10.1161/circresaha.110.223800] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Molecular pathways that control the specification, migration, and number of available smooth muscle progenitor cells play key roles in determining blood vessel size and structure, capacity for tissue repair, and progression of age-related disorders. Defects in these pathways produce malformations of developing blood vessels, depletion of smooth muscle progenitor cell pools for vessel wall maintenance and repair, and aberrant activation of alternative differentiation pathways in vascular disease. A better understanding of the molecular mechanisms that uniquely specify and maintain vascular smooth muscle cell precursors is essential if we are to use advances in stem and progenitor cell biology and somatic cell reprogramming for applications directed to the vessel wall.
Collapse
Affiliation(s)
- Mark W Majesky
- Seattle Children's Research Institute, University of Washington, 1900 Ninth Ave, M/S C9S-5, Seattle, WA 98101, USA.
| | | | | | | |
Collapse
|
24
|
Civelek M, Manduchi E, Riley RJ, Stoeckert CJ, Davies PF. Coronary artery endothelial transcriptome in vivo: identification of endoplasmic reticulum stress and enhanced reactive oxygen species by gene connectivity network analysis. ACTA ACUST UNITED AC 2011; 4:243-52. [PMID: 21493819 DOI: 10.1161/circgenetics.110.958926] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial function is central to the localization of atherosclerosis. The in vivo endothelial phenotypic footprints of arterial bed identity and site-specific atherosusceptibility are addressed. METHODS AND RESULTS Ninety-eight endothelial cell samples from 13 discrete coronary and noncoronary arterial regions of varying susceptibilities to atherosclerosis were isolated from 76 normal swine. Transcript profiles were analyzed to determine the steady-state in vivo endothelial phenotypes. An unsupervised systems biology approach using weighted gene coexpression networks showed highly correlated endothelial genes. Connectivity network analysis identified 19 gene modules, 12 of which showed significant association with circulatory bed classification. Differential expression of 1300 genes between coronary and noncoronary artery endothelium suggested distinct coronary endothelial phenotypes, with highest significance expressed in gene modules enriched for biological functions related to endoplasmic reticulum (ER) stress and unfolded protein binding, regulation of transcription and translation, and redox homeostasis. Furthermore, within coronary arteries, comparison of endothelial transcript profiles of susceptible proximal regions to protected distal regions suggested the presence of ER stress conditions in susceptible sites. Accumulation of reactive oxygen species throughout coronary endothelium was greater than in noncoronary endothelium consistent with coronary artery ER stress and lower endothelial expression of antioxidant genes in coronary arteries. CONCLUSIONS Gene connectivity analyses discriminated between coronary and noncoronary endothelial transcript profiles and identified differential transcript levels associated with increased ER and oxidative stress in coronary arteries consistent with enhanced susceptibility to atherosclerosis.
Collapse
Affiliation(s)
- Mete Civelek
- Department of Bioengineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
25
|
del Monte G, Casanova JC, Guadix JA, MacGrogan D, Burch JB, Pérez-Pomares JM, de la Pompa JL. Differential Notch Signaling in the Epicardium Is Required for Cardiac Inflow Development and Coronary Vessel Morphogenesis. Circ Res 2011; 108:824-36. [DOI: 10.1161/circresaha.110.229062] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
The proepicardium is a transient structure comprising epicardial progenitor cells located at the posterior limit of the embryonic cardiac inflow. A network of signals regulates proepicardial cell fate and defines myocardial and nonmyocardial domains at the venous pole of the heart. During cardiac development, epicardial-derived cells also contribute to coronary vessel morphogenesis.
Objective:
To study Notch function during proepicardium development and coronary vessel formation in the mouse.
Methods and Results:
Using in situ hybridization, RT-PCR, and immunohistochemistry, we find that Notch pathway elements are differentially activated throughout the proepicardial–epicardial–coronary transition. Analysis of
RBPJk
-targeted embryos indicates that Notch ablation causes ectopic procardiogenic signaling in the proepicardium that in turn promotes myocardial differentiation in adjacent mesodermal progenitors, resulting in a premature muscularization of the sinus venosus horns. Epicardium-specific
Notch1
ablation using a
Wt1-Cre
driver line disrupts coronary artery differentiation, reduces myocardium wall thickness and myocyte proliferation, and reduces
Raldh2
expression. Ectopic Notch1 activation disrupts epicardium development and causes thinning of ventricular walls.
