101
|
Sawada H, Rateri DL, Moorleghen JJ, Majesky MW, Daugherty A. Smooth Muscle Cells Derived From Second Heart Field and Cardiac Neural Crest Reside in Spatially Distinct Domains in the Media of the Ascending Aorta-Brief Report. Arterioscler Thromb Vasc Biol 2017; 37:1722-1726. [PMID: 28663257 DOI: 10.1161/atvbaha.117.309599] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/20/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Smooth muscle cells (SMCs) of the proximal thoracic aorta are embryonically derived from the second heart field (SHF) and cardiac neural crest (CNC). However, distributions of these embryonic origins are not fully defined. The regional distribution of SMCs of different origins is speculated to cause region-specific aortopathies. Therefore, the aim of this study was to determine the distribution of SMCs of SHF and CNC origins in the proximal thoracic aorta. APPROACH AND RESULTS Mice with repressed LacZ in the ROSA26 locus were bred to those expressing Cre controlled by either the Wnt1 or Mef2c (myocyte-specific enhancer factor 2c) promoter to trace CNC- and SHF-derived SMCs, respectively. Thoracic aortas were harvested, and activity of β-galactosidase was determined. Aortas from Wnt1-Cre mice had β-galactosidase-positive areas throughout the region from the proximal ascending aorta to just distal of the subclavian arterial branch. Unexpectedly, β-galactosidase-positive areas in Mef2c-Cre mice extended from the aortic root throughout the ascending aorta. This distribution occurred independent of sex and aging. Cross and sagittal aortic sections demonstrated that CNC-derived cells populated the inner medial aspect of the anterior region of the ascending aorta and transmurally in the media of the posterior region. Interestingly, outer medial cells throughout anterior and posterior ascending aortas were derived from the SHF. β-Galactosidase-positive medial cells of both origins colocalized with an SMC marker, α-actin. CONCLUSIONS Both CNC- and SHF-derived SMCs populate the media throughout the ascending aorta. The outer medial cells of the ascending aorta form a sleeve populated by SHF-derived SMCs.
Collapse
Affiliation(s)
- Hisashi Sawada
- From the Saha Cardiovascular Research Center (H.S., D.L.R., J.J.M., A.D.) and Department of Physiology (A.D.), University of Kentucky, Lexington; Seattle Children's Research Institute, Washington (M.W.M.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Debra L Rateri
- From the Saha Cardiovascular Research Center (H.S., D.L.R., J.J.M., A.D.) and Department of Physiology (A.D.), University of Kentucky, Lexington; Seattle Children's Research Institute, Washington (M.W.M.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Jessica J Moorleghen
- From the Saha Cardiovascular Research Center (H.S., D.L.R., J.J.M., A.D.) and Department of Physiology (A.D.), University of Kentucky, Lexington; Seattle Children's Research Institute, Washington (M.W.M.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Mark W Majesky
- From the Saha Cardiovascular Research Center (H.S., D.L.R., J.J.M., A.D.) and Department of Physiology (A.D.), University of Kentucky, Lexington; Seattle Children's Research Institute, Washington (M.W.M.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center (H.S., D.L.R., J.J.M., A.D.) and Department of Physiology (A.D.), University of Kentucky, Lexington; Seattle Children's Research Institute, Washington (M.W.M.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.).
| |
Collapse
|
102
|
Levchuk KA, Malashicheva AB. Regional functional heterogeneity of aortic smooth muscle cells in rats. Russ J Dev Biol 2017. [DOI: 10.1134/s1062360417030080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
103
|
Poelmann RE, Gittenberger-de Groot AC, Biermans MWM, Dolfing AI, Jagessar A, van Hattum S, Hoogenboom A, Wisse LJ, Vicente-Steijn R, de Bakker MAG, Vonk FJ, Hirasawa T, Kuratani S, Richardson MK. Outflow tract septation and the aortic arch system in reptiles: lessons for understanding the mammalian heart. EvoDevo 2017; 8:9. [PMID: 28491275 PMCID: PMC5424407 DOI: 10.1186/s13227-017-0072-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/03/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac outflow tract patterning and cell contribution are studied using an evo-devo approach to reveal insight into the development of aorto-pulmonary septation. RESULTS We studied embryonic stages of reptile hearts (lizard, turtle and crocodile) and compared these to avian and mammalian development. Immunohistochemistry allowed us to indicate where the essential cell components in the outflow tract and aortic sac were deployed, more specifically endocardial, neural crest and second heart field cells. The neural crest-derived aorto-pulmonary septum separates the pulmonary trunk from both aortae in reptiles, presenting with a left visceral and a right systemic aorta arising from the unseptated ventricle. Second heart field-derived cells function as flow dividers between both aortae and between the two pulmonary arteries. In birds, the left visceral aorta disappears early in development, while the right systemic aorta persists. This leads to a fusion of the aorto-pulmonary septum and the aortic flow divider (second heart field population) forming an avian aorto-pulmonary septal complex. In mammals, there is also a second heart field-derived aortic flow divider, albeit at a more distal site, while the aorto-pulmonary septum separates the aortic trunk from the pulmonary trunk. As in birds there is fusion with second heart field-derived cells albeit from the pulmonary flow divider as the right 6th pharyngeal arch artery disappears, resulting in a mammalian aorto-pulmonary septal complex. In crocodiles, birds and mammals, the main septal and parietal endocardial cushions receive neural crest cells that are functional in fusion and myocardialization of the outflow tract septum. Longer-lasting septation in crocodiles demonstrates a heterochrony in development. In other reptiles with no indication of incursion of neural crest cells, there is either no myocardialized outflow tract septum (lizard) or it is vestigial (turtle). Crocodiles are unique in bearing a central shunt, the foramen of Panizza, between the roots of both aortae. Finally, the soft-shell turtle investigated here exhibits a spongy histology of the developing carotid arteries supposedly related to regulation of blood flow during pharyngeal excretion in this species. CONCLUSIONS This is the first time that is shown that an interplay of second heart field-derived flow dividers with a neural crest-derived cell population is a variable but common, denominator across all species studied for vascular patterning and outflow tract septation. The observed differences in normal development of reptiles may have impact on the understanding of development of human congenital outflow tract malformations.
Collapse
Affiliation(s)
- Robert E Poelmann
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | | | - Marcel W M Biermans
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Anne I Dolfing
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Armand Jagessar
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Sam van Hattum
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Amanda Hoogenboom
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Rebecca Vicente-Steijn
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Merijn A G de Bakker
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Freek J Vonk
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands.,Naturalis Biodiversity Center, Darwinweg 2, Leiden, The Netherlands
| | - Tatsuya Hirasawa
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Michael K Richardson
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| |
Collapse
|
104
|
Milewski RK, Habertheuer A, Bavaria JE, Siki M, Szeto WY, Krause E, Korutla V, Desai ND, Vallabhajosyula P. Fate of remnant sinuses of Valsalva in patients with bicuspid and trileaflet valves undergoing aortic valve, ascending aorta, and aortic arch replacement. J Thorac Cardiovasc Surg 2017; 154:421-432. [PMID: 28599977 DOI: 10.1016/j.jtcvs.2017.03.150] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 02/26/2017] [Accepted: 03/26/2017] [Indexed: 11/25/2022]
Abstract
OBJECTIVE In patients presenting with aortic valvulopathy with concomitant ascending aortic aneurysm, surgical management of the sinus of Valsalva segment remains undefined, especially for moderately dilated aortic roots. In patients with this pathology undergoing aortic valve replacement with supracoronary ascending aorta replacement, we assessed the fate of the remnant preserved sinus of Valsalva segment stratified by aortic valve morphology and pathology. METHODS From 2002 to 2015, 428 patients underwent elective aortic valve replacement with supracoronary ascending aorta replacement. Patients were stratified on the basis of valvular morphology (bicuspid aortic valve [n = 254] and tricuspid aortic valve [n = 174]), valvular pathology (bicuspid aortic valve with aortic stenosis [n = 178], bicuspid aortic valve with aortic insufficiency [n = 76], tricuspid aortic valve with aortic stenosis [n = 61], tricuspid aortic valve with aortic insufficiency [n = 113]), and preoperative sinus of Valsalva dimensions (<40, 40-45, >45 mm). RESULTS Kaplan-Meier analysis revealed no significant difference in freedom from reoperation in tricuspid aortic valve versus bicuspid aortic valve (P = .576). Multivariable Cox regression model performed with sinus of Valsalva dimensions at baseline and follow-up as time-varying covariates did not adversely affect survival. A repeated-measure, mixed-effects model constructed to assess longitudinal sinus of Valsalva trends revealed that the retained sinus of Valsalva dimensions remain stable over long-term follow-up (discharge to ≥10 years), irrespective of valvular morphology/pathology (bicuspid aortic valve with aortic insufficiency, tricuspid aortic valve with aortic insufficiency, tricuspid aortic valve with aortic stenosis) and preoperative sinus of Valsalva groups (<40, 40-45, >45 mm). CONCLUSIONS In patients with nonaneurysmal sinuses of Valsalva undergoing aortic valve replacement with supracoronary ascending aorta replacement, the sinus segment can be preserved irrespective of the type of valvular pathology (aortic stenosis vs aortic insufficiency) or valvular morphology (bicuspid aortic valve vs tricuspid aortic valve). Aortic valve replacement with supracoronary ascending aorta replacement may have a stabilizing effect on the sinus segment over long-term follow-up in patients with tricuspid aortic valves or bicuspid aortic valves.
