1
|
Qu X, Harmelink C, Baldwin HS. Endocardial-Myocardial Interactions During Early Cardiac Differentiation and Trabeculation. Front Cardiovasc Med 2022; 9:857581. [PMID: 35600483 PMCID: PMC9116504 DOI: 10.3389/fcvm.2022.857581] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Throughout the continuum of heart formation, myocardial growth and differentiation occurs in concert with the development of a specialized population of endothelial cells lining the cardiac lumen, the endocardium. Once the endocardial cells are specified, they are in close juxtaposition to the cardiomyocytes, which facilitates communication between the two cell types that has been proven to be critical for both early cardiac development and later myocardial function. Endocardial cues orchestrate cardiomyocyte proliferation, survival, and organization. Additionally, the endocardium enables oxygenated blood to reach the cardiomyocytes. Cardiomyocytes, in turn, secrete factors that promote endocardial growth and function. As misregulation of this delicate and complex endocardial-myocardial interplay can result in congenital heart defects, further delineation of underlying genetic and molecular factors involved in cardiac paracrine signaling will be vital in the development of therapies to promote cardiac homeostasis and regeneration. Herein, we highlight the latest research that has advanced the elucidation of endocardial-myocardial interactions in early cardiac morphogenesis, including endocardial and myocardial crosstalk necessary for cellular differentiation and tissue remodeling during trabeculation, as well as signaling critical for endocardial growth during trabeculation.
Collapse
Affiliation(s)
- Xianghu Qu
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cristina Harmelink
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
| | - H. Scott Baldwin
- Department of Pediatrics (Cardiology), Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Cell and Development Biology, Vanderbilt University, Nashville, TN, United States
- *Correspondence: H. Scott Baldwin
| |
Collapse
|
2
|
Rufaihah AJ, Chen CK, Yap CH, Mattar CNZ. Mending a broken heart: In vitro, in vivo and in silico models of congenital heart disease. Dis Model Mech 2021; 14:dmm047522. [PMID: 33787508 PMCID: PMC8033415 DOI: 10.1242/dmm.047522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Birth defects contribute to ∼0.3% of global infant mortality in the first month of life, and congenital heart disease (CHD) is the most common birth defect among newborns worldwide. Despite the significant impact on human health, most treatments available for this heterogenous group of disorders are palliative at best. For this reason, the complex process of cardiogenesis, governed by multiple interlinked and dose-dependent pathways, is well investigated. Tissue, animal and, more recently, computerized models of the developing heart have facilitated important discoveries that are helping us to understand the genetic, epigenetic and mechanobiological contributors to CHD aetiology. In this Review, we discuss the strengths and limitations of different models of normal and abnormal cardiogenesis, ranging from single-cell systems and 3D cardiac organoids, to small and large animals and organ-level computational models. These investigative tools have revealed a diversity of pathogenic mechanisms that contribute to CHD, including genetic pathways, epigenetic regulators and shear wall stresses, paving the way for new strategies for screening and non-surgical treatment of CHD. As we discuss in this Review, one of the most-valuable advances in recent years has been the creation of highly personalized platforms with which to study individual diseases in clinically relevant settings.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Ching Kit Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Choon Hwai Yap
- Division of Cardiology, Department of Paediatrics, Khoo Teck Puat -National University Children's Medical Institute, National University Health System, Singapore 119228
- Department of Bioengineering, Imperial College London, London, UK
| | - Citra N Z Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore 119228
| |
Collapse
|
3
|
Lahrouchi N, Postma AV, Salazar CM, De Laughter DM, Tjong F, Piherová L, Bowling FZ, Zimmerman D, Lodder EM, Ta-Shma A, Perles Z, Beekman L, Ilgun A, Gunst Q, Hababa M, Škorić-Milosavljević D, Stránecký V, Tomek V, de Knijff P, de Leeuw R, Robinson JY, Burn SC, Mustafa H, Ambrose M, Moss T, Jacober J, Niyazov DM, Wolf B, Kim KH, Cherny S, Rousounides A, Aristidou-Kallika A, Tanteles G, Ange-Line B, Denommé-Pichon AS, Francannet C, Ortiz D, Haak MC, Ten Harkel AD, Manten GT, Dutman AC, Bouman K, Magliozzi M, Radio FC, Santen GW, Herkert JC, Brown HA, Elpeleg O, van den Hoff MJ, Mulder B, Airola MV, Kmoch S, Barnett JV, Clur SA, Frohman MA, Bezzina CR. Biallelic loss-of-function variants in PLD1 cause congenital right-sided cardiac valve defects and neonatal cardiomyopathy. J Clin Invest 2021; 131:142148. [PMID: 33645542 PMCID: PMC7919725 DOI: 10.1172/jci142148] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/16/2020] [Indexed: 01/12/2023] Open
Abstract
Congenital heart disease is the most common type of birth defect, accounting for one-third of all congenital anomalies. Using whole-exome sequencing of 2718 patients with congenital heart disease and a search in GeneMatcher, we identified 30 patients from 21 unrelated families of different ancestries with biallelic phospholipase D1 (PLD1) variants who presented predominantly with congenital cardiac valve defects. We also associated recessive PLD1 variants with isolated neonatal cardiomyopathy. Furthermore, we established that p.I668F is a founder variant among Ashkenazi Jews (allele frequency of ~2%) and describe the phenotypic spectrum of PLD1-associated congenital heart defects. PLD1 missense variants were overrepresented in regions of the protein critical for catalytic activity, and, correspondingly, we observed a strong reduction in enzymatic activity for most of the mutant proteins in an enzymatic assay. Finally, we demonstrate that PLD1 inhibition decreased endothelial-mesenchymal transition, an established pivotal early step in valvulogenesis. In conclusion, our study provides a more detailed understanding of disease mechanisms and phenotypic expression associated with PLD1 loss of function.