Conclusions:
Epicardial Notch modulates cell differentiation in the proepicardium and adjacent pericardial mesoderm. Notch1 is later required for arterial endothelium commitment and differentiation and for vessel wall maturation during coronary vessel development and myocardium growth.
Collapse
Affiliation(s)
- Gonzalo del Monte
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - Jesús C. Casanova
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - Juan Antonio Guadix
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - Donal MacGrogan
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - John B.E. Burch
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - José María Pérez-Pomares
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| | - José Luis de la Pompa
- From the Laboratorio de Biología Celular y del Desarrollo (G.d.M., J.C.C., D.M., J.L.d.l.P.), Dpto de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain; Departamento de Biología Animal (J.A.G., J.M.P.-P.), Facultad de Ciencias, Universidad de Málaga, Spain; and Fox Chase Cancer Center (J.B.E.B.), Philadelphia PA
| |
Collapse
|
26
|
Combs MD, Braitsch CM, Lange AW, James JF, Yutzey KE. NFATC1 promotes epicardium-derived cell invasion into myocardium. Development 2011; 138:1747-57. [PMID: 21447555 DOI: 10.1242/dev.060996] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Epicardium-derived cells (EPDCs) contribute to formation of coronary vessels and fibrous matrix of the mature heart. Nuclear factor of activated T-cells cytoplasmic 1 (NFATC1) is expressed in cells of the proepicardium (PE), epicardium and EPDCs in mouse and chick embryos. Conditional loss of NFATC1 expression in EPDCs in mice causes embryonic death by E18.5 with reduced coronary vessel and fibrous matrix penetration into myocardium. In osteoclasts, calcineurin-mediated activation of NFATC1 by receptor activator of NFκB ligand (RANKL) signaling induces cathepsin K (CTSK) expression for extracellular matrix degradation and cell invasion. RANKL/NFATC1 pathway components also are expressed in EPDCs, and loss of NFATC1 in EPDCs causes loss of CTSK expression in the myocardial interstitium in vivo. Likewise, RANKL treatment induces Ctsk expression in PE-derived cell cultures via a calcineurin-dependent mechanism. In chicken embryo hearts, RANKL treatment increases the distance of EPDC invasion into myocardium, and this response is calcineurin dependent. Together, these data demonstrate a crucial role for the RANKL/NFATC1 signaling pathway in promoting invasion of EPDCs into the myocardium by induction of extracellular matrix-degrading enzyme gene expression.
Collapse
Affiliation(s)
- Michelle D Combs
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center ML7020, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
27
|
Tomanek RJ, Christensen LP, Simons M, Murakami M, Zheng W, Schatteman GC. Embryonic coronary vasculogenesis and angiogenesis are regulated by interactions between multiple FGFs and VEGF and are influenced by mesenchymal stem cells. Dev Dyn 2011; 239:3182-91. [PMID: 20981833 DOI: 10.1002/dvdy.22460] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In embryonic hearts explanted on collagen gels, epicardial cells delaminate and form vascular tubes, thus providing a model for coronary tubulogenesis. Using this model, we show that fibroblast growth factors (FGFs) 1, 2, 4, 8, 9, and 18 contribute to tubulogenesis and that the availability of multiple FGFs provides the optimal tubulogenic response. Moreover, the FGF effects are vascular endothelial growth factor (VEGF) -dependent, while VEGF-induced tubulogenesis requires FGF signaling. The number of endothelial cells (ECs) is increased by all of the FGFs, while EC migration is significantly enhanced only by FGF-2 and FGF-18. Finally, addition of embryonic mesenchymal stem cells (EMSC) to the explants markedly enhances EC numbers and a 23-fold increase in stromal derived factor-1α (SDF-1α), which is FGF dependent. Both explants and EMSCs produce SDF-1α. In conclusion, coronary tubulogenesis of embryonic epicardium: (1) is responsive to many FGF family members, (2) requires both FGF and VEGFA signaling, and (3) is responsive to EMSCs.