Collapse
Affiliation(s)
| | - Andreas Habertheuer
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Joseph E Bavaria
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Mary Siki
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Wilson Y Szeto
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Eric Krause
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Varun Korutla
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | - Nimesh D Desai
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pa
| | | |
Collapse
|
105
|
Francou A, De Bono C, Kelly RG. Epithelial tension in the second heart field promotes mouse heart tube elongation. Nat Commun 2017; 8:14770. [PMID: 28357999 PMCID: PMC5379109 DOI: 10.1038/ncomms14770] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 01/27/2017] [Indexed: 12/29/2022] Open
Abstract
Extension of the vertebrate heart tube is driven by progressive addition of second heart field (SHF) progenitor cells to the poles of the heart. Defects in this process cause a spectrum of congenital anomalies. SHF cells form an epithelial layer in splanchnic mesoderm in the dorsal wall of the pericardial cavity. Here we report oriented cell elongation, polarized actomyosin distribution and nuclear YAP/TAZ in a proliferative centre in the posterior dorsal pericardial wall during heart tube extension. These parameters are indicative of mechanical stress, further supported by analysis of cell shape changes in wound assays. Time course and mutant analysis identifies SHF deployment as a source of epithelial tension. Moreover, cell division and oriented growth in the dorsal pericardial wall align with the axis of cell elongation, suggesting that epithelial tension in turn contributes to heart tube extension. Our results implicate tissue-level forces in the regulation of heart tube extension. Epithelial progenitor cell growth in the second heart field contributes to heart morphogenesis but how this is regulated at the tissue level is unclear. Here, the authors show that cell elongation, polarized actomyosin and nuclear YAP/TAZ drive epithelial growth and correlate with mechanical tension.
Collapse
Affiliation(s)
- Alexandre Francou
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Christopher De Bono
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Robert G Kelly
- Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, Campus De Luminy Case 907, 13288 Marseille Cedex 9, France
| |
Collapse
|
106
|
Wang X, Chen D, Chen K, Jubran A, Ramirez A, Astrof S. Endothelium in the pharyngeal arches 3, 4 and 6 is derived from the second heart field. Dev Biol 2016; 421:108-117. [PMID: 27955943 DOI: 10.1016/j.ydbio.2016.12.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 12/22/2022]
Abstract
Oxygenated blood from the heart is directed into the systemic circulation through the aortic arch arteries (AAAs). The AAAs arise by remodeling of three symmetrical pairs of pharyngeal arch arteries (PAAs), which connect the heart with the paired dorsal aortae at mid-gestation. Aberrant PAA formation results in defects frequently observed in patients with lethal congenital heart disease. How the PAAs form in mammals is not understood. The work presented in this manuscript shows that the second heart field (SHF) is the major source of progenitors giving rise to the endothelium of the pharyngeal arches 3 - 6, while the endothelium in the pharyngeal arches 1 and 2 is derived from a different source. During the formation of the PAAs 3 - 6, endothelial progenitors in the SHF extend cellular processes toward the pharyngeal endoderm, migrate from the SHF and assemble into a uniform vascular plexus. This plexus then undergoes remodeling, whereby plexus endothelial cells coalesce into a large PAA in each pharyngeal arch. Taken together, our studies establish a platform for investigating cellular and molecular mechanisms regulating PAA formation and alterations that lead to disease.
Collapse
Affiliation(s)
- Xia Wang
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | - Dongying Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kelley Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Jefferson Medical College of Thomas Jefferson University, Clinical & Translational Research Track, USA
| | - Ali Jubran
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; The Master's of Science Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - AnnJosette Ramirez
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sophie Astrof
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
107
|
Peterss S, Bhandari R, Rizzo JA, Fang H, Kuzmik GA, Ziganshin BA, Elefteriades JA. The Aortic Root: Natural History After Root-Sparing Ascending Replacement in Nonsyndromic Aneurysmal Patients. Ann Thorac Surg 2016; 103:828-833. [PMID: 27659602 DOI: 10.1016/j.athoracsur.2016.06.081] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Leaving native aortic tissue in situ in root-sparing ascending aortic replacement raises concern regarding potential later need for root reoperation or for the potential occurrence of localized dissections or rupture in the residual root. The purpose of this study was to evaluate the natural growth of the aortic root after root-sparing aortic replacement. METHODS In all, 102 consecutive patients (mean age 61.8 ± 12.5 years; 60% male) who had undergone root-sparing aortic replacement had sufficient retrievable information regarding their aortic root diameter at postoperative baseline and follow-up imaging by computed tomography or echocardiography. The annual growth rate was evaluated and also compared according to the influence of valve morphology and concomitant aortic valve replacement. Furthermore, the years of natural history that would require for root enlargement to meet a 50 mm threshold of the root diameter were calculated. RESULTS The estimated growth rate of the aortic root after root-sparing aortic replacement is between 0.27 and 0.51 mm per year (mean 0.41 mm, varying according to the underlying diameter) and therefore fivefold less than other aortic regions. Accordingly, a root aneurysm indicating reoperation would not be expected for 29.1 years on average. Only patients with a diameter of 45 mm or more are at risk for reoperation, and not until at least after 10.4 years have passed. Neither the valve morphology (p = 0.62) nor concomitant aortic valve replacement (p = 0.86) influenced rate of root dilation. CONCLUSIONS In nonsyndromic patients, the aortic root is the slowest growing portion of the thoracic aorta. Leaving the native root, as in root-sparing ascending aortic replacement, is a safe approach regarding secondary root intervention for aortic root diameters of 45 mm or less.
Collapse
Affiliation(s)
- Sven Peterss
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut; Department of Cardiac Surgery, University Hospital Munich, Ludwig Maximilian University, Munich, Germany
| | - Rohan Bhandari
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - John A Rizzo
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut; Departments of Economics and Preventive Medicine, Stony Brook University, Stony Brook, New York
| | - Hai Fang
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Gregory A Kuzmik
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut
| | - Bulat A Ziganshin
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut; Department of Surgical Diseases 2, Kazan State Medical University, Kazan, Russia
| | - John A Elefteriades
- Aortic Institute at Yale-New Haven Hospital, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
108
|
Nakajima Y. Mechanism responsible for D-transposition of the great arteries: Is this part of the spectrum of right isomerism? Congenit Anom (Kyoto) 2016; 56:196-202. [PMID: 27329052 DOI: 10.1111/cga.12176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/26/2016] [Accepted: 06/15/2016] [Indexed: 12/25/2022]
Abstract
D-transposition of the great arteries (TGA) is one of the most common conotruncal heart defects at birth and is characterized by a discordant ventriculoarterial connection with a concordant atrioventricular connection. The morphological etiology of TGA is an inverted or arrested rotation of the heart outflow tract (OFT, conotruncus), by which the aorta is transposed in the right ventral direction to the pulmonary trunk. The rotational defect of the OFT is thought to be attributed to hypoplasia of the subpulmonic conus, which originates from the left anterior heart field (AHF) residing in the mesodermal core of the first and second pharyngeal arches. AHF, especially on the left, at the early looped heart stage (corresponding to Carnegie stage 10-11 in the human embryo) is one of the regions responsible for the impediment that causes TGA morphology. In human or experimentally produced right isomerism, malposition of the great arteries including D-TGA is frequently associated. Mutations in genes involving left-right (L-R) asymmetry, such as NODAL, ACTRIIB and downstream target FOXH1, have been found in patients with right isomerism as well as in isolated TGA. The downstream pathways of Nodal-Foxh1 play a critical role not only in L-R determination in the lateral plate mesoderm but also in myocardial specification and differentiation in the AHF, suggesting that TGA is a phenotype in heterotaxia as well as the primary developmental defect of the AHF.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
109
|
Jahangiri L, Sharpe M, Novikov N, González-Rosa JM, Borikova A, Nevis K, Paffett-Lugassy N, Zhao L, Adams M, Guner-Ataman B, Burns CE, Burns CG. The AP-1 transcription factor component Fosl2 potentiates the rate of myocardial differentiation from the zebrafish second heart field. Development 2016; 143:113-22. [PMID: 26732840 DOI: 10.1242/dev.126136] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vertebrate heart forms through successive phases of cardiomyocyte differentiation. Initially, cardiomyocytes derived from first heart field (FHF) progenitors assemble the linear heart tube. Thereafter, second heart field (SHF) progenitors differentiate into cardiomyocytes that are accreted to the poles of the heart tube over a well-defined developmental window. Although heart tube elongation deficiencies lead to life-threatening congenital heart defects, the variables controlling the initiation, rate and duration of myocardial accretion remain obscure. Here, we demonstrate that the AP-1 transcription factor, Fos-like antigen 2 (Fosl2), potentiates the rate of myocardial accretion from the zebrafish SHF. fosl2 mutants initiate accretion appropriately, but cardiomyocyte production is sluggish, resulting in a ventricular deficit coupled with an accumulation of SHF progenitors. Surprisingly, mutant embryos eventually correct the myocardial deficit by extending the accretion window. Overexpression of Fosl2 also compromises production of SHF-derived ventricular cardiomyocytes, a phenotype that is consistent with precocious depletion of the progenitor pool. Our data implicate Fosl2 in promoting the progenitor to cardiomyocyte transition and uncover the existence of regulatory mechanisms to ensure appropriate SHF-mediated cardiomyocyte contribution irrespective of embryonic stage.