Collapse
Affiliation(s)
- Najim Lahrouchi
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Alex V. Postma
- Department of Clinical Genetics, and
- Department of Medical Biology, Amsterdam UMC, Amsterdam, Netherlands
| | - Christian M. Salazar
- Department of Pharmacological Sciences and Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
| | - Daniel M. De Laughter
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Fleur Tjong
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Lenka Piherová
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Forrest Z. Bowling
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Dominic Zimmerman
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Elisabeth M. Lodder
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Asaf Ta-Shma
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Zeev Perles
- Department of Pediatric Cardiology, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | - Leander Beekman
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Aho Ilgun
- Department of Medical Biology, Amsterdam UMC, Amsterdam, Netherlands
| | - Quinn Gunst
- Department of Medical Biology, Amsterdam UMC, Amsterdam, Netherlands
| | - Mariam Hababa
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Doris Škorić-Milosavljević
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Viktor Tomek
- Children’s Heart Centre, 2nd Faculty of Medicine, Charles University in Prague, Motol University Hospital, Prague, Czech Republic
| | - Peter de Knijff
- Department of Human Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Rick de Leeuw
- Department of Human Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Jamille Y. Robinson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | - Hiba Mustafa
- Department of Obstetrics, Gynecology and Women’s Health
| | - Matthew Ambrose
- Department of Pediatrics, Division of Pediatric Cardiology, and
| | - Timothy Moss
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jennifer Jacober
- Department of Pediatrics, Ochsner Clinic, Tulane University, University of Queensland, New Orleans, Louisiana, USA
| | - Dmitriy M. Niyazov
- Department of Pediatrics, Ochsner Clinic, Tulane University, University of Queensland, New Orleans, Louisiana, USA
| | - Barry Wolf
- Division of Genetics, Birth Defects and Metabolic Disorders, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Katherine H. Kim
- Division of Genetics, Birth Defects and Metabolic Disorders, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Sara Cherny
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Division of Cardiology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | | | | | - George Tanteles
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Department of Clinical Genetics, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Bruel Ange-Line
- UMR 1231 INSERM, GAD, Université Bourgogne Franche-Comté, Dijon, France
- Unité Fonctionnelle d’Innovation en Diagnostique Génomique des Maladies Rares, FHU-TRANSLAD, Centre Hospitalier Universitaire Estaing (CHU), Dijon Bourgogne, Dijon, France
| | - Anne-Sophie Denommé-Pichon
- UMR 1231 INSERM, GAD, Université Bourgogne Franche-Comté, Dijon, France
- Unité Fonctionnelle d’Innovation en Diagnostique Génomique des Maladies Rares, FHU-TRANSLAD, Centre Hospitalier Universitaire Estaing (CHU), Dijon Bourgogne, Dijon, France
| | | | - Damara Ortiz
- Medical Genetics Department, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Arend D.J. Ten Harkel
- Department of Pediatric Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | | | - Annemiek C. Dutman
- Department of Pathology, Isala Women and Children’s Hospital, Zwolle, Netherlands
| | - Katelijne Bouman
- University Medical Center Groningen, Department of Genetics, University of Groningen, Groningen, Netherlands
| | - Monia Magliozzi
- Genetic and Rare Disease Research Division, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | - Gijs W.E. Santen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Johanna C. Herkert
- University Medical Center Groningen, Department of Genetics, University of Groningen, Groningen, Netherlands
| | - H. Alex Brown
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Orly Elpeleg
- Department of Genetics, Hadassah, Hebrew University Medical Center, Jerusalem, Israel
| | | | - Barbara Mulder
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| | - Michael V. Airola
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, USA
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Joey V. Barnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Sally-Ann Clur
- Department of Pediatric Cardiology, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Michael A. Frohman
- Department of Pharmacological Sciences and Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, New York, USA
| | - Connie R. Bezzina
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences
| |
Collapse
|
4
|
Urban JD, Wikoff DS, Chappell GA, Harris C, Haws LC. Systematic evaluation of mechanistic data in assessing in utero exposures to trichloroethylene and development of congenital heart defects. Toxicology 2020; 436:152427. [PMID: 32145346 DOI: 10.1016/j.tox.2020.152427] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 01/23/2023]
Abstract
The hypothesis that in utero exposures to low levels of trichloroethylene (TCE) may increase the risk of congenital heart defects (CHDs) in offspring remains a subject of substantial controversy within the scientific community due primarily to the reliance on an inconsistent and unreproducible experimental study in rats. To build on previous assessments that have primarily focused on epidemiological and experimental animal studies in developing conclusions, the objective of the current study is to conduct a systematic evaluation of mechanistic data related to in utero exposures to TCE and the development of CHDs. The evidence base was heterogeneous; 79 mechanistic datasets were identified, characterizing endpoints which ranged from molecular to organismal responses in seven species, involving both in vivo and in vitro study designs in mammalian and non-mammalian models. Of these, 24 datasets were considered reliable following critical appraisal using a study quality tool that employs metrics specific to the study type. Subsequent synthesis and integration demonstrated that the available mechanistic data: 1) did not support the potential for CHD hazard in humans, 2) did not support the biological plausibility of a response in humans based on organization via a putative adverse outcome pathway for valvulo-septal cardiac defects, and 3) were not suitable for serving as candidate studies in risk assessment. Findings supportive of an association were generally limited to in ovo chicken studies, in which TCE was administered in high concentration solutions via direct injection. Results of these in ovo studies were difficult to interpret for human health risk assessment given the lack of generalizability of the study models (including dose relevance, species-specific biological differences, variations in the construct of the study design, etc.). When the mechanistic data are integrated with findings from previous evaluations of human and animal evidence streams, the totality of evidence does not support CHDs as a critical effect in TCE human health risk assessment.
Collapse
Affiliation(s)
- Jonathan D Urban
- ToxStrategies, Inc., 9390 Research Blvd, Ste. 100, Austin, TX, 78759, USA.
| | - Daniele S Wikoff
- ToxStrategies, Inc., 31 College Place, Ste. B118, Asheville, NC, 28801, USA
| | - Grace A Chappell
- ToxStrategies, Inc., 31 College Place, Ste. B118, Asheville, NC, 28801, USA
| | - Craig Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Laurie C Haws
- ToxStrategies, Inc., 9390 Research Blvd, Ste. 100, Austin, TX, 78759, USA
| |
Collapse
|
5
|
Maleki S, Poujade FA, Bergman O, Gådin JR, Simon N, Lång K, Franco-Cereceda A, Body SC, Björck HM, Eriksson P. Endothelial/Epithelial Mesenchymal Transition in Ascending Aortas of Patients With Bicuspid Aortic Valve. Front Cardiovasc Med 2019; 6:182. [PMID: 31921896 PMCID: PMC6928128 DOI: 10.3389/fcvm.2019.00182] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) is the progressive enlargement of the aorta due to destructive changes in the connective tissue of the aortic wall. Aneurysm development is silent and often first manifested by the drastic events of aortic dissection or rupture. As yet, therapeutic agents that halt or reverse the process of aortic wall deterioration are absent, and the only available therapeutic recommendation is elective prophylactic surgical intervention. Being born with a bicuspid instead of the normal tricuspid aortic valve (TAV) is a major risk factor for developing aneurysm in the ascending aorta later in life. Although the pathophysiology of the increased aneurysm susceptibility is not known, recent studies are suggestive of a transformation of aortic endothelium into a more mesenchymal state i.e., an endothelial-to-mesenchymal transition in these individuals. This process involves the loss of endothelial cell features, resulting in junction instability and enhanced vascular permeability of the ascending aorta that may lay the ground for increased aneurysm susceptibility. This finding differentiates and further emphasizes the specific characteristics of aneurysm development in individuals with a bicuspid aortic valve (BAV). This review discusses the possibility of a developmental fate shared between the aortic endothelium and aortic valves. It further speculates about the impact of aortic endothelium phenotypic shift on aneurysm development in individuals with a BAV and revisits previous studies in the light of the new findings.