Collapse
Affiliation(s)
- Robert J Tomanek
- Department of Anatomy and Cell Biology, The University of Iowa Carver College of Medicine and The Cardiovascular Center, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Holtz ML, Misra RP. Serum response factor is required for cell contact maintenance but dispensable for proliferation in visceral yolk sac endothelium. BMC DEVELOPMENTAL BIOLOGY 2011; 11:18. [PMID: 21401944 PMCID: PMC3065428 DOI: 10.1186/1471-213x-11-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 03/14/2011] [Indexed: 01/14/2023]
Abstract
Background Endothelial-specific knockout of the transcription factor serum response factor (SRF) results in embryonic lethality by mid-gestation. The associated phenotype exhibits vascular failure in embryos as well as visceral yolk sac (VYS) tissues. Previous data suggest that this vascular failure is caused by alterations in cell-cell and cell-matrix contacts. In the current study, we sought to more carefully address the role of SRF in endothelial function and cell contact interactions in VYS tissues. Results Tie2-Cre recombinase-mediated knockout of SRF expression resulted in loss of detectable SRF from VYS mesoderm by E12.5. This loss was accompanied by decreased expression of smooth muscle alpha-actin as well as vascular endothelial cadherin and claudin 5, endothelial-specific components of adherens and tight junctions, respectively. Focal adhesion (FA) integrins alpha5 and beta1 were largely unchanged in contrast to loss of the FA-associated molecule vinculin. The integrin binding partner fibronectin-1 was also profoundly decreased in the extracellular matrix, indicating another aspect of impaired adhesive function and integrin signaling. Additionally, cells in SRF-null VYS mesoderm failed to reduce proliferation, suggesting not only that integrin-mediated contact inhibition is impaired but also that SRF protein is not required for proliferation in these cells. Conclusions Our data support a model in which SRF is critical in maintaining functional cell-cell and cell-matrix adhesion in endothelial cells. Furthermore, we provide evidence that supports a model in which loss of SRF protein results in a sustained proliferation defect due in part to failed integrin signaling.
Collapse
Affiliation(s)
- Mary L Holtz
- Department of Biochemistry, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
29
|
Coronary arteries form by developmental reprogramming of venous cells. Nature 2010; 464:549-53. [PMID: 20336138 DOI: 10.1038/nature08873] [Citation(s) in RCA: 401] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 02/04/2010] [Indexed: 12/31/2022]
Abstract
Coronary artery disease is the leading cause of death worldwide. Determining the coronary artery developmental program could aid understanding of the disease and lead to new treatments, but many aspects of the process, including their developmental origin, remain obscure. Here we show, using histological and clonal analysis in mice and cardiac organ culture, that coronary vessels arise from angiogenic sprouts of the sinus venosus-the vein that returns blood to the embryonic heart. Sprouting venous endothelial cells dedifferentiate as they migrate over and invade the myocardium. Invading cells differentiate into arteries and capillaries; cells on the surface redifferentiate into veins. These results show that some differentiated venous cells retain developmental plasticity, and indicate that position-specific cardiac signals trigger their dedifferentiation and conversion into coronary arteries, capillaries and veins. Understanding this new reprogramming process and identifying the endogenous signals should suggest more natural ways of engineering coronary bypass grafts and revascularizing the heart.
Collapse
|
30
|
Endothelial cell lineages of the heart. Cell Tissue Res 2008; 335:67-73. [PMID: 18682987 DOI: 10.1007/s00441-008-0663-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2008] [Accepted: 06/10/2008] [Indexed: 02/07/2023]
Abstract
During early gastrulation, vertebrate embryos begin to produce endothelial cells (ECs) from the mesoderm. ECs first form primitive vascular plexus de novo and later differentiate into arterial, venous, capillary, and lymphatic ECs. In the heart, the five distinct EC types (endocardial, coronary arterial, venous, capillary, and lymphatic) have distinct phenotypes. For example, coronary ECs establish a typical vessel network throughout the myocardium, whereas endocardial ECs form a large epithelial sheet with no angiogenic sprouting into the myocardium. Neither coronary arteries, veins, and capillaries, nor lymphatic vessels fuse with the endocardium or open to the heart chamber. The developmental stage during which the specific phenotype of each cardiac EC type is determined remains unclear. The mechanisms involved in EC commitment and diversity can however be more precisely defined by tracking the migratory patterns and lineage decisions of the precursors of cardiac ECs.