Collapse
Affiliation(s)
- Leila Jahangiri
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Michka Sharpe
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Natasha Novikov
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Juan Manuel González-Rosa
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Asya Borikova
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen Nevis
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Noelle Paffett-Lugassy
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Long Zhao
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Meghan Adams
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Burcu Guner-Ataman
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| | - Caroline E Burns
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - C Geoffrey Burns
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Charlestown, MA 02129, USA Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
110
|
Stieger-Vanegas SM, Scollan KF, Meadows L, Sisson D, Schlipf J, Riebold T, Löhr CV. Cardiac-gated computed tomography angiography in three alpacas with complex congenital heart disease. J Vet Cardiol 2016; 18:88-98. [PMID: 26803197 DOI: 10.1016/j.jvc.2015.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 09/03/2015] [Accepted: 09/21/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND The prevalence of congenital heart disease is higher in camelids than in other domestic species and complex defects, often involving the great vessels, are more frequently encountered in llamas and alpacas than in other species. Some of these complex defects can be difficult to accurately characterize via echocardiography, the most commonly used diagnostic imaging technique to evaluate the heart in veterinary patients. Contrast-enhanced, electrocardiogram (ECG)-gated computed tomography (CT) has proven utility for the evaluation of human patients with certain congenital heart defects, including those with conotruncal septation defects and other abnormalities involving the formation of the great vessels. METHODS Three alpaca crias, 4 days, 5 weeks and 14 months of age were clinically evaluated and subjected to a complete color-flow Doppler echocardiogram and a contrast-enhanced ECG-gated CT. RESULTS These alpacas exhibited a variety of clinical findings including lethargy, failure to thrive, exercise intolerance, heart murmur, and/or respiratory difficulty. All three crias were subsequently diagnosed with complex cardiac defects including pulmonary atresia with a ventricular septal defect (VSD), a truncus arteriosus with a large VSD, and a double outlet right ventricle with a large VSD and aortic hypoplasia. In each case, the diagnosis was confirmed by postmortem examination. CONCLUSION Color flow echocardiographic evaluation identified all of the intra-cardiac lesions and associated flow anomalies but contrast-enhanced ECG-gated CT permitted more accurate assessment of the morphology of the extracardiac structures and permitted a more precise determination of the exact nature and anatomy of the great vessels.
Collapse
Affiliation(s)
- S M Stieger-Vanegas
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA.
| | - K F Scollan
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| | - L Meadows
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| | - D Sisson
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| | - J Schlipf
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| | - T Riebold
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| | - C V Löhr
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Magruder Hall, Corvallis, OR 97331, USA
| |
Collapse
|
111
|
Yang L, Geng Z, Nickel T, Johnson C, Gao L, Dutton J, Hou C, Zhang J. Differentiation of Human Induced-Pluripotent Stem Cells into Smooth-Muscle Cells: Two Novel Protocols. PLoS One 2016; 11:e0147155. [PMID: 26771193 PMCID: PMC4714916 DOI: 10.1371/journal.pone.0147155] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/29/2015] [Indexed: 01/18/2023] Open
Abstract
Conventional protocols for differentiating human induced-pluripotent stem cells (hiPSCs) into smooth-muscle cells (SMCs) can be inefficient and generally fail to yield cells with a specific SMC phenotype (i.e., contractile or synthetic SMCs). Here, we present two novel hiPSC-SMC differentiation protocols that yield SMCs with predominantly contractile or synthetic phenotypes. Flow cytometry analyses of smooth-muscle actin (SMA) expression indicated that ~45% of the cells obtained with each protocol assumed an SMC phenotype, and that the populations could be purified to ~95% via metabolic selection. Assessments of cellular mRNA and/or protein levels indicated that SMA, myosin heavy chain II, collagen 1, calponin, transgelin, connexin 43, and vimentin expression in the SMCs obtained via the Contractile SMC protocol and in SMCs differentiated via a traditional protocol were similar, while SMCs produced via the Sythetic SMC protocol expressed less calponin, more collagen 1, and more connexin 43. Differences were also observed in functional assessments of the two SMC populations: the two-dimensional surface area of Contractile SMCs declined more extensively (to 12% versus 44% of original size) in response to carbachol treatment, while quantification of cell migration and proliferation were greater in Synthetic SMCs. Collectively, these data demonstrate that our novel differentiation protocols can efficiently generate SMCs from hiPSCs.
Collapse
Affiliation(s)
- Libang Yang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zhaohui Geng
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Thomas Nickel
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Caitlin Johnson
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Lin Gao
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - James Dutton
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Cody Hou
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Jianyi Zhang
- Division of Cardiology, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
112
|
Ruiz-Villalba A, Hoppler S, van den Hoff MJB. Wnt signaling in the heart fields: Variations on a common theme. Dev Dyn 2016; 245:294-306. [PMID: 26638115 DOI: 10.1002/dvdy.24372] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 12/27/2022] Open
Abstract
Wnt signaling plays an essential role in development and differentiation. Heart development is initiated with the induction of precardiac mesoderm requiring the tightly and spatially controlled regulation of canonical and noncanonical Wnt signaling pathways. The role of Wnt signaling in subsequent development of the heart fields is to a large extent unclear. We will discuss the role of Wnt signaling in the development of the arterial and venous pole of the heart, highlighting the dual roles of Wnt signaling with respect to its time- and dosage-dependent effects and the balance between the canonical and noncanonical signaling. Canonical signaling appears to be involved in retaining the cardiac precursors in a proliferative and precursor state, whereas noncanonical signaling promotes their differentiation. Thereafter, both canonical and noncanonical signaling regulate specific steps in differentiation of the cardiac compartments. Because heart development is a contiguous, rather than a sequential, process, analyses tend only to show a single timeframe of development. The repetitive alternating and reciprocal effect of canonical and noncanonical signaling is lost when studied in homogenates. Without the simultaneous in vivo visualization of the different Wnt signaling pathways, the mechanism of Wnt signaling in heart development remains elusive.
Collapse
Affiliation(s)
- Adrián Ruiz-Villalba
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| | - Stefan Hoppler
- Cardiovascular Biology and Medicine Research Programme, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Maurice J B van den Hoff
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| |
Collapse
|
113
|
Steinbach SK, Husain M. Vascular smooth muscle cell differentiation from human stem/progenitor cells. Methods 2015; 101:85-92. [PMID: 26678794 DOI: 10.1016/j.ymeth.2015.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 01/16/2023] Open
Abstract
Transplantation of vascular smooth muscle cells (VSMCs) is a promising cellular therapy to promote angiogenesis and wound healing. However, VSMCs are derived from diverse embryonic sources which may influence their role in the development of vascular disease and in its therapeutic modulation. Despite progress in understanding the mechanisms of VSMC differentiation, there remains a shortage of robust methods for generating lineage-specific VSMCs from pluripotent and adult stem/progenitor cells in serum-free conditions. Here we describe a method for differentiating pluripotent stem cells, such as embryonic and induced pluripotent stem cells, as well as skin-derived precursors, into lateral plate-derived VSMCs including 'coronary-like' VSMCs and neural crest-derived VSMC, respectively. We believe this approach will have broad applications in modeling origin-specific disease vulnerability and in developing personalized cell-based vascular grafts for regenerative medicine.
Collapse
Affiliation(s)
- Sarah K Steinbach
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada
| | - Mansoor Husain
- McEwen Centre for Regenerative Medicine, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Division of Experimental Therapeutics, Toronto General Research Institute, 101 College St., Toronto, Ontario M5G-1L7, Canada; Departments of Medicine, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Physiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Departments of Laboratory Medicine & Pathobiology, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Ted Rogers Centre for Heart Research, University of Toronto, 1 Kings College Circle, Toronto, Ontario M5S-1A8, Canada; Peter Munk Cardiac Centre, University Health Network, 200 Elizabeth St., Toronto, Ontario M5G-2C4, Canada.
| |
Collapse
|
114
|
Shi N, Chen SY. Smooth Muscle Cell Differentiation: Model Systems, Regulatory Mechanisms, and Vascular Diseases. J Cell Physiol 2015; 231:777-87. [DOI: 10.1002/jcp.25208] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ning Shi
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| | - Shi-You Chen
- Department of Physiology and Pharmacology; University of Georgia; Athens Georgia
| |
Collapse
|
115
|
Kshitiz, Afzal J, Kim DH, Levchenko A. Concise review: Mechanotransduction via p190RhoGAP regulates a switch between cardiomyogenic and endothelial lineages in adult cardiac progenitors. Stem Cells 2015; 32:1999-2007. [PMID: 24710857 DOI: 10.1002/stem.1700] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/18/2014] [Indexed: 01/01/2023]
Abstract
Mechanical cues can have pleiotropic influence on stem cell shape, proliferation, differentiation, and morphogenesis, and are increasingly realized to play an instructive role in regeneration and maintenance of tissue structure and functions. To explore the putative effects of mechanical cues in regeneration of the cardiac tissue, we investigated therapeutically important cardiosphere-derived cells (CDCs), a heterogeneous patient- or animal-specific cell population containing c-Kit(+) multipotent stem cells. We showed that mechanical cues can instruct c-Kit(+) cell differentiation along two lineages with corresponding morphogenic changes, while also serving to amplify the initial c-Kit(+) subpopulation. In particular, mechanical cues mimicking the structure of myocardial extracellular matrix specify cardiomyogenic fate, while cues mimicking myocardium rigidity specify endothelial fates. Furthermore, we found that these cues dynamically regulate the same molecular species, p190RhoGAP, which then acts through both RhoA-dependent and independent mechanisms. Thus, differential regulation of p190RhoGAP molecule by either mechanical inputs or genetic manipulation can determine lineage type specification. Since human CDCs are already in phase II clinical trials, the potential therapeutic use of mechanical or genetic manipulation of the cell fate could enhance effectiveness of these progenitor cells in cardiac repair, and shed new light on differentiation mechanisms in cardiac and other tissues.
Collapse
Affiliation(s)
- Kshitiz
- Department of Bioengineering, Institute of Stem Cells and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA; Institute of Stem Cells and Regenerative Medicine and Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| | | | | | | |
Collapse
|
116
|
Martin PS, Kloesel B, Norris RA, Lindsay M, Milan D, Body SC. Embryonic Development of the Bicuspid Aortic Valve. J Cardiovasc Dev Dis 2015; 2:248-272. [PMID: 28529942 PMCID: PMC5438177 DOI: 10.3390/jcdd2040248] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital valvular heart defect with an overall frequency of 0.5%–1.2%. BAVs result from abnormal aortic cusp formation during valvulogenesis, whereby adjacent cusps fuse into a single large cusp resulting in two, instead of the normal three, aortic cusps. Individuals with BAV are at increased risk for ascending aortic disease, aortic stenosis and coarctation of the aorta. The frequent occurrence of BAV and its anatomically discrete but frequent co-existing diseases leads us to suspect a common cellular origin. Although autosomal-dominant transmission of BAV has been observed in a few pedigrees, notably involving the gene NOTCH1, no single-gene model clearly explains BAV inheritance, implying a complex genetic model involving interacting genes. Several sequencing studies in patients with BAV have identified rare and uncommon mutations in genes of cardiac embryogenesis. But the extensive cell-cell signaling and multiple cellular origins involved in cardiac embryogenesis preclude simplistic explanations of this disease. In this review, we examine the series of events from cellular and transcriptional embryogenesis of the heart, to development of the aortic valve.