Collapse
Affiliation(s)
- Shohreh Maleki
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Flore-Anne Poujade
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Otto Bergman
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Jesper R Gådin
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Nancy Simon
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Karin Lång
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Simon C Body
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Hanna M Björck
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Per Eriksson
- Cardiovascular Medicine Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
6
|
Qu X, Violette K, Sewell-Loftin MK, Soslow J, Saint-Jean L, Hinton RB, Merryman WD, Baldwin HS. Loss of flow responsive Tie1 results in Impaired
Aortic valve remodeling. Dev Biol 2019; 455:73-84. [PMID: 31319059 DOI: 10.1016/j.ydbio.2019.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 10/26/2022]
Abstract
The mechanisms regulating endothelial cell response to hemodynamic forces required for heart valve development, especially valve remodeling, remain elusive. Tie1, an endothelial specific receptor tyrosine kinase, is up-regulated by oscillating shear stress and is required for lymphatic valve development. In this study, we demonstrate that valvular endothelial Tie1 is differentially expressed in a dynamic pattern predicted by disturbed flow during valve remodeling. Following valvular endocardial specific deletion of Tie1 in mice, we observed enlarged aortic valve leaflets, decreased valve stiffness and valvular insufficiency. Valve abnormalities were only detected in late gestation and early postnatal mutant animals and worsened with age. The mutant mice developed perturbed extracellular matrix (ECM) deposition and remodeling characterized by increased glycosaminoglycan and decreased collagen content, as well as increased valve interstitial cell expression of Sox9, a transcription factor essential for normal ECM maturation during heart valve development. This study provides the first evidence that Tie1 is involved in modulation of late valve remodeling and suggests that an important Tie1-Sox9 signaling axis exists through which disturbed flows are converted by endocardial cells to paracrine Sox9 signals to modulate normal matrix remodeling of the aortic valve.
Collapse
Affiliation(s)
- Xianghu Qu
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - Kate Violette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - M K Sewell-Loftin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jonathan Soslow
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA
| | - LeShana Saint-Jean
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Robert B Hinton
- Division of Cardiology, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - H Scott Baldwin
- Division of Pediatrics Cardiology, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
7
|
Saint-Jean L, Barkas N, Harmelink C, Tompkins KL, Oakey RJ, Baldwin HS. Myocardial differentiation is dependent upon endocardial signaling during early cardiogenesis in vitro. Development 2019; 146:dev.172619. [PMID: 31023876 DOI: 10.1242/dev.172619] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/10/2019] [Indexed: 01/18/2023]
Abstract
The endocardium interacts with the myocardium to promote proliferation and morphogenesis during the later stages of heart development. However, the role of the endocardium in early cardiac ontogeny remains under-explored. Given the shared origin, subsequent juxtaposition, and essential cell-cell interactions of endocardial and myocardial cells throughout heart development, we hypothesized that paracrine signaling from the endocardium to the myocardium is crucial for initiating early differentiation of myocardial cells. To test this, we generated an in vitro, endocardial-specific ablation model using the diphtheria toxin receptor under the regulatory elements of the Nfat c1 genomic locus (NFATc1-DTR). Early treatment of NFATc1-DTR mouse embryoid bodies with diphtheria toxin efficiently ablated endocardial cells, which significantly attenuated the percentage of beating EBs in culture and expression of early and late myocardial differentiation markers. The addition of Bmp2 during endocardial ablation partially rescued myocyte differentiation, maturation and function. Therefore, we conclude that early stages of myocardial differentiation rely on endocardial paracrine signaling mediated in part by Bmp2. Our findings provide novel insight into early endocardial-myocardial interactions that can be explored to promote early myocardial development and growth.
Collapse
Affiliation(s)
- Leshana Saint-Jean
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nikolaos Barkas
- Department of Medical & Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - Cristina Harmelink
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kevin L Tompkins
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Rebecca J Oakey
- Department of Medical & Molecular Genetics, King's College London, London, SE1 9RT, UK
| | - H Scott Baldwin
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA .,Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
8
|
Luxán G, D'Amato G, MacGrogan D, de la Pompa JL. Endocardial Notch Signaling in Cardiac Development and Disease. Circ Res 2015; 118:e1-e18. [PMID: 26635389 DOI: 10.1161/circresaha.115.305350] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/22/2015] [Indexed: 01/03/2023]
Abstract
The Notch signaling pathway is an ancient and highly conserved signaling pathway that controls cell fate specification and tissue patterning in the embryo and in the adult. Region-specific endocardial Notch activity regulates heart morphogenesis through the interaction with multiple myocardial-, epicardial-, and neural crest-derived signals. Mutations in NOTCH signaling elements cause congenital heart disease in humans and mice, demonstrating its essential role in cardiac development. Studies in model systems have provided mechanistic understanding of Notch function in cardiac development, congenital heart disease, and heart regeneration. Notch patterns the embryonic endocardium into prospective territories for valve and chamber formation, and later regulates the signaling processes leading to outflow tract and valve morphogenesis and ventricular trabeculae compaction. Alterations in NOTCH signaling in the endocardium result in congenital structural malformations that can lead to disease in the neonate and adult heart.
Collapse
Affiliation(s)
- Guillermo Luxán
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - Gaetano D'Amato
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - Donal MacGrogan
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.)
| | - José Luis de la Pompa
- From the Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovascular (CNIC), Melchor Fernández Almagro, Madrid, Spain (G.L., G.D'A., D.M., J.L.d.l.P.); and Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany (G.L.).
| |
Collapse
|
9
|
Coram RJ, Stillwagon SJ, Guggilam A, Jenkins MW, Swanson MS, Ladd AN. Muscleblind-like 1 is required for normal heart valve development in vivo. BMC DEVELOPMENTAL BIOLOGY 2015; 15:36. [PMID: 26472242 PMCID: PMC4608261 DOI: 10.1186/s12861-015-0087-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/09/2015] [Indexed: 12/26/2022]
Abstract
Background Development of the valves and septa of the heart depends on the formation and remodeling of the endocardial cushions in the atrioventricular canal and outflow tract. These cushions are populated by mesenchyme produced from the endocardium by epithelial-mesenchymal transition (EMT). The endocardial cushions are remodeled into the valves at post-EMT stages via differentiation of the mesenchyme and changes in the extracellular matrix (ECM). Transforming growth factor β (TGFβ) signaling has been implicated in both the induction of EMT in the endocardial cushions and the remodeling of the valves at post-EMT stages. We previously identified the RNA binding protein muscleblind-like 1 (MBNL1) as a negative regulator of TGFβ signaling and EMT in chicken endocardial cushions ex vivo. Here, we investigate the role of MBNL1 in endocardial cushion development and valvulogenesis in Mbnl1∆E3/∆E3 mice, which are null for MBNL1 protein. Methods Collagen gel invasion assays, histology, immunohistochemistry, real-time RT-PCR, optical coherence tomography, and echocardiography were used to evaluate EMT and TGFβ signaling in the endocardial cushions, and morphogenesis, ECM composition, and function of the heart valves. Results As in chicken, the loss of MBNL1 promotes precocious TGFβ signaling and EMT in the endocardial cushions. Surprisingly, this does not lead to the production of excess mesenchyme, but later valve morphogenesis is aberrant. Adult Mbnl1∆E3/∆E3 mice exhibit valve dysmorphia with elevated TGFβ signaling, changes in ECM composition, and increased pigmentation. This is accompanied by a high incidence of regurgitation across both inflow and outflow valves. Mbnl1∆E3/∆E3 mice also have a high incidence of ostium secundum septal defects accompanied by atrial communication, but do not develop overt cardiomyopathy. Conclusions Together, these data indicate that MBNL1 plays a conserved role in negatively regulating TGFβ signaling, and is required for normal valve morphogenesis and homeostasis in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0087-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan J Coram
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Present Address: Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA.