Collapse
|
31
|
The influence of proepicardial cells on the osteogenic potential of marrow stromal cells in a three-dimensional tubular scaffold. Biomaterials 2008; 29:2203-16. [PMID: 18289664 DOI: 10.1016/j.biomaterials.2008.01.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 01/27/2008] [Indexed: 11/22/2022]
Abstract
It is well established that the process of neovascularization or neoangiogenesis is coupled to the development and maturation of bone. Bone marrow stromal cells (BMSCs) or mesenchymal stem cells (MSCs) comprise a heterogeneous population of cells that can be differentiated in vitro into both mesenchymal and non-mesenchymal cell lineages. When both rat BMSCs and quail proepicardia (PEs) were seeded onto a three-dimensional (3-D) tubular scaffold engineered from aligned collagen type I strands and co-cultured in osteogenic media, the maturation and co-differentiation into osteoblastic and vascular cell lineages were observed. In addition, these cells produced abundant mineralized extracellular matrix materials and vessel-like structures. BMSCs were seeded at a density of 2 x 10(6)cells/15 mm tube and cultured in basal media for 3 days. Subsequently, on day 3, PEs were seeded onto the same tubes and the co-culture was continued for another 3, 6 or 9 days either in basal or in osteogenic media. Differentiated cells were subjected to immunohistochemical, cytochemical and biochemical analyses. Phenotypic induction was analyzed at mRNA level by reverse transcriptase quantitative polymerase chain reaction (RT-qPCR). Immunolocalization of key osteogenic and vasculogenic lineage specific markers were examined using confocal scanning laser microscopy. In osteogenic tube cultures, both early and late osteogenic markers were observed and were reminiscent of in vivo expression pattern. Alkaline phosphatase activity and calcium content significantly increased over the observed period of time in osteogenic medium. Abundant interlacing fascicles of QCPN, QH1, isolectin and alpha-smooth muscle actin (alpha-SMA) positive cells were observed in these tube cultures. These cells formed extensive arborizations of nascent capillary-like structures and were seen amidst the developing osteoblasts in osteogenic cultures. The 3-D culture system not only generated de novo vessel-like structures but also augmented the maturation and differentiation of BMSCs into osteoblasts. Thus, this novel co-culture system provides a useful in vitro model to investigate the functional role and effects of neovascularization in the proliferation, differentiation and maturation of BMSC derived osteoblasts.
Collapse
|
32
|
Abstract
This chapter summarizes experimental techniques used to study coronary vessel development from its origins in the proepicardium (PE) to the final assembled network of arteries, veins, and capillaries present in the mature heart. Methods are described for microdissection and culture of the PE and embryonic epicardial cells, isolation of total RNA from single PE primordia and analysis by RT-PCR, imaging of the epicardium and coronary vessels by whole-mount confocal microscopy and by scanning electron microscopy, and the preparation of coronary vascular corrosion casts to visualize the entire coronary artery network structure. These techniques form the basic tools to study the cellular and molecular pathways that guide development and remodeling of coronary vessels.
Collapse
Affiliation(s)
- Xiu Rong Dong
- Carolina Cardiovascular Biology Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | |
Collapse
|
33
|
Montano MM, Doughman YQ, Deng H, Chaplin L, Yang J, Wang N, Zhou Q, Ward NL, Watanabe M. Mutation of the HEXIM1 gene results in defects during heart and vascular development partly through downregulation of vascular endothelial growth factor. Circ Res 2007; 102:415-22. [PMID: 18079413 DOI: 10.1161/circresaha.107.157859] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Our previous studies and those of others indicated that the transcription factor Hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) is a tumor suppressor and cyclin-dependent kinase inhibitor, and that these HEXIM1 functions are mainly dependent on its C-terminal region. We provide evidence here that the HEXIM1 C-terminal region is critical for cardiovascular development. HEXIM1 protein was detected in the heart during critical time periods in cardiac growth and chamber maturation. We created mice carrying an insertional mutation in the HEXIM1 gene that disrupted its C-terminal region and found that this resulted in prenatal lethality. Heart defects in HEXIM1(1 to 312) mice included abnormal coronary patterning and thin ventricular walls. The thin myocardium can be partly attributed to increased apoptosis. Platelet endothelial cell adhesion molecular precursor-1 staining of HEXIM1(1 to 312) heart sections revealed decreased vascularization of the myocardium despite the presence of coronary vasculature in the epicardium. The expression of vascular endothelial growth factor (VEGF), known to affect angioblast invasion and myocardial proliferation and survival, was decreased in HEXIM1(1 to 312) mice compared with control littermates. We also observed decreased fibroblast growth factor 9 (FGF9) expression, suggesting that effects of HEXIM1 in the myocardium are partly mediated through epicardial FGF9 signaling. Together our results suggest that HEXIM1 plays critical roles in coronary vessel development and myocardial growth. The basis for this role of HEXIM1 is that VEGF is a direct transcriptional target of HEXIM1, and involves attenuation a repressive effects of C/EBPalpha on VEGF gene transcription.