Collapse
Affiliation(s)
- Peter S. Martin
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Benjamin Kloesel
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell Biology, Children’s Research Institute, Medical University of South Carolina, 173 Ashley St, Charleston, SC 29403, USA; E-Mail:
| | - Mark Lindsay
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - David Milan
- Cardiovascular Research Center, Richard B. Simches Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; E-Mails: (M.L.); (D.M.)
| | - Simon C. Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St., Th724, Boston, MA 02115, USA; E-Mails: (P.S.M.); (B.K.)
- Author to whom correspondence should be addressed: E-Mail: ; Tel.: +1-617-732-7330; Fax: +1-617-730-2813
| |
Collapse
|
117
|
Roux M, Laforest B, Capecchi M, Bertrand N, Zaffran S. Hoxb1 regulates proliferation and differentiation of second heart field progenitors in pharyngeal mesoderm and genetically interacts with Hoxa1 during cardiac outflow tract development. Dev Biol 2015; 406:247-58. [PMID: 26284287 DOI: 10.1016/j.ydbio.2015.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 08/06/2015] [Accepted: 08/14/2015] [Indexed: 01/28/2023]
Abstract
Outflow tract (OFT) anomalies are among the most common congenital heart defects found at birth. The embryonic OFT grows by the progressive addition of cardiac progenitors, termed the second heart field (SHF), which originate from splanchnic pharyngeal mesoderm. Development of the SHF is controlled by multiple intercellular signals and transcription factors; however the relationship between different SHF regulators remains unclear. We have recently shown that Hoxa1 and Hoxb1 are expressed in a sub-population of the SHF contributing to the OFT. Here, we report that Hoxb1 deficiency results in a shorter OFT and ventricular septal defects (VSD). Mechanistically, we show that both FGF/ERK and BMP/SMAD signaling, which regulate proliferation and differentiation of cardiac progenitor cells and OFT morphogenesis, are enhanced in the pharyngeal region in Hoxb1 mutants. Absence of Hoxb1 also perturbed SHF development through premature myocardial differentiation. Hence, the positioning and remodeling of the mutant OFT is disrupted. Hoxa1(-/-) embryos, in contrast, have low percentage of VSD and normal SHF development. However, compound Hoxa1(-/-); Hoxb1(+/-) embryos display OFT defects associated with premature SHF differentiation, demonstrating redundant roles of these factors during OFT development. Our findings provide new insights into the gene regulatory network controlling SHF and OFT formation.
Collapse
Affiliation(s)
- Marine Roux
- Aix Marseille Université, GMGF, 13385 Marseille, France; Inserm, UMR_S910, 13385 Marseille, France
| | - Brigitte Laforest
- Aix Marseille Université, GMGF, 13385 Marseille, France; Inserm, UMR_S910, 13385 Marseille, France
| | - Mario Capecchi
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Bertrand
- Aix Marseille Université, GMGF, 13385 Marseille, France; Inserm, UMR_S910, 13385 Marseille, France
| | - Stéphane Zaffran
- Aix Marseille Université, GMGF, 13385 Marseille, France; Inserm, UMR_S910, 13385 Marseille, France.
| |
Collapse
|
118
|
Lindsey SE, Menon PG, Kowalski WJ, Shekhar A, Yalcin HC, Nishimura N, Schaffer CB, Butcher JT, Pekkan K. Growth and hemodynamics after early embryonic aortic arch occlusion. Biomech Model Mechanobiol 2015; 14:735-51. [PMID: 25416845 PMCID: PMC4452432 DOI: 10.1007/s10237-014-0633-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 11/05/2014] [Indexed: 01/28/2023]
Abstract
The majority of severe clinically significant forms of congenital heart disease (CHD) are associated with great artery lesions, including hypoplastic, double, right or interrupted aortic arch morphologies. While fetal and neonatal interventions are advancing, their potential ability to restore cardiac function, optimal timing, location, and intensity required for intervention remain largely unknown. Here, we combine computational fluid dynamics (CFD) simulations with in vivo experiments to test how individual pharyngeal arch artery hemodynamics alter as a result of local interventions obstructing individual arch artery flow. Simulated isolated occlusions within each pharyngeal arch artery were created with image-derived three-dimensional (3D) reconstructions of normal chick pharyngeal arch anatomy at Hamburger-Hamilton (HH) developmental stages HH18 and HH24. Acute flow redistributions were then computed using in vivo measured subject-specific aortic sinus inflow velocity profiles. A kinematic vascular growth-rendering algorithm was then developed and implemented to test the role of changing local wall shear stress patterns in downstream 3D morphogenesis of arch arteries. CFD simulations predicted that altered pressure gradients and flow redistributions were most sensitive to occlusion of the IVth arches. To evaluate these simulations experimentally, a novel in vivo experimental model of pharyngeal arch occlusion was developed and implemented using two-photon microscopy-guided femtosecond laser-based photodisruption surgery. The right IVth arch was occluded at HH18, and resulting diameter changes were followed for up to 24 h. Pharyngeal arch diameter responses to acute hemodynamic changes were predicted qualitatively but poorly quantitatively. Chronic growth and adaptation to hemodynamic changes, however, were predicted in a subset of arches. Our findings suggest that this complex biodynamic process is governed through more complex forms of mechanobiological vascular growth rules. Other factors in addition to wall shear stress or more complex WSS rules are likely important in the long-term arterial growth and patterning. Combination in silico/experimental platforms are essential for accelerating our understanding and prediction of consequences from embryonic/fetal cardiovascular occlusions and lay the foundation for noninvasive methods to guide CHD diagnosis and fetal intervention.
Collapse
Affiliation(s)
| | - Prahlad G. Menon
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | - William J. Kowalski
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| | - Akshay Shekhar
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Huseyin C. Yalcin
- Department of Mechanical Engineering, Dogus University, Istanbul, Turkey
| | - Nozomi Nishimura
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Chris B. Schaffer
- Department of Biomedical Engineering, Cornell University, Ithaca, NY
| | | | - Kerem Pekkan
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA
| |
Collapse
|
119
|
Parikh A, Wu J, Blanton RM, Tzanakakis ES. Signaling Pathways and Gene Regulatory Networks in Cardiomyocyte Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:377-92. [PMID: 25813860 DOI: 10.1089/ten.teb.2014.0662] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Strategies for harnessing stem cells as a source to treat cell loss in heart disease are the subject of intense research. Human pluripotent stem cells (hPSCs) can be expanded extensively in vitro and therefore can potentially provide sufficient quantities of patient-specific differentiated cardiomyocytes. Although multiple stimuli direct heart development, the differentiation process is driven in large part by signaling activity. The engineering of hPSCs to heart cell progeny has extensively relied on establishing proper combinations of soluble signals, which target genetic programs thereby inducing cardiomyocyte specification. Pertinent differentiation strategies have relied as a template on the development of embryonic heart in multiple model organisms. Here, information on the regulation of cardiomyocyte development from in vivo genetic and embryological studies is critically reviewed. A fresh interpretation is provided of in vivo and in vitro data on signaling pathways and gene regulatory networks (GRNs) underlying cardiopoiesis. The state-of-the-art understanding of signaling pathways and GRNs presented here can inform the design and optimization of methods for the engineering of tissues for heart therapies.