| | - Samantha J Stillwagon
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Present Address: Department of Obstetrics and Gynecology, Women's Health Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Anuradha Guggilam
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Michael W Jenkins
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Maurice S Swanson
- Department of Molecular Genetics & Microbiology, College of Medicine, Center for NeuroGenetics and the Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Andrea N Ladd
- Department of Cellular & Molecular Medicine, Lerner Research Institute, 9500 Euclid Ave. NC10, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
10
|
Krishnan A, Samtani R, Dhanantwari P, Lee E, Yamada S, Shiota K, Donofrio MT, Leatherbury L, Lo CW. A detailed comparison of mouse and human cardiac development. Pediatr Res 2014; 76:500-7. [PMID: 25167202 PMCID: PMC4233008 DOI: 10.1038/pr.2014.128] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 05/29/2014] [Indexed: 11/09/2022]
Abstract
BACKGROUND Mouse mutants are used to model human congenital cardiovascular disease. Few studies exist comparing normal cardiovascular development in mice vs. humans. We carried out a systematic comparative analysis of mouse and human fetal cardiovascular development. METHODS Episcopic fluorescence image capture (EFIC) was performed on 66 wild-type mouse embryos from embryonic day (E) 9.5 to birth; 2-dimensional and 3-dimensional datasets were compared with EFIC and magnetic resonance images from a study of 52 human fetuses (Carnegie stage 13-23). RESULTS Time course of atrial, ventricular, and outflow septation were outlined and followed a similar sequence in both species. Bilateral venae cavae and prominent atrial appendages were seen in the mouse fetus; in human fetuses, atrial appendages were small, and a single right superior vena cava was present. In contrast to humans with separate pulmonary vein orifices, a pulmonary venous confluence with one orifice enters the left atrium in mice. CONCLUSION The cardiac developmental sequences observed in mouse and human fetuses are comparable, with minor differences in atrial and venous morphology. These comparisons of mouse and human cardiac development strongly support that mouse morphogenesis is a good model for human development.
Collapse
Affiliation(s)
- Anita Krishnan
- Laboratory of Developmental Biology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD; United States,Children’s National Heart Institute; Children’s National Medical Center; Washington, DC; United States
| | - Rajeev Samtani
- Laboratory of Developmental Biology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD; United States
| | - Preeta Dhanantwari
- Division of Pediatric Cardiology; Schneider Children’s Hospital; New Hyde Park, NY; United States
| | - Elaine Lee
- Laboratory of Developmental Biology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD; United States
| | - Shigehito Yamada
- Congenital Anomaly Research Center, Kyoto University Graduate School of Medicine; Kyoto, Japan
| | - Kohei Shiota
- Congenital Anomaly Research Center, Kyoto University Graduate School of Medicine; Kyoto, Japan
| | - Mary T. Donofrio
- Children’s National Heart Institute; Children’s National Medical Center; Washington, DC; United States
| | - Linda Leatherbury
- Laboratory of Developmental Biology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD; United States,Children’s National Heart Institute; Children’s National Medical Center; Washington, DC; United States
| | - Cecilia W. Lo
- Laboratory of Developmental Biology; National Heart, Lung, and Blood Institute; National Institutes of Health; Bethesda, MD; United States,Department of Developmental Biology; University of Pittsburgh School of Medicine; Pittsburgh, PA; United States
| |
Collapse
|
11
|
Peng Y, Song L, Zhao M, Harmelink C, Debenedittis P, Cui X, Wang Q, Jiao K. Critical roles of miRNA-mediated regulation of TGFβ signalling during mouse cardiogenesis. Cardiovasc Res 2014; 103:258-67. [PMID: 24835278 DOI: 10.1093/cvr/cvu126] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS MicroRNAs (miRNAs) play critical roles during the development of the cardiovascular system. Blocking miRNA biosynthesis in embryonic hearts through a conditional gene inactivation approach led to differential cardiac defects depending on the Cre drivers used in different studies. The goal of this study is to reveal the cardiogenic pathway that is regulated by the miRNA mechanism at midgestation, a stage that has not been evaluated in previous publications. METHODS AND RESULTS We specifically inactivated Dicer1, which is essential for generation of functional mature miRNAs, in the myocardium by crossing cTnt-Cre mice with Dicer1(loxP) mice. cTnt-Cre efficiently inactivates target genes in cardiomyocytes at midgestation. All mutants died between E14.5 and E16.5 with severe myocardial wall defects, including reduced cell proliferation, increased cell death, and spongy myocardial wall. Expression of TGFβ type I receptor (Tgfbr1), which encodes the Type I receptor of TGFβ ligands, was up-regulated in mutant hearts. As expected, TGFβ activity was increased in Dicer1-inactivated hearts. Our further molecular analysis suggested that Tgfbr1 is a direct target of three miRNAs. Reducing TGFβ activities using a pharmacological inhibitor on in vitro cultured hearts, or through an in vivo genetic approach, partially rescued the cardiac defects caused by Dicer1 inactivation. CONCLUSIONS We show for the first time that TGFβ signalling is directly regulated by the miRNA mechanism during myocardial wall morphogenesis. Increased TGFβ activity plays a major role in the cardiac defects caused by myocardial deletion of Dicer1. Thus, miRNA-mediated regulation of TGFβ signalling is indispensable for normal cardiogenesis.
Collapse
Affiliation(s)
- Yin Peng
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| | - Lanying Song
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| | - Mei Zhao
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| | - Cristina Harmelink
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| | - Paige Debenedittis
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| | - Xiangqin Cui
- Department of Biostatistics, The University of Alabama at Birmingham, Birmingham, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, USA
| | - Kai Jiao
- Division of Research, Department of Genetics, The University of Alabama at Birmingham, 720 20th St. S., 768 Kaul Building, Birmingham AL 35294, USA
| |
Collapse
|
12
|
Sewell-Loftin MK, DeLaughter DM, Peacock JR, Brown CB, Baldwin HS, Barnett JV, Merryman WD. Myocardial contraction and hyaluronic acid mechanotransduction in epithelial-to-mesenchymal transformation of endocardial cells. Biomaterials 2014; 35:2809-15. [PMID: 24433835 DOI: 10.1016/j.biomaterials.2013.12.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) of endocardial cells is a critical initial step in the formation of heart valves. The collagen gel in vitro model has provided significant information on the role of growth factors regulating EMT but has not permitted investigation of mechanical factors. Therefore we sought to develop a system to probe the effects of mechanical inputs on endocardial EMT by incorporating hyaluronic acid (HA), the primary component of endocardial cushions in developing heart valves, into the gel assay. This was achieved using a combination collagen and crosslinkable methacrylated HA hydrogel (Coll-MeHA). Avian atrioventricular canal explants on Coll-MeHA gels showed increased numbers of transformed cells. Analysis of the mechanical properties of Coll-MeHA gels shows that stiffness does not directly affect EMT. Hydrogel deformation from the beating myocardium of explants directly led to higher levels of regional gel deformation and larger average strain magnitudes associated with invaded cells on Coll-MeHA gels. Inhibition of this contraction reduced EMT on all gel types, although to a lesser extent on Coll-MeHA gels. Using the system we have developed, which permits the manipulation of mechanical factors, we have demonstrated that active mechanical forces play a role in the regulation of endocardial EMT.
