51
|
Lombardo VA, Heise M, Moghtadaei M, Bornhorst D, Männer J, Abdelilah-Seyfried S. Morphogenetic control of zebrafish cardiac looping by Bmp signaling. Development 2019; 146:dev.180091. [PMID: 31628109 DOI: 10.1242/dev.180091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/15/2019] [Indexed: 12/23/2022]
Abstract
Cardiac looping is an essential and highly conserved morphogenetic process that places the different regions of the developing vertebrate heart tube into proximity of their final topographical positions. High-resolution 4D live imaging of mosaically labelled cardiomyocytes reveals distinct cardiomyocyte behaviors that contribute to the deformation of the entire heart tube. Cardiomyocytes acquire a conical cell shape, which is most pronounced at the superior wall of the atrioventricular canal and contributes to S-shaped bending. Torsional deformation close to the outflow tract contributes to a torque-like winding of the entire heart tube between its two poles. Anisotropic growth of cardiomyocytes based on their positions reinforces S-shaping of the heart. During cardiac looping, bone morphogenetic protein pathway signaling is strongest at the future superior wall of the atrioventricular canal. Upon pharmacological or genetic inhibition of bone morphogenetic protein signaling, myocardial cells at the superior wall of the atrioventricular canal maintain cuboidal cell shapes and S-shaped bending is impaired. This description of cellular rearrangements and cardiac looping regulation may also be relevant for understanding the etiology of human congenital heart defects.
Collapse
Affiliation(s)
- Verónica A Lombardo
- Instituto de Biología Molecular y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas and Universidad Nacional de Rosario, 2000 Rosario, Argentina .,Centro de Estudios Interdisciplinarios, Universidad Nacional de Rosario, 2000 Rosario, Argentina
| | - Melina Heise
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany
| | - Motahareh Moghtadaei
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany.,Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| | - Dorothee Bornhorst
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany.,Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| | - Jörg Männer
- Institute of Anatomy and Embryology, UMG, Göttingen University, D-37075 Göttingen, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany .,Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| |
Collapse
|
52
|
Duchemin AL, Vignes H, Vermot J, Chow R. Mechanotransduction in cardiovascular morphogenesis and tissue engineering. Curr Opin Genet Dev 2019; 57:106-116. [PMID: 31586750 DOI: 10.1016/j.gde.2019.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/06/2019] [Accepted: 08/10/2019] [Indexed: 12/13/2022]
Abstract
Cardiovascular morphogenesis involves cell behavior and cell identity changes that are activated by mechanical forces associated with heart function. Recently, advances in in vivo imaging, methods to alter blood flow, and computational modelling have greatly advanced our understanding of how forces produced by heart contraction and blood flow impact different morphogenetic processes. Meanwhile, traditional genetic approaches have helped to elucidate how endothelial cells respond to forces at the cellular and molecular level. Here we discuss the principles of endothelial mechanosensitity and their interplay with cellular processes during cardiovascular morphogenesis. We then discuss their implications in the field of cardiovascular tissue engineering.
Collapse
Affiliation(s)
- Anne-Laure Duchemin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Helene Vignes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France.
| | - Renee Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, 67404 Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, 67404 Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, 67404 Illkirch, France; Université de Strasbourg, 67404 Illkirch, France
| |
Collapse
|
53
|
Defects in Trabecular Development Contribute to Left Ventricular Noncompaction. Pediatr Cardiol 2019; 40:1331-1338. [PMID: 31342111 DOI: 10.1007/s00246-019-02161-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
Left ventricular noncompaction (LVNC) is a genetically heterogeneous disorder the etiology of which is still debated. During fetal development, trabecular cardiomyocytes contribute extensively to the working myocardium and the ventricular conduction system. The impact of developmental defects in trabecular myocardium in the etiology of LVNC has been debated. Recently we generated new mouse models of LVNC by the conditional deletion of the key cardiac transcription factor encoding gene Nkx2-5 in trabecular myocardium at critical steps of trabecular development. These conditional mutant mice recapitulate pathological features similar to those observed in LVNC patients, including a hypertrabeculated left ventricle with deep endocardial recesses, subendocardial fibrosis, conduction defects, strain defects, and progressive heart failure. After discussing recent findings describing the respective contribution of trabecular and compact myocardium during ventricular morphogenesis, this review will focus on new data reflecting the link between trabecular development and LVNC.