Collapse
Affiliation(s)
- Abhirath Parikh
- 1 Lonza Walkersville, Inc. , Lonza Group, Walkersville, Maryland
| | - Jincheng Wu
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts
| | - Robert M Blanton
- 3 Division of Cardiology, Molecular Cardiology Research Institute , Tufts Medical Center, Tufts School of Medicine, Boston, Massachusetts
| | - Emmanuel S Tzanakakis
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts.,4 Tufts Clinical and Translational Science Institute (CTSI) , Boston, Massachusetts
| |
Collapse
|
120
|
Imanaka-Yoshida K, Yoshida T, Miyagawa-Tomita S. Tenascin-C in development and disease of blood vessels. Anat Rec (Hoboken) 2015; 297:1747-57. [PMID: 25125186 DOI: 10.1002/ar.22985] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 01/25/2014] [Accepted: 01/25/2014] [Indexed: 12/30/2022]
Abstract
Tenascin-C (TNC) is an extracellular glycoprotein categorized as a matricellular protein. It is highly expressed during embryonic development, wound healing, inflammation, and cancer invasion, and has a wide range of effects on cell response in tissue morphogenesis and remodeling including the cardiovascular system. In the heart, TNC is sparsely detected in normal adults but transiently expressed at restricted sites during embryonic development and in response to injury, playing an important role in myocardial remodeling. Although TNC in the vascular system appears more complex than in the heart, the expression of TNC in normal adult blood vessels is generally low. During embryonic development, vascular smooth muscle cells highly express TNC on maturation of the vascular wall, which is controlled in a way that depends on the embryonic site of cell origin. Strong expression of TNC is also linked with several pathological conditions such as cerebral vasospasm, intimal hyperplasia, pulmonary artery hypertension, and aortic aneurysm/ dissection. TNC synthesized by smooth muscle cells in response to developmental and environmental cues regulates cell responses such as proliferation, migration, differentiation, and survival in an autocrine/paracrine fashion and in a context-dependent manner. Thus, TNC can be a key molecule in controlling cellular activity in adaptation during normal vascular development as well as tissue remodeling in pathological conditions.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie, 514-8507, Japan; Mie University Research Center for Matrix Biology, Tsu, Mie, 514-8507, Japan
| | | | | |
Collapse
|
121
|
Turner CJ, Badu-Nkansah K, Crowley D, van der Flier A, Hynes RO. α5 and αv integrins cooperate to regulate vascular smooth muscle and neural crest functions in vivo. Development 2015; 142:797-808. [PMID: 25670798 DOI: 10.1242/dev.117572] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The RGD-binding α5 and αv integrins have been shown to be key regulators of vascular smooth muscle cell (vSMC) function in vitro. However, their role on vSMCs during vascular development in vivo remains unclear. To address this issue, we have generated mice that lack α5, αv or both α5 and αv integrins on their vSMCs, using the SM22α-Cre transgenic mouse line. To our surprise, neither α5 nor αv mutants displayed any obvious vascular defects during embryonic development. By contrast, mice lacking both α5 and αv integrins developed interrupted aortic arches, large brachiocephalic/carotid artery aneurysms and cardiac septation defects, but developed extensive and apparently normal vasculature in the skin. Cardiovascular defects were also found, along with cleft palates and ectopically located thymi, in Wnt1-Cre α5/αv mutants, suggesting that α5 and αv cooperate on neural crest-derived cells to control the remodelling of the pharyngeal arches and the septation of the heart and outflow tract. Analysis of cultured α5/αv-deficient vSMCs suggests that this is achieved, at least in part, through proper assembly of RGD-containing extracellular matrix proteins and the correct incorporation and activation of latent TGF-β.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Denise Crowley
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
122
|
Wu M, Li J. Numb family proteins: novel players in cardiac morphogenesis and cardiac progenitor cell differentiation. Biomol Concepts 2015; 6:137-48. [PMID: 25883210 PMCID: PMC4589147 DOI: 10.1515/bmc-2015-0003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/16/2015] [Indexed: 11/15/2022] Open
Abstract
Vertebrate heart formation is a spatiotemporally regulated morphogenic process that initiates with bilaterally symmetric cardiac primordial cells migrating toward the midline to form a linear heart tube. The heart tube then elongates and undergoes a series of looping morphogenesis, followed by expansions of regions that are destined to become primitive heart chambers. During the cardiac morphogenesis, cells derived from the first heart field contribute to the primary heart tube, and cells from the secondary heart field, cardiac neural crest, and pro-epicardial organ are added to the heart tube in a precise spatiotemporal manner. The coordinated addition of these cells and the accompanying endocardial cushion morphogenesis yield the atrial, ventricular, and valvular septa, resulting in the formation of a four-chambered heart. Perturbation of progenitor cells' deployment and differentiation leads to a spectrum of congenital heart diseases. Two of the genes that were recently discovered to be involved in cardiac morphogenesis are Numb and Numblike. Numb, an intracellular adaptor protein, distinguishes sibling cell fates by its asymmetric distribution between the two daughter cells and its ability to inhibit Notch signaling. Numb regulates cardiac progenitor cell differentiation in Drosophila and controls heart tube laterality in Zebrafish. In mice, Numb and Numblike, the Numb family proteins (NFPs), function redundantly and have been shown to be essential for epicardial development, cardiac progenitor cell differentiation, outflow tract alignment, atrioventricular septum morphogenesis, myocardial trabeculation, and compaction. In this review, we will summarize the functions of NFPs in cardiac development and discuss potential mechanisms of NFPs in the regulation of cardiac development.
Collapse
Affiliation(s)
- M Wu
- Cardiovascular Science Center, Albany Medical College, Albany NY 12208
| | - J Li
- Cardiovascular Science Center, Albany Medical College, Albany NY 12208
| |
Collapse
|
123
|
Abstract
The heart is the first organ to form during embryonic development. Given the complex nature of cardiac differentiation and morphogenesis, it is not surprising that some form of congenital heart disease is present in ≈1 percent of newborns. The molecular determinants of heart development have received much attention over the past several decades. This has been driven in large part by an interest in understanding the causes of congenital heart disease coupled with the potential of using knowledge from developmental biology to generate functional cells and tissues that could be used for regenerative medicine purposes. In this review, we highlight the critical signaling pathways and transcription factor networks that regulate cardiomyocyte lineage specification in both in vivo and in vitro models. Special focus will be given to epigenetic regulators that drive the commitment of cardiomyogenic cells from nascent mesoderm and their differentiation into chamber-specific myocytes, as well as regulation of myocardial trabeculation.
Collapse
Affiliation(s)
- Sharon L Paige
- From the Division of Pediatric Cardiology and Department of Pediatrics (S.L.P., S.M.W.), Cardiovascular Institute (K.P., A.X., S.M.W.), Division of Cardiovascular Medicine, Department of Medicine, Institute for Stem Cell Biology and Institute for Stem Cell Biology and Regenerative Medicine Regenerative Medicine, Child Health Research Institute (S.M.W.), Stanford University School of Medicine, CA
| | - Karolina Plonowska
- From the Division of Pediatric Cardiology and Department of Pediatrics (S.L.P., S.M.W.), Cardiovascular Institute (K.P., A.X., S.M.W.), Division of Cardiovascular Medicine, Department of Medicine, Institute for Stem Cell Biology and Institute for Stem Cell Biology and Regenerative Medicine Regenerative Medicine, Child Health Research Institute (S.M.W.), Stanford University School of Medicine, CA
| | - Adele Xu
- From the Division of Pediatric Cardiology and Department of Pediatrics (S.L.P., S.M.W.), Cardiovascular Institute (K.P., A.X., S.M.W.), Division of Cardiovascular Medicine, Department of Medicine, Institute for Stem Cell Biology and Institute for Stem Cell Biology and Regenerative Medicine Regenerative Medicine, Child Health Research Institute (S.M.W.), Stanford University School of Medicine, CA
| | - Sean M Wu
- From the Division of Pediatric Cardiology and Department of Pediatrics (S.L.P., S.M.W.), Cardiovascular Institute (K.P., A.X., S.M.W.), Division of Cardiovascular Medicine, Department of Medicine, Institute for Stem Cell Biology and Institute for Stem Cell Biology and Regenerative Medicine Regenerative Medicine, Child Health Research Institute (S.M.W.), Stanford University School of Medicine, CA.
| |
Collapse
|
124
|
Induced pluripotent stem cell-derived vascular smooth muscle cells: methods and application. Biochem J 2015; 465:185-94. [PMID: 25559088 DOI: 10.1042/bj20141078] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Vascular smooth muscle cells (VSMCs) play a major role in the pathophysiology of cardiovascular diseases. The advent of induced pluripotent stem cell (iPSC) technology and the capability of differentiating into virtually every cell type in the human body make this field a ray of hope for vascular regenerative therapy and understanding of the disease mechanism. In the present review, we first discuss the recent iPSC technology and vascular smooth muscle development from an embryo and then examine different methodologies to derive VSMCs from iPSCs, and their applications in regenerative therapy and disease modelling.
Collapse
|
125
|
Mostefa-Kara M, Bonnet D, Belli E, Fadel E, Houyel L. Anatomy of the ventricular septal defect in outflow tract defects: Similarities and differences. J Thorac Cardiovasc Surg 2015; 149:682-8.e1. [DOI: 10.1016/j.jtcvs.2014.11.087] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 11/17/2014] [Accepted: 11/29/2014] [Indexed: 02/03/2023]
|
126
|
Granata A, Bernard WG, Zhao N, Mccafferty J, Lilly B, Sinha S. Temporal and embryonic lineage-dependent regulation of human vascular SMC development by NOTCH3. Stem Cells Dev 2015; 24:846-56. [PMID: 25539150 PMCID: PMC4367523 DOI: 10.1089/scd.2014.0520] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vascular smooth muscle cells (SMCs), which arise from multiple embryonic progenitors, have unique lineage-specific properties and this diversity may contribute to spatial patterns of vascular diseases. We developed in vitro methods to generate distinct vascular SMC subtypes from human pluripotent stem cells, allowing us to explore their intrinsic differences and the mechanisms involved in SMC development. Since Notch signaling is thought to be one of the several key regulators of SMC differentiation and function, we profiled the expression of Notch receptors, ligands, and downstream elements during the development of origin-specific SMC subtypes. NOTCH3 expression in our in vitro model varied in a lineage- and developmental stage-specific manner so that the highest expression in mature SMCs was in those derived from paraxial mesoderm (PM). This pattern was consistent with the high expression level of NOTCH3 observed in the 8-9 week human fetal descending aorta, which is populated by SMCs of PM origin. Silencing NOTCH3 in mature SMCs in vitro reduced SMC markers in cells of PM origin preferentially. Conversely, during early development, NOTCH3 was highly expressed in vitro in SMCs of neuroectoderm (NE) origin. Inhibition of NOTCH3 in early development resulted in a significant downregulation of specific SMC markers exclusively in the NE lineage. Corresponding to this prediction, the Notch3-null mouse showed reduced expression of Acta2 in the neural crest-derived SMCs of the aortic arch. Thus, Notch3 signaling emerges as one of the key regulators of vascular SMC differentiation and maturation in vitro and in vivo in a lineage- and temporal-dependent manner.
Collapse
Affiliation(s)
- Alessandra Granata
- 1 Anne Mclaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, University of Cambridge , Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
127
|
|
128
|
Uribe V, Badía-Careaga C, Casanova JC, Domínguez JN, de la Pompa JL, Sanz-Ezquerro JJ. Arid3b is essential for second heart field cell deployment and heart patterning. Development 2014; 141:4168-81. [PMID: 25336743 DOI: 10.1242/dev.109918] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Arid3b, a member of the conserved ARID family of transcription factors, is essential for mouse embryonic development but its precise roles are poorly understood. Here, we show that Arid3b is expressed in the myocardium of the tubular heart and in second heart field progenitors. Arid3b-deficient embryos show cardiac abnormalities, including a notable shortening of the poles, absence of myocardial differentiation and altered patterning of the atrioventricular canal, which also lacks epithelial-to-mesenchymal transition. Proliferation and death of progenitors as well as early patterning of the heart appear normal. However, DiI labelling of second heart field progenitors revealed a defect in the addition of cells to the heart. RNA microarray analysis uncovered a set of differentially expressed genes in Arid3b-deficient tissues, including Bhlhb2, a regulator of cardiomyocyte differentiation, and Lims2, a gene involved in cell migration. Arid3b is thus required for heart development by regulating the motility and differentiation of heart progenitors. These findings identify Arid3b as a candidate gene involved in the aetiology of human congenital malformations.