Collapse
Affiliation(s)
| | - Daniel M DeLaughter
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Jon R Peacock
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Christopher B Brown
- Department of Pediatrics, Division of Cardiology, Vanderbilt University, Nashville, TN 37232, USA
| | - H Scott Baldwin
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Pediatrics, Division of Cardiology, Vanderbilt University, Nashville, TN 37232, USA
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
13
|
Kain KH, Miller JWI, Jones-Paris CR, Thomason RT, Lewis JD, Bader DM, Barnett JV, Zijlstra A. The chick embryo as an expanding experimental model for cancer and cardiovascular research. Dev Dyn 2013; 243:216-28. [PMID: 24357262 DOI: 10.1002/dvdy.24093] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 10/28/2013] [Accepted: 10/28/2013] [Indexed: 12/17/2022] Open
Abstract
A long and productive history in biomedical research defines the chick as a model for human biology. Fundamental discoveries, including the description of directional circulation propelled by the heart and the link between oncogenes and the formation of cancer, indicate its utility in cardiac biology and cancer. Despite the more recent arrival of several vertebrate and invertebrate animal models during the last century, the chick embryo remains a commonly used model for vertebrate biology and provides a tractable biological template. With new molecular and genetic tools applied to the avian genome, the chick embryo is accelerating the discovery of normal development and elusive disease processes. Moreover, progress in imaging and chick culture technologies is advancing real-time visualization of dynamic biological events, such as tissue morphogenesis, angiogenesis, and cancer metastasis. A rich background of information, coupled with new technologies and relative ease of maintenance, suggest an expanding utility for the chick embryo in cardiac biology and cancer research.
Collapse
|
14
|
Schumacher JA, Bloomekatz J, Garavito-Aguilar ZV, Yelon D. tal1 Regulates the formation of intercellular junctions and the maintenance of identity in the endocardium. Dev Biol 2013; 383:214-26. [PMID: 24075907 DOI: 10.1016/j.ydbio.2013.09.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 09/13/2013] [Accepted: 09/16/2013] [Indexed: 11/26/2022]
Abstract
The endocardium forms the inner lining of the heart tube, where it enables blood flow and also interacts with the myocardium during the formation of valves and trabeculae. Although a number of studies have identified regulators in the morphogenesis of the myocardium, relatively little is known about the molecules that control endocardial morphogenesis. Prior work has implicated the bHLH transcription factor Tal1 in endocardial tube formation: in zebrafish embryos lacking Tal1, endocardial cells form a disorganized mass within the ventricle and do not populate the atrium. Through blastomere transplantation, we find that tal1 plays a cell-autonomous role in regulating endocardial extension, suggesting that Tal1 activity influences the behavior of individual endocardial cells. The defects in endocardial behavior in tal1-deficient embryos originate during the earliest steps of endocardial morphogenesis: tal1-deficient endocardial cells fail to generate a cohesive monolayer at the midline and instead pack tightly together into a multi-layered aggregate. Moreover, the tight junction protein ZO-1 is mislocalized in the tal1-deficient endocardium, indicating a defect in intercellular junction formation. In addition, we find that the tal1-deficient endocardium fails to maintain its identity; over time, a progressively increasing number of tal1-deficient endocardial cells initiate myocardial gene expression. However, the onset of defects in intercellular junction formation precedes the onset of ectopic myocardial gene expression in the tal1-deficient endocardium. We therefore propose a model in which Tal1 has distinct roles in regulating the formation of endocardial intercellular junctions and maintaining endocardial identity.
Collapse
Affiliation(s)
- Jennifer A Schumacher
- Developmental Genetics Program and Department of Cell Biology, Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
15
|
Blech-Hermoni Y, Ladd AN. RNA binding proteins in the regulation of heart development. Int J Biochem Cell Biol 2013; 45:2467-78. [PMID: 23973289 DOI: 10.1016/j.biocel.2013.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 11/28/2022]
Abstract
In vivo, RNA molecules are constantly accompanied by RNA binding proteins (RBPs), which are intimately involved in every step of RNA biology, including transcription, editing, splicing, transport and localization, stability, and translation. RBPs therefore have opportunities to shape gene expression at multiple levels. This capacity is particularly important during development, when dynamic chemical and physical changes give rise to complex organs and tissues. This review discusses RBPs in the context of heart development. Since the targets and functions of most RBPs--in the heart and at large--are not fully understood, this review focuses on the expression and roles of RBPs that have been implicated in specific stages of heart development or developmental pathology. RBPs are involved in nearly every stage of cardiogenesis, including the formation, morphogenesis, and maturation of the heart. A fuller understanding of the roles and substrates of these proteins could ultimately provide attractive targets for the design of therapies for congenital heart defects, cardiovascular disease, or cardiac tissue repair.
Collapse
Affiliation(s)
- Yotam Blech-Hermoni
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Program in Cell Biology, Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | |
Collapse
|
16
|
Allison P, Huang T, Broka D, Parker P, Barnett JV, Camenisch TD. Disruption of canonical TGFβ-signaling in murine coronary progenitor cells by low level arsenic. Toxicol Appl Pharmacol 2013; 272:147-53. [PMID: 23732083 DOI: 10.1016/j.taap.2013.04.035] [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: 01/10/2013] [Revised: 04/11/2013] [Accepted: 04/30/2013] [Indexed: 11/30/2022]
Abstract
Exposure to arsenic results in several types of cancers as well as heart disease. A major contributor to ischemic heart pathologies is coronary artery disease, however the influences by environmental arsenic in this disease process are not known. Similarly, the impact of toxicants on blood vessel formation and function during development has not been studied. During embryogenesis, the epicardium undergoes proliferation, migration, and differentiation into several cardiac cell types including smooth muscle cells which contribute to the coronary vessels. The TGFβ family of ligands and receptors is essential for developmental cardiac epithelial to mesenchymal transition (EMT) and differentiation into coronary smooth muscle cells. In this in vitro study, 18hour exposure to 1.34μM arsenite disrupted developmental EMT programming in murine epicardial cells causing a deficit in cardiac mesenchyme. The expression of EMT genes including TGFβ2, TGFβ receptor-3, Snail, and Has-2 are decreased in a dose-dependent manner following exposure to arsenite. TGFβ2 cell signaling is abrogated as detected by decreases in phosphorylated Smad2/3 when cells are exposed to 1.34μM arsenite. There is also loss of nuclear accumulation pSmad due to arsenite exposure. These observations coincide with a decrease in vimentin positive mesenchymal cells invading three-dimensional collagen gels. However, arsenite does not block TGFβ2 mediated smooth muscle cell differentiation by epicardial cells. Overall these results show that arsenic exposure blocks developmental EMT gene programming in murine coronary progenitor cells by disrupting TGFβ2 signals and Smad activation, and that smooth muscle cell differentiation is refractory to this arsenic toxicity.