Collapse
|
54
|
Biomechanical signaling within the developing zebrafish heart attunes endocardial growth to myocardial chamber dimensions. Nat Commun 2019; 10:4113. [PMID: 31511517 PMCID: PMC6739419 DOI: 10.1038/s41467-019-12068-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Intra-organ communication guides morphogenetic processes that are essential for an organ to carry out complex physiological functions. In the heart, the growth of the myocardium is tightly coupled to that of the endocardium, a specialized endothelial tissue that lines its interior. Several molecular pathways have been implicated in the communication between these tissues including secreted factors, components of the extracellular matrix, or proteins involved in cell-cell communication. Yet, it is unknown how the growth of the endocardium is coordinated with that of the myocardium. Here, we show that an increased expansion of the myocardial atrial chamber volume generates higher junctional forces within endocardial cells. This leads to biomechanical signaling involving VE-cadherin, triggering nuclear localization of the Hippo pathway transcriptional regulator Yap1 and endocardial proliferation. Our work suggests that the growth of the endocardium results from myocardial chamber volume expansion and ends when the tension on the tissue is relaxed. It is unknown how endocardium growth is coordinated with that of the myocardium in the zebrafish. Here, the authors show that myocardial chamber volume expansion causes increased endocardial tissue tension, which in turn triggers Hippo signaling-mediated proliferation within the endocardium.
Collapse
|
55
|
Mechanical Forces Regulate Cardiomyocyte Myofilament Maturation via the VCL-SSH1-CFL Axis. Dev Cell 2019; 51:62-77.e5. [PMID: 31495694 DOI: 10.1016/j.devcel.2019.08.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2019] [Accepted: 08/07/2019] [Indexed: 01/07/2023]
Abstract
Mechanical forces regulate cell behavior and tissue morphogenesis. During cardiac development, mechanical stimuli from the heartbeat are required for cardiomyocyte maturation, but the underlying molecular mechanisms remain unclear. Here, we first show that the forces of the contracting heart regulate the localization and activation of the cytoskeletal protein vinculin (VCL), which we find to be essential for myofilament maturation. To further analyze the role of VCL in this process, we examined its interactome in contracting versus non-contracting cardiomyocytes and, in addition to several known interactors, including actin regulators, identified the slingshot protein phosphatase SSH1. We show how VCL recruits SSH1 and its effector, the actin depolymerizing factor cofilin (CFL), to regulate F-actin rearrangement and promote cardiomyocyte myofilament maturation. Overall, our results reveal that mechanical forces generated by cardiac contractility regulate cardiomyocyte maturation through the VCL-SSH1-CFL axis, providing further insight into how mechanical forces are transmitted intracellularly to regulate myofilament maturation.
Collapse
|
56
|
Abstract
Cardiogenesis is a complex developmental process involving multiple overlapping stages of cell fate specification, proliferation, differentiation, and morphogenesis. Precise spatiotemporal coordination between the different cardiogenic processes is ensured by intercellular signalling crosstalk and tissue-tissue interactions. Notch is an intercellular signalling pathway crucial for cell fate decisions during multicellular organismal development and is aptly positioned to coordinate the complex signalling crosstalk required for progressive cell lineage restriction during cardiogenesis. In this Review, we describe the role of Notch signalling and the crosstalk with other signalling pathways during the differentiation and patterning of the different cardiac tissues and in cardiac valve and ventricular chamber development. We examine how perturbation of Notch signalling activity is linked to congenital heart diseases affecting the neonate and adult, and discuss studies that shed light on the role of Notch signalling in heart regeneration and repair after injury.
Collapse
|
57
|
Jaques C, Pignat E, Calinon S, Liebling M. Temporal super-resolution microscopy using a hue-encoded shutter. BIOMEDICAL OPTICS EXPRESS 2019; 10:4727-4741. [PMID: 31565521 PMCID: PMC6757482 DOI: 10.1364/boe.10.004727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 06/10/2023]
Abstract
Limited time-resolution in microscopy is an obstacle to many biological studies. Despite recent advances in hardware, digital cameras have limited operation modes that constrain frame-rate, integration time, and color sensing patterns. In this paper, we propose an approach to extend the temporal resolution of a conventional digital color camera by leveraging a multi-color illumination source. Our method allows for the imaging of single-hue objects at an increased frame-rate by trading spectral for temporal information (while retaining the ability to measure base hue). It also allows rapid switching to standard RGB acquisition. We evaluated the feasibility and performance of our method via experiments with mobile resolution targets. We observed a time-resolution increase by a factor 2.8 with a three-fold increase in temporal sampling rate. We further illustrate the use of our method to image the beating heart of a zebrafish larva, allowing the display of color or fast grayscale images. Our method is particularly well-suited to extend the capabilities of imaging systems where the flexibility of rapidly switching between high frame rate and color imaging are necessary.