Collapse
Affiliation(s)
- Verónica Uribe
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Claudio Badía-Careaga
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Jesús C Casanova
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Jorge N Domínguez
- Departamento de Biología Experimental, Facultad de Ciencias Experimentales, Universidad de Jaén, CU Las Lagunillas, Jáen 23071, Spain
| | - José Luis de la Pompa
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Juan José Sanz-Ezquerro
- Departamento de Desarrollo y Reparación Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain Departamento de Biología Molecular y Celular, Centro Nacional de Biotecnología (CSIC), Darwin, 3, Madrid 28049, Spain
| |
Collapse
|
129
|
Gittenberger-de Groot AC, Calkoen EE, Poelmann RE, Bartelings MM, Jongbloed MRM. Morphogenesis and molecular considerations on congenital cardiac septal defects. Ann Med 2014; 46:640-52. [PMID: 25307363 DOI: 10.3109/07853890.2014.959557] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The primary unseptated heart tube undergoes extensive remodeling including septation at the atrial, atrioventricular, ventricular, and ventriculo-arterial level. Alignment and fusion of the septal components is required to ensure full septation of the heart. Deficiencies lead to septal defects at various levels. Addition of myocardium and mesenchymal tissues from the second heart field (SHF) to the primary heart tube, as well as a population of neural crest cells, provides the necessary cellular players. Surprisingly, the study of the molecular background of these defects does not show a great diversity of responsible transcription factors and downstream gene pathways. Epigenetic modulation and mutations high up in several transcription factor pathways (e.g. NODAL and GATA4) may lead to defects at all levels. Disturbance of modulating pathways, involving primarily the SHF-derived cell populations and the genes expressed therein, results at the arterial pole (e.g. TBX1) in a spectrum of ventricular septal defects located at the level of the outflow tract. At the venous pole (e.g. TBX5), it can explain a variety of atrial septal defects. The various defects can occur as isolated anomalies or within families. In this review developmental, morphological, genetic, as well as epigenetic aspects of septal defects are discussed.
Collapse
|
130
|
Sinha T, Li D, Théveniau-Ruissy M, Hutson MR, Kelly RG, Wang J. Loss of Wnt5a disrupts second heart field cell deployment and may contribute to OFT malformations in DiGeorge syndrome. Hum Mol Genet 2014; 24:1704-16. [PMID: 25410658 DOI: 10.1093/hmg/ddu584] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Outflow tract (OFT) malformation accounts for ∼30% of human congenital heart defects and manifests frequently in TBX1 haplo-insufficiency associated DiGeorge (22q11.2 deletion) syndrome. OFT myocardium originates from second heart field (SHF) progenitors in the pharyngeal and splanchnic mesoderm (SpM), but how these progenitors are deployed to the OFT is unclear. We find that SHF progenitors in the SpM gradually gain epithelial character and are deployed to the OFT as a cohesive sheet. Wnt5a, a non-canonical Wnt, is expressed specifically in the caudal SpM and may regulate oriented cell intercalation to incorporate SHF progenitors into an epithelial-like sheet, thereby generating the pushing force to deploy SHF cells rostrally into the OFT. Using enhancer trap and Cre transgenes, our lineage tracing experiments show that in Wnt5a null mice, SHF progenitors are trapped in the SpM and fail to be deployed to the OFT efficiently, resulting in a reduction in the inferior OFT myocardial wall and its derivative, subpulmonary myocardium. Concomitantly, the superior OFT and subaortic myocardium are expanded. Finally, in chick embryos, blocking the Wnt5a function in the caudal SpM perturbs polarized elongation of SHF progenitors, and compromises their deployment to the OFT. Collectively, our results highlight a critical role for Wnt5a in deploying SHF progenitors from the SpM to the OFT. Given that Wnt5a is a putative transcriptional target of Tbx1, and the similar reduction of subpulmonary myocardium in Tbx1 mutant mice, our results suggest that perturbing Wnt5a-mediated SHF deployment may be an important pathogenic mechanism contributing to OFT malformations in DiGeorge syndrome.
Collapse
Affiliation(s)
- Tanvi Sinha
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | - Ding Li
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA
| | | | - Mary R Hutson
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina
| | - Robert G Kelly
- Aix Marseille Université, CNRS, IBDM UMR 7288, Marseille 13288, France
| | - Jianbo Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Alabama, USA,
| |
Collapse
|
131
|
Meilhac SM, Lescroart F, Blanpain C, Buckingham ME. Cardiac cell lineages that form the heart. Cold Spring Harb Perspect Med 2014; 4:a013888. [PMID: 25183852 DOI: 10.1101/cshperspect.a013888] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Myocardial cells ensure the contractility of the heart, which also depends on other mesodermal cell types for its function. Embryological experiments had identified the sources of cardiac precursor cells. With the advent of genetic engineering, novel tools have been used to reconstruct the lineage tree of cardiac cells that contribute to different parts of the heart, map the development of cardiac regions, and characterize their genetic signature. Such knowledge is of fundamental importance for our understanding of cardiogenesis and also for the diagnosis and treatment of heart malformations.
Collapse
Affiliation(s)
- Sigolène M Meilhac
- Institut Pasteur, Department of Developmental and Stem Cell Biology, CNRS URA2578, 75015 Paris, France
| | | | - Cédric Blanpain
- Université Libre de Bruxelles, IRIBHM, Brussels B-1070, Belgium WELBIO, Université Libre de Bruxelles, Brussels B-1070, Belgium
| | - Margaret E Buckingham
- Institut Pasteur, Department of Developmental and Stem Cell Biology, CNRS URA2578, 75015 Paris, France
| |
Collapse
|
132
|
Krishnamurthy VK, Evans AN, Wansapura JP, Osinska H, Maddy KE, Biechler SV, Narmoneva DA, Goodwin RL, Hinton RB. Asymmetric cell-matrix and biomechanical abnormalities in elastin insufficiency induced aortopathy. Ann Biomed Eng 2014; 42:2014-28. [PMID: 25099772 DOI: 10.1007/s10439-014-1072-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/14/2014] [Indexed: 01/28/2023]
Abstract
Aortopathy is characterized by vascular smooth muscle cell (VSMC) abnormalities and elastic fiber fragmentation. Elastin insufficient (Eln (+/-)) mice demonstrate latent aortopathy similar to human disease. We hypothesized that aortopathy manifests primarily in the aorto-pulmonary septal (APS) side of the thoracic aorta due to asymmetric cardiac neural crest (CNC) distribution. Anatomic (aortic root vs. ascending aorta) and molecular (APS vs. non-APS) regions of proximal aorta tissue were examined in adult and aged wild type (WT) and mutant (Eln (+/-)) mice. CNC, VSMCs, elastic fiber architecture, proteoglycan expression, morphometrics and biomechanical properties were examined using histology, 3D reconstruction, micropipette aspiration and in vivo magnetic resonance imaging (MRI). In the APS side of Eln (+/-) aorta, Sonic Hedgehog (SHH) is decreased while SM22 is increased. Elastic fiber architecture abnormalities are present in the Eln (+/-) aortic root and APS ascending aorta, and biglycan is increased in the aortic root while aggrecan is increased in the APS aorta. The Eln (+/-) ascending aorta is stiffer than the aortic root, the APS side is thicker and stiffer than the non-APS side, and significant differences in the individual aortic root sinuses are observed. Asymmetric structure-function abnormalities implicate regional CNC dysregulation in the development and progression of aortopathy.
Collapse
Affiliation(s)
- Varun K Krishnamurthy
- Division of Cardiology, the Heart Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC 7020, Cincinnati, OH, 45229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Liu CF, Cheung JW, Thomas G, Ip JE, Markowitz SM, Lerman BB. Ubiquitous Myocardial Extensions Into the Pulmonary Artery Demonstrated by Integrated Intracardiac Echocardiography and Electroanatomic Mapping. Circ Arrhythm Electrophysiol 2014; 7:691-700. [DOI: 10.1161/circep.113.001347] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Christopher F. Liu
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| | - Jim W. Cheung
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| | - George Thomas
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| | - James E. Ip
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| | - Steven M. Markowitz
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| | - Bruce B. Lerman
- From the Division of Cardiology, Weill Cornell Medical College, New York, NY
| |
Collapse
|
134
|
White PS, Xie HM, Werner P, Glessner J, Latney B, Hakonarson H, Goldmuntz E. Analysis of chromosomal structural variation in patients with congenital left-sided cardiac lesions. ACTA ACUST UNITED AC 2014; 100:951-64. [DOI: 10.1002/bdra.23279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Peter S. White
- The Center for Biomedical Informatics; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
- Department of Pediatrics; Perelman School of Medicine, University of Pennsylvania; Philadelphia Pennsylvania
| | - Hongbo M. Xie
- The Center for Biomedical Informatics; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| | - Petra Werner
- The Division of Cardiology; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| | - Joseph Glessner
- The Center for Applied Genomics, Department of Pediatrics; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| | - Brande Latney
- The Division of Cardiology; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| | - Hakon Hakonarson
- Department of Pediatrics; Perelman School of Medicine, University of Pennsylvania; Philadelphia Pennsylvania
- The Center for Applied Genomics, Department of Pediatrics; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| | - Elizabeth Goldmuntz
- Department of Pediatrics; Perelman School of Medicine, University of Pennsylvania; Philadelphia Pennsylvania
- The Division of Cardiology; The Children's Hospital of Philadelphia; Philadelphia Pennsylvania
| |
Collapse
|
135
|
|
136
|
Liang S, Li HC, Wang YX, Wu SS, Cai YJ, Cui HL, Yang YP, Ya J. Pulmonary endoderm, second heart field and the morphogenesis of distal outflow tract in mouse embryonic heart. Dev Growth Differ 2014; 56:276-92. [PMID: 24697670 DOI: 10.1111/dgd.12129] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 02/21/2014] [Accepted: 02/27/2014] [Indexed: 01/01/2023]
Abstract
The second heart field (SHF), foregut endoderm and sonic hedgehog (SHH) signaling pathway are all reported to associate with normal morphogenesis and septation of outflow tract (OFT). However, the morphological relationships of the development of foregut endoderm and expression of SHH signaling pathway members with the development of surrounding SHF and OFT are seldom described. In this study, serial sections of mouse embryos from ED9 to ED13 (midgestation) were stained with a series of marker antibodies for specifically highlighting SHF (Isl-1), endoderm (Foxa2), basement membrane (Laminin), myocardium (MHC) and smooth muscle (α-SMA) respectively, or SHH receptors antibodies including patched1 (Ptc1), patched2 (Ptc2) and smoothened, to observe the spatiotemporal relationship between them and their contributions to OFT morphogenesis. Our results demonstrated that the development of an Isl-1 positive field in the splanchnic mesoderm ventral to foregut, a subset of SHF, is closely coupled with pulmonary endoderm or tracheal groove, the Isl-1 positive cells surrounding pulmonary endoderm are distributed in a special cone-shaped pattern and take part in the formation of the lateral walls of the intrapericardial aorta and pulmonary trunk and the transient aortic-pulmonary septum, and Ptc1 and Ptc2 are exclusively expressed in pulmonary endoderm during this Isl-l positive field development, suggesting special roles played in inducing the Isl-l positive field formation by pulmonary endoderm. It is indicated that pulmonary endoderm plays a role in the development and specification of SHF in midgestation, and that pulmonary endoderm-associated Isl-l positive field is involved in patterning the morphogenesis and septation of the intrapericardial arterial trunks.