Collapse
Affiliation(s)
- Patrick Allison
- Department of Pharmacology and Toxicology College of Pharmacy, Southwest Environmental Health Sciences Center, Steele Children's Research Center and Bio5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | | | | | | | | | | |
Collapse
|
17
|
Wu B, Baldwin HS, Zhou B. Nfatc1 directs the endocardial progenitor cells to make heart valve primordium. Trends Cardiovasc Med 2013; 23:294-300. [PMID: 23669445 DOI: 10.1016/j.tcm.2013.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/09/2013] [Accepted: 04/10/2013] [Indexed: 11/26/2022]
Abstract
Heart valves arise from the cardiac endocardial cushions located at the atrioventricular canal (AVC) and cardiac outflow tract (OFT) during development. A subpopulation of cushion endocardial cells undergoes endocardial to mesenchymal transformation (EMT) and generates the cushion mesenchyme, which is then remodeled into the interstitial tissue of the mature valves. The cushion endocardial cells that do not undertake EMT proliferate to elongate valve leaflets. During EMT and the post-EMT valve remodeling, endocardial cells at the cushions highly express nuclear factor in activated T cell, cytoplasmic 1 (Nfatc1), a transcription factor required for valve formation in mice. In this review, we present the current knowledge of Nfatc1 roles in the ontogeny of heart valves with a focus on the fate decision of the endocardial cells in the processes of EMT and valve remodeling.
Collapse
Affiliation(s)
- Bingruo Wu
- Department of Genetics, Division of Cardiology, Wilf Cardiovascular Institute, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA; Department of Pediatrics, Division of Cardiology, Wilf Cardiovascular Institute, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA; Department of Medicine, Division of Cardiology, Wilf Cardiovascular Institute, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | | | | |
Collapse
|
18
|
DeLaughter DM, Christodoulou DC, Robinson JY, Seidman CE, Baldwin HS, Seidman JG, Barnett JV. Spatial transcriptional profile of the chick and mouse endocardial cushions identify novel regulators of endocardial EMT in vitro. J Mol Cell Cardiol 2013; 59:196-204. [PMID: 23557753 DOI: 10.1016/j.yjmcc.2013.03.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/15/2013] [Accepted: 03/21/2013] [Indexed: 11/17/2022]
Abstract
Valvular Interstitial Cells (VICs) are a common substrate for congenital and adult heart disease yet the signaling mechanisms governing their formation during early valvulogenesis are incompletely understood. We developed an unbiased strategy to identify genes important in endocardial epithelial-to-mesenchymal transformation (EMT) using a spatial transcriptional profile. Endocardial cells overlaying the cushions of the atrioventricular canal (AVC) and outflow tract (OFT) undergo an EMT to yield VICs. RNA sequencing (RNA-seq) analysis of gene expression between AVC, OFT, and ventricles (VEN) isolated from chick and mouse embryos at comparable stages of development (chick HH18; mouse E11.0) was performed. EMT occurs in the AVC and OFT cushions, but not VEN at this time. 198 genes in the chick (n=1) and 105 genes in the mouse (n=2) were enriched 2-fold in the cushions. Gene regulatory networks (GRN) generated from cushion-enriched gene lists confirmed TGFβ as a nodal point and identified NF-κB as a potential node. To reveal previously unrecognized regulators of EMT four candidate genes, Hapln1, Id1, Foxp2, and Meis2, and a candidate pathway, NF-κB, were selected. In vivo spatial expression of each gene was confirmed by in situ hybridization and a functional role for each in endocardial EMT was determined by siRNA knockdown in a collagen gel assay. Our spatial-transcriptional profiling strategy yielded gene lists which reflected the known biology of the system. Further analysis accurately identified and validated previously unrecognized novel candidate genes and the NF-κB pathway as regulators of endocardial cell EMT in vitro.
Collapse
Affiliation(s)
- Daniel M DeLaughter
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Jun is required in Isl1-expressing progenitor cells for cardiovascular development. PLoS One 2013; 8:e57032. [PMID: 23437302 PMCID: PMC3578783 DOI: 10.1371/journal.pone.0057032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/18/2013] [Indexed: 01/20/2023] Open
Abstract
Jun is a highly conserved member of the multimeric activator protein 1 transcription factor complex and plays an important role in human cancer where it is known to be critical for proliferation, cell cycle regulation, differentiation, and cell death. All of these biological functions are also crucial for embryonic development. Although all Jun null mouse embryos die at mid-gestation with persistent truncus arteriosus, a severe cardiac outflow tract defect also seen in human congenital heart disease, the developmental mechanisms are poorly understood. Here we show that murine Jun is expressed in a restricted pattern in several cell populations important for cardiovascular development, including the second heart field, pharyngeal endoderm, outflow tract and atrioventricular endocardial cushions and post-migratory neural crest derivatives. Several genes, including Isl1, molecularly mark the second heart field. Isl1 lineages include myocardium, smooth muscle, neural crest, endocardium, and endothelium. We demonstrate that conditional knockout mouse embryos lacking Jun in Isl1-expressing progenitors display ventricular septal defects, double outlet right ventricle, semilunar valve hyperplasia and aortic arch artery patterning defects. In contrast, we show that conditional deletion of Jun in Tie2-expressing endothelial and endocardial precursors does not result in aortic arch artery patterning defects or embryonic death, but does result in ventricular septal defects and a low incidence of semilunar valve defects, atrioventricular valve defects and double outlet right ventricle. Our results demonstrate that Jun is required in Isl1-expressing progenitors and, to a lesser extent, in endothelial cells and endothelial-derived endocardium for cardiovascular development but is dispensable in both cell types for embryonic survival. These data provide a cellular framework for understanding the role of Jun in the pathogenesis of congenital heart disease.
Collapse
|
20
|
Wilson CL, Gough PJ, Chang CA, Chan CK, Frey JM, Liu Y, Braun KR, Chin MT, Wight TN, Raines EW. Endothelial deletion of ADAM17 in mice results in defective remodeling of the semilunar valves and cardiac dysfunction in adults. Mech Dev 2013; 130:272-89. [PMID: 23354118 DOI: 10.1016/j.mod.2013.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/21/2012] [Accepted: 01/07/2013] [Indexed: 12/24/2022]
Abstract
Global inactivation of the metalloproteinase ADAM17 during mouse development results in perinatal lethality and abnormalities of the heart, including late embryonic cardiomegaly and thickened semilunar and atrioventricular valves. These defects have been attributed in part to a lack of ADAM17-mediated processing of HB-EGF, as absence of soluble HB-EGF results in similar phenotypes. Because valvular mesenchymal cells are largely derived from cardiac endothelial cells, we generated mice with a floxed Adam17 allele and crossed these animals with Tie2-Cre transgenics to focus on the role of endothelial ADAM17 in valvulogenesis. We find that although hearts from late-stage embryos with ablation of endothelial ADAM17 appear normal, an increase in valve size and cell number is evident, but only in the semilunar cusps. Unlike Hbegf(-/-) valves, ADAM17-null semilunar valves do not differ from controls in acute cell proliferation at embryonic day 14.5 (E14.5), suggesting compensatory processing of HB-EGF. However, levels of the proteoglycan versican are significantly reduced in mutant hearts early in valve remodeling (E12.5). After birth, aortic valve cusps from mutants are not only hyperplastic but also show expansion of the glycosaminoglycan-rich component, with the majority of adults exhibiting aberrant compartmentalization of versican and increased deposition of collagen. The inability of mutant outflow valve precursors to transition into fully mature cusps is associated with decreased postnatal viability, progressive cardiomegaly, and systolic dysfunction. Together, our data indicate that ADAM17 is required in valvular endothelial cells for regulating cell content as well as extracellular matrix composition and organization in semilunar valve remodeling and homeostasis.