Collapse
Affiliation(s)
- Christian Jaques
- Idiap Research Institute, Rue Marconi 19, 1920 Martigny, Switzerland
- École Polytechnique Fédérale de Lausanne, Switzerland
| | - Emmanuel Pignat
- Idiap Research Institute, Rue Marconi 19, 1920 Martigny, Switzerland
- École Polytechnique Fédérale de Lausanne, Switzerland
| | - Sylvain Calinon
- Idiap Research Institute, Rue Marconi 19, 1920 Martigny, Switzerland
- École Polytechnique Fédérale de Lausanne, Switzerland
| | - Michael Liebling
- Idiap Research Institute, Rue Marconi 19, 1920 Martigny, Switzerland
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
58
|
Foo YY, Pant S, Tay HS, Imangali N, Chen N, Winkler C, Yap CH. 4D modelling of fluid mechanics in the zebrafish embryonic heart. Biomech Model Mechanobiol 2019; 19:221-232. [PMID: 31446522 DOI: 10.1007/s10237-019-01205-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 07/20/2019] [Indexed: 11/30/2022]
Abstract
Abnormal blood flow mechanics can result in pathological heart malformation, underlining the importance of understanding embryonic cardiac fluid mechanics. In the current study, we performed image-based computational fluid dynamics simulation of the zebrafish embryonic heart ventricles and characterized flow mechanics, organ dynamics, and energy dynamics in detail. 4D scans of 5 days post-fertilization embryonic hearts with GFP-labelled myocardium were acquired using line-scan focal modulation microscopy. This revealed that the zebrafish hearts exhibited a wave-like contractile/relaxation motion from the inlet to the outlet during both systole and diastole, which we showed to be an energy efficient configuration. No impedance pumping effects of pressure and velocity waves were observed. Due to its tube-like configuration, inflow velocities were higher near the inlet and smaller at the outlet and vice versa for outflow velocities. This resulted in an interesting spatial wall shear stress (WSS) pattern where WSS waveforms near the inlet and those near the outlet were out of phase. There was large spatial variability in WSS magnitudes. Peak WSS was in the range of 47.5-130 dyne/cm2 at the inflow and outflow tracts, but were much smaller, in the range of 4-11 dyne/cm2, in the mid-ventricular segment. Due to very low Reynolds number and the highly viscous environment, intraventricular pressure gradients were high, suggesting substantial energy losses of flow through the heart.
Collapse
Affiliation(s)
- Yoke Yin Foo
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Shilpa Pant
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Huiping Shermaine Tay
- Department of Biological Science, National University of Singapore, Singapore, Singapore
| | - Nurgul Imangali
- Department of Biological Science, National University of Singapore, Singapore, Singapore
| | - Nanguang Chen
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Christoph Winkler
- Department of Biological Science, National University of Singapore, Singapore, Singapore
| | - Choon Hwai Yap
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
59
|
Cardiomyocyte orientation modulated by the Numb family proteins-N-cadherin axis is essential for ventricular wall morphogenesis. Proc Natl Acad Sci U S A 2019; 116:15560-15569. [PMID: 31300538 PMCID: PMC6681736 DOI: 10.1073/pnas.1904684116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The roles of cellular orientation during trabecular and ventricular wall morphogenesis are unknown, and so are the underlying mechanisms that regulate cellular orientation. Myocardial-specific Numb and Numblike double-knockout (MDKO) hearts display a variety of defects, including in cellular orientation, patterns of mitotic spindle orientation, trabeculation, and ventricular compaction. Furthermore, Numb- and Numblike-null cardiomyocytes exhibit cellular behaviors distinct from those of control cells during trabecular morphogenesis based on single-cell lineage tracing. We investigated how Numb regulates cellular orientation and behaviors and determined that N-cadherin levels and membrane localization are reduced in MDKO hearts. To determine how Numb regulates N-cadherin membrane localization, we generated an mCherry:Numb knockin line and found that Numb localized to diverse endocytic organelles but mainly to the recycling endosome. Consistent with this localization, cardiomyocytes in MDKO did not display defects in N-cadherin internalization but rather in postendocytic recycling to the plasma membrane. Furthermore, N-cadherin overexpression via a mosaic model partially rescued the defects in cellular orientation and trabeculation of MDKO hearts. Our study unravels a phenomenon that cardiomyocytes display spatiotemporal cellular orientation during ventricular wall morphogenesis, and its disruption leads to abnormal trabecular and ventricular wall morphogenesis. Furthermore, we established a mechanism by which Numb modulates cellular orientation and consequently trabecular and ventricular wall morphogenesis by regulating N-cadherin recycling to the plasma membrane.