Collapse
Affiliation(s)
- Shi Liang
- Department of Histology and Embryology, Shanxi Medical University, 56 Xin Jian Nan Road, Taiyuan, 030001, Shanxi, China
| | | | | | | | | | | | | | | |
Collapse
|
137
|
Chester AH, El-Hamamsy I, Butcher JT, Latif N, Bertazzo S, Yacoub MH. The living aortic valve: From molecules to function. Glob Cardiol Sci Pract 2014; 2014:52-77. [PMID: 25054122 PMCID: PMC4104380 DOI: 10.5339/gcsp.2014.11] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 04/28/2014] [Indexed: 12/12/2022] Open
Abstract
The aortic valve lies in a unique hemodynamic environment, one characterized by a range of stresses (shear stress, bending forces, loading forces and strain) that vary in intensity and direction throughout the cardiac cycle. Yet, despite its changing environment, the aortic valve opens and closes over 100,000 times a day and, in the majority of human beings, will function normally over a lifespan of 70–90 years. Until relatively recently heart valves were considered passive structures that play no active role in the functioning of a valve, or in the maintenance of its integrity and durability. However, through clinical experience and basic research the aortic valve can now be characterized as a living, dynamic organ with the capacity to adapt to its complex mechanical and biomechanical environment through active and passive communication between its constituent parts. The clinical relevance of a living valve substitute in patients requiring aortic valve replacement has been confirmed. This highlights the importance of using tissue engineering to develop heart valve substitutes containing living cells which have the ability to assume the complex functioning of the native valve.
Collapse
|
138
|
Sinha S, Iyer D, Granata A. Embryonic origins of human vascular smooth muscle cells: implications for in vitro modeling and clinical application. Cell Mol Life Sci 2014; 71:2271-88. [PMID: 24442477 PMCID: PMC4031394 DOI: 10.1007/s00018-013-1554-3] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 12/03/2013] [Accepted: 12/30/2013] [Indexed: 01/06/2023]
Abstract
Vascular smooth muscle cells (SMCs) arise from multiple origins during development, raising the possibility that differences in embryological origins between SMCs could contribute to site-specific localization of vascular diseases. In this review, we first examine the developmental pathways and embryological origins of vascular SMCs and then discuss in vitro strategies for deriving SMCs from human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We then review in detail the potential for vascular disease modeling using iPSC-derived SMCs and consider the pathological implications of heterogeneous embryonic origins. Finally, we touch upon the role of human ESC-derived SMCs in therapeutic revascularization and the challenges remaining before regenerative medicine using ESC- or iPSC-derived cells comes of age.
Collapse
Affiliation(s)
- Sanjay Sinha
- Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK,
| | | | | |
Collapse
|
139
|
Saremi F, Gera A, Yen Ho S, Hijazi ZM, Sánchez-Quintana D. CT and MR Imaging of the Pulmonary Valve. Radiographics 2014; 34:51-71. [DOI: 10.1148/rg.341135026] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
140
|
Shi N, Chen SY. Mechanisms simultaneously regulate smooth muscle proliferation and differentiation. J Biomed Res 2013; 28:40-6. [PMID: 24474962 PMCID: PMC3904173 DOI: 10.7555/jbr.28.20130130] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 11/13/2013] [Indexed: 01/01/2023] Open
Abstract
Vascular smooth muscle cell (VSMC) differentiation and proliferation are two important physiological processes during vascular development. The phenotypic alteration from differentiated to proliferative VSMC contributes to the development of several major cardiovascular diseases including atherosclerosis, hypertension, restenosis after angioplasty or bypass, diabetic vascular complications, and transplantation arteriopathy. Since the VSMC phenotype in these pathological conditions resembles that of developing VSMC during embryonic development, understanding of the molecular mechanisms that control VSMC differentiation will provide fundamental insights into the pathological processes of these cardiovascular diseases. Although VSMC differentiation is usually accompanied by an irreversible cell cycle exit, VSMC proliferation and differentiation occur concurrently during embryonic development. The molecular mechanisms simultaneously regulating these two processes, however, remain largely unknown. Our recent study demonstrates that cell division cycle 7, a key regulator of cell cycle, promotes both VSMC differentiation and proliferation through different mechanisms during the initial phase of VSMC differentiation. Conversely, Krüppel-like factor 4 appears to be a repressor for both VSMC differentiation and proliferation. This review attempts to highlight the novel role of cell division cycle 7 in TGF-β-induced VSMC differentiation and proliferation. The role of Krüppel-like factor 4 in suppressing these two processes will also be discussed.
Collapse
Affiliation(s)
- Ning Shi
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
141
|
Della Corte A, Bancone C, Buonocore M, Dialetto G, Covino FE, Manduca S, Scognamiglio G, D'Oria V, De Feo M. Pattern of Ascending Aortic Dimensions Predicts the Growth Rate of the Aorta in Patients With Bicuspid Aortic Valve. JACC Cardiovasc Imaging 2013; 6:1301-10. [DOI: 10.1016/j.jcmg.2013.07.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Accepted: 07/18/2013] [Indexed: 11/29/2022]
|
142
|
Abu-Issa R. Heart fields: spatial polarity and temporal dynamics. Anat Rec (Hoboken) 2013; 297:175-82. [PMID: 24443184 DOI: 10.1002/ar.22831] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/10/2013] [Accepted: 10/14/2013] [Indexed: 11/06/2022]
Abstract
In chick and mouse, heart fields undergo dynamic morphological spatiotemporal changes during heart tube formation. Here, the dynamic change in spatial polarity of such fields is discussed and a new perspective on the heart fields is proposed. The heart progenitor cells delaminate through the primitive streak and migrate in a semicircular trajectory craniolaterally forming the bilateral heart fields as part of the splanchnic mesoderm. They switch their polarity from anteroposterior to mediolateral. The anterior intestinal portal posterior descent inverts the newly formed heart field mediolateral polarity into lateromedial by 125° bending. The heart fields revert back to their original anteroposterior polarity and fuse at the midline forming a semi heart tube by completing their half circle movement. Several names and roles were assigned to different portions of the heart fields: posterior versus anterior, first versus second, and primary versus secondary heart field. The posterior and anterior heart fields define basically physical fields that form the inflow-outflow axis of the heart tube. The first and second heart fields are, in contrast, temporal fields of differentiating cardiomyocytes expressing myosin light chain 2a and undifferentiated and proliferating precardiac mesoderm expressing Isl1 gene, respectively. The two markers present a complementary pattern and are expressed transiently in all myocardial lineages. Thus, Isl1 is not restricted to a portion of the heart field or one of the two heart lineages as has been often assumed.
Collapse
Affiliation(s)
- Radwan Abu-Issa
- Department of Natural Sciences, University of Michigan-Dearborn, Michigan
| |
Collapse
|
143
|
Harmon AW, Nakano A. Nkx2-5 lineage tracing visualizes the distribution of second heart field-derived aortic smooth muscle. Genesis 2013; 51:862-9. [PMID: 24133047 DOI: 10.1002/dvg.22721] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 10/10/2013] [Accepted: 10/11/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Andrew W Harmon
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | | |
Collapse
|
144
|
Moretti A, Laugwitz KL, Dorn T, Sinnecker D, Mummery C. Pluripotent stem cell models of human heart disease. Cold Spring Harb Perspect Med 2013; 3:3/11/a014027. [PMID: 24186488 DOI: 10.1101/cshperspect.a014027] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Understanding the molecular basis of many cardiac diseases has been hampered by the lack of appropriate in vitro cell culture models that accurately reflect the human disease phenotypes. In the past few years, remarkable advances in stem cell biology have made possible this long-standing ambition-the generation of human and even patient-specific cellular models of diseases. Combined with other novel technologies in the fields of human genetics, tissue engineering, and gene-targeted manipulation, disease modeling with pluripotent stem cells has the promise to influence modern cardiovascular medicine on several fronts: molecular understanding of pathological mechanisms, early diagnosis, drug development, and effective treatment.