Collapse
Affiliation(s)
- Carole L Wilson
- Department of Pathology, University of Washington, Seattle, WA 98104, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Vijayaraj P, Le Bras A, Mitchell N, Kondo M, Juliao S, Wasserman M, Beeler D, Spokes K, Aird WC, Baldwin HS, Oettgen P. Erg is a crucial regulator of endocardial-mesenchymal transformation during cardiac valve morphogenesis. Development 2012; 139:3973-85. [PMID: 22932696 PMCID: PMC3472597 DOI: 10.1242/dev.081596] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During murine embryogenesis, the Ets factor Erg is highly expressed in endothelial cells of the developing vasculature and in articular chondrocytes of developing bone. We identified seven isoforms for the mouse Erg gene. Four share a common translational start site encoded by exon 3 (Ex3) and are enriched in chondrocytes. The other three have a separate translational start site encoded by Ex4 and are enriched in endothelial cells. Homozygous ErgΔEx3/ΔEx3 knockout mice are viable, fertile and do not display any overt phenotype. By contrast, homozygous ErgΔEx4/ΔEx4 knockout mice are embryonic lethal, which is associated with a marked reduction in endocardial-mesenchymal transformation (EnMT) during cardiac valve morphogenesis. We show that Erg is required for the maintenance of the core EnMT regulatory factors that include Snail1 and Snail2 by binding to their promoter and intronic regions.
Collapse
Affiliation(s)
- Preethi Vijayaraj
- Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lencinas A, Tavares ALP, Barnett JV, Runyan RB. Collagen gel analysis of epithelial-mesenchymal transition in the embryo heart: an in vitro model system for the analysis of tissue interaction, signal transduction, and environmental effects. ACTA ACUST UNITED AC 2012; 93:298-311. [PMID: 22271679 DOI: 10.1002/bdrc.20222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The cellular process of epithelial-mesenchymal cell transition (EMT) is a critical event in development that is reiterated in adult pathologies of metastasis and organ fibrosis. An initial understanding of the cellular and molecular events of this process emerged from an in vitro examination of heart valve development. Explants of the chick atrioventricular valve-forming region were placed on collagen gels and removed to show that EMT was regulated by a tissue interaction. Subsequent studies showed that specific TGFβ isoforms and receptors were required and steps of activation and invasion could be distinguished. The assay was modified for mouse hearts and has been used to explore signal transduction and gene expression in both species. The principle advantages of the system are a defined temporal window, when EMT takes place and the ability to isolate cells at various stages of the EMT process. These advantages are largely unavailable in other developmental or adult models. As the mesenchymal cells produced by EMT in the heart are involved in defects found in congenital heart disease, there is also a direct relevance of cardiac EMT to human birth defects. This relationship has been explored in relation to environmental exposures and in a number of genetic models. This review provides both an overview of the findings developed from the assay and protocols to enable the use of the assay by other laboratories. The assay provides a versatile platform to explore roles of specific gene products, drugs, and environmental agents on a critical cellular process.
Collapse
Affiliation(s)
- Alejandro Lencinas
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | | | | | | |
Collapse
|
23
|
VanDusen NJ, Firulli AB. Twist factor regulation of non-cardiomyocyte cell lineages in the developing heart. Differentiation 2012; 84:79-88. [PMID: 22516205 DOI: 10.1016/j.diff.2012.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 02/14/2012] [Accepted: 03/07/2012] [Indexed: 12/31/2022]
Abstract
The heart is a complex organ that is composed of numerous cell types, which must integrate their programs for proper specification, differentiation and cardiac morphogenesis. During cardiogenesis members of the Twist-family of basic helix-loop-helix (bHLH) transcription factors play distinct roles within cardiac lineages such as the endocardium and extra-cardiac lineages such as the cardiac neural crest (cNCC) and epicardium. While the study of these cell populations is often eclipsed by that of cardiomyocytes, the contributions of non-cardiomyocytes to development and disease are increasingly being appreciated as both dynamic and essential. This review summarizes what is known regarding Twist-family bHLH function in extra-cardiac cell populations and the endocardium, with a focus on regulatory mechanisms, downstream targets, and expression profiles. Improving our understanding of the molecular pathways that Twist-family bHLH factors mediate in these lineages will be necessary to ascertain how their dysfunction leads to congenital disease and adult pathologies such as myocardial infarctions and cardiac fibroblast induced fibrosis. Indeed, this knowledge will prove to be critical to clinicians seeking to improve current treatments.
Collapse
Affiliation(s)
- Nathan J VanDusen
- Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Department of Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
| | | |
Collapse
|
24
|
Sánchez NS, Barnett JV. TGFβ and BMP-2 regulate epicardial cell invasion via TGFβR3 activation of the Par6/Smurf1/RhoA pathway. Cell Signal 2012; 24:539-548. [PMID: 22033038 PMCID: PMC3237859 DOI: 10.1016/j.cellsig.2011.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 10/10/2011] [Indexed: 01/19/2023]
Abstract
Coronary vessel development requires transfer of mesothelial cells to the heart surface to form the epicardium where some cells subsequently undergo epithelial-mesenchymal transformation (EMT) and invade the subepicardial matrix. Tgfbr3(-/-) mice die due to failed coronary vessel formation associated with decreased epicardial cell invasion but the mediators downstream of TGFβR3 are not well described. TGFβR3-dependent endocardial EMT stimulated by either TGFβ2 or BMP-2 requires activation of the Par6/Smurf1/RhoA 1pathway where Activin Receptor Like Kinase (ALK5) signals Par6 to act downstream of TGFβ to recruit Smurf1 to target RhoA for degradation to regulate apical-basal polarity and tight junction dissolution. Here we asked if this pathway was operant in epicardial cells and if TGFβR3 was required to access this pathway. Targeting of ALK5 in Tgfbr3(+/+) cells inhibited loss of epithelial character and invasion. Overexpression of wild-type (wt) Par6, but not dominant negative (dn) Par6, induced EMT and invasion while targeting Par6 by siRNA inhibited EMT and invasion. Overexpression of Smurf1 and dnRhoA induced loss of epithelial character and invasion. Targeting of Smurf1 by siRNA or overexpression of constitutively active (ca) RhoA inhibited EMT and invasion. In Tgfbr3(-/-) epicardial cells which have a decreased ability to invade collagen gels in response to TGFβ2, overexpression of wtPar6, Smurf1, or dnRhoA had a diminished ability to induce invasion. Overexpression of TGFβR3 in Tgfbr3(-/-) cells, followed by siRNA targeting of Par6 or Smurf1, diminished the ability of TGFβR3 to rescue invasion demonstrating that the Par6/Smurf1/RhoA pathway is activated downstream of TGFβR3 in epicardial cells.