Collapse
Affiliation(s)
- Monica M Montano
- Department of Pharmacology, Case Western Reserve University School of Medicine, H.G. Wood Bldg W307, 2109 Adelbert Rd, Cleveland, Ohio 44106, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ishii Y, Langberg JD, Hurtado R, Lee S, Mikawa T. Induction of proepicardial marker gene expression by the liver bud. Development 2007; 134:3627-37. [PMID: 17855432 DOI: 10.1242/dev.005280] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cells of the coronary vessels arise from a unique extracardiac mesothelial cell population, the proepicardium, which develops posterior to the sinoatrial region of the looping-stage heart. Although contribution of the proepicardial cells to cardiac development has been studied extensively, it remains unresolved how the proepicardium is induced and specified in the mesoderm during embryogenesis. It is known, however, that the proepicardium develops from the mesothelium that overlays the liver bud. Here, we show that the expression of proepicardial marker genes - Wt1, capsulin (epicardin, pod1, Tcf21) and Tbx18, can be induced in naïve mesothelial cells by the liver bud, both in vitro and in vivo. Lateral embryonic explants, when co-cultured with the liver bud, were induced to express these proepicardial marker genes. The same induction of the marker genes was detected in vivo when a quail liver bud was implanted in the posterior-lateral regions of a chick embryo. This ectopic induction of marker gene expression was not evident when other endodermal tissues, such as the lung bud or stomach, were implanted. This inductive response to the liver bud was not detectable in host embryos before stage 12 (16-somite stage). These results suggest that, after a specific developmental stage, a large area of the mesothelium becomes competent to express proepicardial marker genes in response to localized liver-derived signal(s). The developmentally regulated competency of mesothelium and a localized inductive signal might play a role in restricting the induction of the proepicardial marker gene expression to a specific region of the mesothelium. The data might also provide a foundation for future engineering of a coronary vascular progenitor population.
Collapse
Affiliation(s)
- Yasuo Ishii
- University of California San Francisco, Cardiovascular Research Institute, Box 2711, Rock Hall Room 384D, 1550 4th Street, San Francisco, CA 94158-2324, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
The origins of vascular smooth muscle are far more diverse than previously thought. Lineage mapping studies show that the segmental organization of early vertebrate embryos leaves footprints on the adult vascular system in the form of a mosaic pattern of different smooth muscle types. Moreover, evolutionarily conserved tissue forming pathways produce vascular smooth muscle from a variety of unanticipated sources. A closer look at the diversity of smooth muscle origins in vascular development provides new perspectives about how blood vessels differ from one another and why they respond in disparate ways to common risk factors associated with vascular disease. The origins of vascular smooth muscle are far more diverse than previously thought. A closer look at the diversity of smooth muscle origins in vascular development provides new perspectives about how blood vessels differ from one another and why they respond in disparate ways to common risk factors associated with vascular disease.
Collapse
Affiliation(s)
- Mark W Majesky
- Department of Medicine, Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599-7126, USA.
| |
Collapse
|
36
|
Tomanek RJ, Hansen HK, Dedkov EI. Vascular patterning of the quail coronary system during development. ACTA ACUST UNITED AC 2006; 288:989-99. [PMID: 16892426 DOI: 10.1002/ar.a.20365] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Recent studies have provided insights into specific events that contribute to vasculogenesis and angiogenesis in the developing coronary vasculature. This study focused on the developmental progression of coronary vascularization beginning with tube formation and ending with the establishment of a coronary arterial tree. We used electron microscopy, histology of serial sections, and immunohistochemistry in order to provide a comprehensive view of coronary vessel formation during the embryonic and fetal periods of the quail heart, a species that has been used in a number of studies addressing myocardial vascularization. Our data reveal features of progenitor cells and blood islands, tubular formation, and the anatomical relationship of a transformed periarterial tubular network and sympathetic ganglia to the emergence and branching of the right and left coronary arteries. We have traced the pattern of coronary artery branching and documented its innervation. Finally, our data include the relationship of fibronectin, laminin, and apoptosis to coronary artery growth. Our findings bring together morphological events that occur over the embryonic and fetal periods and provide a baseline for studies into the mechanisms that regulate the various events that occur during these time periods.