Collapse
Affiliation(s)
- Alessandra Moretti
- Klinikum rechts der Isar-Technische Universität München, I. Medical Department-Cardiology, 81675 Munich, Germany
| | | | | | | | | |
Collapse
|
145
|
Yu Z, Kong J, Pan B, Sun H, Lv T, Zhu J, Huang G, Tian J. Islet-1 may function as an assistant factor for histone acetylation and regulation of cardiac development-related transcription factor Mef2c expression. PLoS One 2013; 8:e77690. [PMID: 24147056 PMCID: PMC3798409 DOI: 10.1371/journal.pone.0077690] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/03/2013] [Indexed: 01/15/2023] Open
Abstract
Objective Islet-1 is an important transcription factor for cardiac development through mediating extensive interactions between DNA and proteins. The present study was to investigate the role of Islet-1 in regulating the expression of cardiac development-related transcription factors and mechanism. Methods and Results The expression of Islet-1 and histone acetylases (HATs) subtype p300 was determined in newborn mouse hearts and mouse embryonic hearts at different development stages using Western blot. The expression of Islet-1 and cardiac development-related transcription factors Mef2c, GATA4 and Tbx5 as well as histone H3 acetylation level were determined in cardiac progenitor cells with and without transfection of Islet-1 interference RNA (RNAi) in lentivirus using PCR and Western blot. Islet-1 peak expression occurred on day E14.5 in mouse embryonic heart, and was present in the promoter regions of Mef2c, GATA4 and Tbx5 that were precipitated with p300 antibody. When Islet-1 was inhibited with specific RNAi in cardiac progenitor cells, the expression of Mef2c and Tbx5, but not GATA4, was significantly suppressed along with selective reduction in histone H3 acetylation in the promoter region of Mef2c, but not GATA4 and Tbx5. The level of Mef2c DNA, not GATA4 and Tbx5, in the complex associated with p300 was significantly decreased in the cells with Islet-1 knockdown. Conclusions These data suggested that Islet-1 might function as an assistant factor that was involved in the regulation of histone acetylation and Mef2c expression via assisting p300 on specifically targeting the promoter of Mef2c.
Collapse
Affiliation(s)
- Zhongsu Yu
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China
| | - Juanjuan Kong
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China
| | - Bo Pan
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Huichao Sun
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Tiewei Lv
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jing Zhu
- Key Laboratory of Developmental Disease in Childhood (Chongqing Medical University), Ministry of Education, Chongqing, PR China
| | - Guoying Huang
- Pediatric Heart Centre, Children's Hospital of Fudan University, Shanghai, PR China
| | - Jie Tian
- Heart Centre, Children's Hospital of Chongqing Medical University, Chongqing, PR China
- * E-mail:
| |
Collapse
|
146
|
Schleich JM, Abdulla T, Summers R, Houyel L. An overview of cardiac morphogenesis. Arch Cardiovasc Dis 2013; 106:612-23. [PMID: 24138816 DOI: 10.1016/j.acvd.2013.07.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/09/2013] [Accepted: 07/16/2013] [Indexed: 01/06/2023]
Abstract
Accurate knowledge of normal cardiac development is essential for properly understanding the morphogenesis of congenital cardiac malformations that represent the most common congenital anomaly in newborns. The heart is the first organ to function during embryonic development and is fully formed at 8 weeks of gestation. Recent studies stemming from molecular genetics have allowed specification of the role of cellular precursors in the field of heart development. In this article we review the different steps of heart development, focusing on the processes of alignment and septation. We also show, as often as possible, the links between abnormalities of cardiac development and the main congenital heart defects. The development of animal models has permitted the unraveling of many mechanisms that potentially lead to cardiac malformations. A next step towards a better knowledge of cardiac development could be multiscale cardiac modelling.
Collapse
Affiliation(s)
- Jean-Marc Schleich
- Service de Cardiologie et de Maladies Vasculaires, Hôpital de Pontchaillou, 35033 Rennes cedex 09, France; INSERM U 1099, Université de Rennes, LTSI, 35042 Rennes, France.
| | | | | | | |
Collapse
|
147
|
Wu D, Shen YH, Russell L, Coselli JS, LeMaire SA. Molecular mechanisms of thoracic aortic dissection. J Surg Res 2013; 184:907-24. [PMID: 23856125 PMCID: PMC3788606 DOI: 10.1016/j.jss.2013.06.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/31/2013] [Accepted: 06/05/2013] [Indexed: 12/22/2022]
Abstract
Thoracic aortic dissection (TAD) is a highly lethal vascular disease. In many patients with TAD, the aorta progressively dilates and ultimately ruptures. Dissection formation, progression, and rupture cannot be reliably prevented pharmacologically because the molecular mechanisms of aortic wall degeneration are poorly understood. The key histopathologic feature of TAD is medial degeneration, a process characterized by smooth muscle cell depletion and extracellular matrix degradation. These structural changes have a profound impact on the functional properties of the aortic wall and can result from excessive protease-mediated destruction of the extracellular matrix, altered signaling pathways, and altered gene expression. Review of the literature reveals differences in the processes that lead to ascending versus descending and sporadic versus hereditary TAD. These differences add to the complexity of this disease. Although tremendous progress has been made in diagnosing and treating TAD, a better understanding of the molecular, cellular, and genetic mechanisms that cause this disease is necessary to developing more effective preventative and therapeutic treatment strategies.
Collapse
Affiliation(s)
- Darrell Wu
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, BCM 390, One Baylor Plaza, Houston, Texas 77030
- Department of Cardiovascular Surgery, Texas Heart Institute at St. Luke’s Episcopal Hospital, 6770 Bertner Ave., Houston, Texas 77030
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, BCM 335, One Baylor Plaza, Houston, Texas 77030
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, BCM 390, One Baylor Plaza, Houston, Texas 77030
- Department of Cardiovascular Surgery, Texas Heart Institute at St. Luke’s Episcopal Hospital, 6770 Bertner Ave., Houston, Texas 77030
| | - Ludivine Russell
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, BCM 390, One Baylor Plaza, Houston, Texas 77030
- Department of Cardiovascular Surgery, Texas Heart Institute at St. Luke’s Episcopal Hospital, 6770 Bertner Ave., Houston, Texas 77030
| | - Joseph S. Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, BCM 390, One Baylor Plaza, Houston, Texas 77030
- Department of Cardiovascular Surgery, Texas Heart Institute at St. Luke’s Episcopal Hospital, 6770 Bertner Ave., Houston, Texas 77030
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, BCM 390, One Baylor Plaza, Houston, Texas 77030
- Department of Cardiovascular Surgery, Texas Heart Institute at St. Luke’s Episcopal Hospital, 6770 Bertner Ave., Houston, Texas 77030
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, BCM 335, One Baylor Plaza, Houston, Texas 77030
| |
Collapse
|
148
|
Abstract
The mammalian heart is a highly specialized organ, comprised of many different cell types arising from distinct embryonic progenitor populations during cardiogenesis. Three precursor populations have been identified to contribute to different myocytic and nonmyocytic cell lineages of the heart: cardiogenic mesoderm cells (CMC), the proepicardium (PE), and cardiac neural crest cells (CNCCs). This review will focus on molecular cues necessary for proper induction, expansion, and lineage-specific differentiation of these progenitor populations during cardiac development in vivo. Moreover, we will briefly discuss how the knowledge gained on embryonic heart progenitor biology can be used to develop novel therapeutic strategies for the management of congenital heart disease as well as for improvement of cardiac function in ischemic heart disease.
Collapse
|
149
|
Huhta J, Linask KK. Environmental origins of congenital heart disease: the heart-placenta connection. Semin Fetal Neonatal Med 2013; 18:245-50. [PMID: 23751925 DOI: 10.1016/j.siny.2013.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although the mammalian embryo is well protected in the uterus, environmental chemicals, drugs, and maternal nutritional imbalances can interfere with regulatory pathways directing placental and embryonic development early in gestation. Embryonic cells are most susceptible to environmental influences during cellular specification and differentiation stages. Because biochemical differentiation precedes morphological outcome often by days, the period of susceptibility to environmental chemicals expectedly precedes visible morphogenic effects. The cellular mechanisms by which drugs and other environmental factors disrupt embryonic development and induce cardiac abnormalities have remained undefined.
Collapse
Affiliation(s)
- James Huhta
- All Children's Hospital, Johns Hopkins Medicine, St Petersburg, FL, USA.
| | | |
Collapse
|
150
|
Liu D, Wang CJ, Judge DP, Halushka MK, Ni J, Habashi JP, Moslehi J, Bedja D, Gabrielson KL, Xu H, Qian F, Huso D, Dietz HC, Germino GG, Watnick T. A Pkd1-Fbn1 genetic interaction implicates TGF-β signaling in the pathogenesis of vascular complications in autosomal dominant polycystic kidney disease. J Am Soc Nephrol 2013; 25:81-91. [PMID: 24071006 DOI: 10.1681/asn.2012050486] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure that is due to mutations in two genes, PKD1 and PKD2. Vascular complications, including aneurysms, are a well recognized feature of ADPKD, and a subgroup of families exhibits traits reminiscent of Marfan syndrome (MFS). MFS is caused by mutations in fibrillin-1 (FBN1), which encodes an extracellular matrix protein with homology to latent TGF-β binding proteins. It was recently demonstrated that fibrillin-1 deficiency is associated with upregulation of TGF-β signaling. We investigated the overlap between ADPKD and MFS by breeding mice with targeted mutations in Pkd1 and Fbn1. Double heterozygotes displayed an exacerbation of the typical Fbn1 heterozygous aortic phenotype. We show that the basis of this genetic interaction results from further upregulation of TGF-β signaling caused by Pkd1 haploinsufficiency. In addition, we demonstrate that loss of PKD1 alone is sufficient to induce a heightened responsiveness to TGF-β. Our data link the interaction of two important diseases to a fundamental signaling pathway.
Collapse
Affiliation(s)
- Dongyan Liu
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|