Collapse
Affiliation(s)
- Nora S Sánchez
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| |
Collapse
|
25
|
Hill CR, Sanchez NS, Love JD, Arrieta JA, Hong CC, Brown CB, Austin AF, Barnett JV. BMP2 signals loss of epithelial character in epicardial cells but requires the Type III TGFβ receptor to promote invasion. Cell Signal 2012; 24:1012-22. [PMID: 22237159 DOI: 10.1016/j.cellsig.2011.12.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 12/28/2011] [Indexed: 12/21/2022]
Abstract
Coronary vessel development depends on a subpopulation of epicardial cells that undergo epithelial to mesenchymal transformation (EMT) and invade the subepicardial space and myocardium. These cells form the smooth muscle of the vessels and fibroblasts, but the mechanisms that regulate these processes are poorly understood. Mice lacking the Type III Transforming Growth Factor β Receptor (TGFβR3) die by E14.5 due to failed coronary vessel development accompanied by reduced epicardial cell invasion. BMP2 signals via TGFβR3 emphasizing the importance of determining the relative contributions of the canonical BMP signaling pathway and TGFβR3-dependent signaling to BMP2 responsiveness. Here we examined the role of TGFβR3 in BMP2 signaling in epicardial cells. Whereas TGFβ induced loss of epithelial character and smooth muscle differentiation, BMP2 induced an ALK3-dependent loss of epithelial character and modestly inhibited TGFβ-stimulated differentiation. Tgfbr3(-/-) cells respond to BMP2 indicating that TGFβR3 is not required. However, Tgfbr3(-/-) cells show decreased invasion in response to BMP2 and overexpression of TGFβR3 in Tgfbr3(-/-) cells rescued invasion. Invasion was dependent on ALK5, ALK2, ALK3, and Smad4. Expression of TGFβR3 lacking the 3 C-terminal amino acids required to interact with the scaffolding protein GIPC (GAIP-interacting protein, C terminus) did not rescue. Knockdown of GIPC in Tgfbr3(+/+) or Tgfbr3(-/-) cells rescued with TGFβR3 decreased BMP2-stimulated invasion confirming a requirement for TGFβR3/GIPC interaction. Our results reveal the relative roles of TGFβR3-dependent and TGFβR3-independent signaling in the actions of BMP2 on epicardial cell behavior and demonstrate the critical role of TGFβR3 in mediating BMP2-stimulated invasion.
Collapse
|
26
|
Townsend TA, Robinson JY, How T, DeLaughter DM, Blobe GC, Barnett JV. Endocardial cell epithelial-mesenchymal transformation requires Type III TGFβ receptor interaction with GIPC. Cell Signal 2011; 24:247-56. [PMID: 21945156 DOI: 10.1016/j.cellsig.2011.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/02/2011] [Accepted: 09/05/2011] [Indexed: 12/19/2022]
Abstract
An early event in heart valve formation is the epithelial-mesenchymal transformation (EMT) of a subpopulation of endothelial cells in specific regions of the heart tube, the endocardial cushions. The Type III TGFβ receptor (TGFβR3) is required for TGFβ2- or BMP-2-stimulated EMT in atrioventricular endocardial cushion (AVC) explants in vitro but the mediators downstream of TGFβR3 are not well described. Using AVC and ventricular explants as an in vitro assay, we found an absolute requirement for specific TGFβR3 cytoplasmic residues, GAIP-interacting protein, C terminus (GIPC), and specific Activin Receptor-Like Kinases (ALK)s for TGFβR3-mediated EMT when stimulated by TGFβ2 or BMP-2. The introduction of TGFβR3 into nontransforming ventricular endocardial cells, followed by the addition of either TGFβ2 or BMP-2, results in EMT. TGFβR3 lacking the entire cytoplasmic domain, or only the 3C-terminal amino acids that are required to bind GIPC, fails to support EMT in response to TGFβ2 or BMP-2. Overexpression of GIPC in AVC endocardial cells enhanced EMT while siRNA-mediated silencing of GIPC in ventricular cells overexpressing TGFβR3 significantly inhibited EMT. Targeting of specific ALKs by siRNA revealed that TGFβR3-mediated EMT requires ALK2 and ALK3, in addition to ALK5, but not ALK4 or ALK6. Taken together, these data identify GIPC, ALK2, ALK3, and ALK5 as signaling components required for TGFβR3-mediated endothelial cell EMT.
Collapse
Affiliation(s)
- Todd A Townsend
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Azhar M, Brown K, Gard C, Chen H, Rajan S, Elliott DA, Stevens MV, Camenisch TD, Conway SJ, Doetschman T. Transforming growth factor Beta2 is required for valve remodeling during heart development. Dev Dyn 2011; 240:2127-41. [PMID: 21780244 DOI: 10.1002/dvdy.22702] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2011] [Indexed: 01/31/2023] Open
Abstract
Although the function of transforming growth factor beta2 (TGFβ2) in epithelial mesenchymal transition (EMT) is well studied, its role in valve remodeling remains to be fully explored. Here, we used histological, morphometric, immunohistochemical and molecular approaches and showed that significant dysregulation of major extracellular matrix (ECM) components contributed to valve remodeling defects in Tgfb2(-/-) embryos. The data indicated that cushion mesenchymal cell differentiation was impaired in Tgfb2(-/-) embryos. Hyaluronan and cartilage link protein-1 (CRTL1) were increased in hyperplastic valves of Tgfb2(-/-) embryos, indicating increased expansion and diversification of cushion mesenchyme into the cartilage cell lineage during heart development. Finally, Western blot and immunohistochemistry analyses indicate that the activation of SMAD2/3 was decreased in Tgfb2(-/-) embryos during valve remodeling. Collectively, the data indicate that TGFβ2 promotes valve remodeling and differentiation by inducing matrix organization and suppressing cushion mesenchyme differentiation into cartilage cell lineage during heart development.
Collapse
Affiliation(s)
- Mohamad Azhar
- BIO5 Institute, University of Arizona, Tucson, Arizona; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sewell-Loftin MK, Chun YW, Khademhosseini A, Merryman WD. EMT-inducing biomaterials for heart valve engineering: taking cues from developmental biology. J Cardiovasc Transl Res 2011; 4:658-71. [PMID: 21751069 DOI: 10.1007/s12265-011-9300-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 06/20/2011] [Indexed: 12/20/2022]
Abstract
Although artificial prostheses for diseased heart valves have been around for several decades, viable heart valve replacements have yet to be developed due to their complicated nature. The majority of research in heart valve replacement technology seeks to improve decellularization techniques for porcine valves or bovine pericardium as an effort to improve current clinically used valves. The drawback of clinically used valves is that they are nonviable and thus do not grow or remodel once implanted inside patients. This is particularly detrimental for pediatric patients, who will likely need several reoperations over the course of their lifetimes to implant larger valves as the patient grows. Due to this limitation, additional biomaterials, both synthetic and natural in origin, are also being investigated as novel scaffolds for tissue-engineered heart valves, specifically for the pediatric population. Here, we provide a brief overview of valves in clinical use as well as of the materials being investigated as novel tissue-engineered heart valve scaffolds. Additionally, we focus on natural-based biomaterials for promoting cell behavior that is indicative of the developmental biology process that occurs in the formation of heart valves in utero, such as epithelial-to-mesenchymal transition or transformation. By engineering materials that promote native developmental biology cues and signaling, while also providing mechanical integrity once implanted, a viable tissue-engineered heart valve may one day be realized. A viable tissue-engineered heart valve, capable of growing and remodeling actively inside a patient, could reduce risks and complications associated with current valve replacement options and improve overall quality of life in the thousands of patients who received such valves each year, particularly for children.
Collapse
Affiliation(s)
- M K Sewell-Loftin
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232-0493, USA
| | | | | | | |
Collapse
|