Collapse
Affiliation(s)
- Robert J Tomanek
- Department of Anatomy and Cell Biology and Cardiovascular Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
37
|
Nesbitt T, Lemley A, Davis J, Yost MJ, Goodwin RL, Potts JD. Epicardial development in the rat: a new perspective. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2006; 12:390-8. [PMID: 16984665 DOI: 10.1017/s1431927606060533] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2006] [Accepted: 06/02/2006] [Indexed: 05/11/2023]
Abstract
Development of the epicardium is critical to proper heart formation. It provides all of the precursor cells that form the coronary system and supplies signals that stimulate cardiac myocyte proliferation. The epicardium forms from mesothelial cells associated with the septum transversum and is referred to as the proepicardium (PE). Two different methods by which these PE cells colonize the developing heart have been described. In avians, PE cells form a bridge to the heart over which PE cells migrate onto the heart. In fish and mammals, PE cells form vesicles of cells that detach from the mesothelium, float through the pericardial cavity, and attach to the heart. A previous study of rat PE development investigated this process at the histological level. Protein markers have been developed since this study. Thus, we investigated this important developmental process coupled with these new markers using other visualization techniques such as scanning electron microscopy (SEM) and confocal microscopy. Finally, a novel, three-dimensional (3-D) culture system was used to confirm the identity of the PE cells. In this study, we found convincing evidence that the rat PE cells directly attach to the heart in a manner similar to that observed in avians.
Collapse
Affiliation(s)
- Tresa Nesbitt
- Department of Cell & Developmental Biology and Anatomy, University of South Carolina, School of Medicine, Columbia, SC 29209, USA
| | | | | | | | | | | |
Collapse
|
38
|
Tomanek RJ, Ishii Y, Holifield JS, Sjogren CL, Hansen HK, Mikawa T. VEGF family members regulate myocardial tubulogenesis and coronary artery formation in the embryo. Circ Res 2006; 98:947-53. [PMID: 16527987 DOI: 10.1161/01.res.0000216974.75994.da] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This study tested the hypothesis that coronary tubulogenesis and coronary artery formation require VEGF family members. Quail embryos were injected with soluble vascular endothelial growth factor (VEGF) receptors R1 (Flt-1), R2 (Flk-1), R3 (Flt-4), VEGF-Trap (a chimera of R1 and R2), or neutralizing antibodies to VEGF-A, VEGF-B, or fibroblast growth factor (FGF)-2. Our data document that tubulogenesis is temporally dependent on multiple VEGF family members, because the early stage of tubulogenesis was markedly inhibited by VEGF-Trap and to a lesser extent by soluble VEGFR-1. Some inhibition of tubulogenesis was documented when anti-FGF-2, but not anti-VEGF-A, antibodies were injected at embryonic day 6 (E6). Most importantly, we found that VEGF-Trap injected at either E6 or E7 prevented the formation of coronary arteries. Soluble VEGFR-1 and soluble VEGFR-2 modified the formation of coronary arteries, whereas soluble VEGFR-3 was without effect. Antibodies to VEGF-B, but not VEGF-A, had a strong inhibitory effect on coronary artery development. The absence of coronary artery stems, and thus a functional coronary circulation, in the embryos injected with VEGF-Trap caused an accumulation of erythrocytes in the subepicardium and muscular interventricular septum. Using retroviral cell tagging, we showed that some of the erythrocytes in blood islands and small vascular tubes were progeny of the proepicardium. Thus, another salient finding of this study is the first definitive documentation of proepicardially derived hemangioblasts, which can differentiate into erythrocytes.
Collapse
Affiliation(s)
- Robert J Tomanek
- Department of Anatomy & Cell Biology, Cardiovascular Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Ferguson JE, Kelley RW, Patterson C. Mechanisms of endothelial differentiation in embryonic vasculogenesis. Arterioscler Thromb Vasc Biol 2005; 25:2246-54. [PMID: 16123328 DOI: 10.1161/01.atv.0000183609.55154.44] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The formation of new blood vessels in the adult organism not only contributes to the progression of diseases such as cancer and diabetic retinopathy but also can be promoted in therapeutic approaches to various ischemic pathologies. Because many of the signals important to blood vessel development during embryogenesis are recapitulated during adult blood vessel formation, much work has been performed to better-understand the molecular control of endothelial differentiation in the developing embryo. In this review, we describe the current understanding of where endothelial differentiation from pluripotent progenitor cells occurs during development, how this process is controlled at the molecular level, and what model systems can be used to investigate the earliest steps of blood vessel formation.
Collapse
Affiliation(s)
- J E Ferguson
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599-7126, USA
| | | | | |
Collapse
|