Collapse
|
60
|
Cantu Gutierrez A, Cantu Gutierrez M, Rhyner AM, Ruiz OE, Eisenhoffer GT, Wythe JD. FishNET: An automated relational database for zebrafish colony management. PLoS Biol 2019; 17:e3000343. [PMID: 31220074 PMCID: PMC6605666 DOI: 10.1371/journal.pbio.3000343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 07/02/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022] Open
Abstract
The zebrafish Danio rerio is a powerful model system to study the genetics of development and disease. However, maintenance of zebrafish husbandry records is both time intensive and laborious, and a standardized way to manage and track the large amount of unique lines in a given laboratory or centralized facility has not been embraced by the field. Here, we present FishNET, an intuitive, open-source, relational database for managing data and information related to zebrafish husbandry and maintenance. By creating a "virtual facility," FishNET enables users to remotely inspect the rooms, racks, tanks, and lines within a given facility. Importantly, FishNET scales from one laboratory to an entire facility with several laboratories to multiple facilities, generating a cohesive laboratory and community-based platform. Automated data entry eliminates confusion regarding line nomenclature and streamlines maintenance of individual lines, while flexible query forms allow researchers to retrieve database records based on user-defined criteria. FishNET also links associated embryonic and adult biological samples with data, such as genotyping results or confocal images, to enable robust and efficient colony management and storage of laboratory information. A shared calendar function with email notifications and automated reminders for line turnover, automated tank counts, and census reports promote communication with both end users and administrators. The expected benefits of FishNET are improved vivaria efficiency, increased quality control for experimental numbers, and flexible data reporting and retrieval. FishNET's easy, intuitive record management and open-source, end-user-modifiable architecture provides an efficient solution to real-time zebrafish colony management for users throughout a facility and institution and, in some cases, across entire research hubs.
Collapse
Affiliation(s)
- Abiud Cantu Gutierrez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Manuel Cantu Gutierrez
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander M. Rhyner
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Oscar E. Ruiz
- Department of Genetics, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - George T. Eisenhoffer
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Genetics, MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Joshua D. Wythe
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
61
|
Abstract
Heart formation involves a complex series of tissue rearrangements, during which regions of the developing organ expand, bend, converge, and protrude in order to create the specific shapes of important cardiac components. Much of this morphogenesis takes place while cardiac function is underway, with blood flowing through the rapidly contracting chambers. Fluid forces are therefore likely to influence the regulation of cardiac morphogenesis, but it is not yet clear how these biomechanical cues direct specific cellular behaviors. In recent years, the optical accessibility and genetic amenability of zebrafish embryos have facilitated unique opportunities to integrate the analysis of flow parameters with the molecular and cellular dynamics underlying cardiogenesis. Consequently, we are making progress toward a comprehensive view of the biomechanical regulation of cardiac chamber emergence, atrioventricular canal differentiation, and ventricular trabeculation. In this review, we highlight a series of studies in zebrafish that have provided new insight into how cardiac function can shape cardiac morphology, with a particular focus on how hemodynamics can impact cardiac cell behavior. Over the long-term, this knowledge will undoubtedly guide our consideration of the potential causes of congenital heart disease.
Collapse
Affiliation(s)
- Pragya Sidhwani
- Division of Biological Sciences, University of California, San Diego, CA, United States
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, CA, United States.
| |
Collapse
|
62
|
Gardner LD, Peck KA, Goetz GW, Linbo TL, Cameron J, Scholz NL, Block BA, Incardona JP. Cardiac remodeling in response to embryonic crude oil exposure involves unconventional NKX family members and innate immunity genes. J Exp Biol 2019; 222:jeb.205567. [DOI: 10.1242/jeb.205567] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 10/04/2019] [Indexed: 01/08/2023]
Abstract
Cardiac remodeling results from both physiological and pathological stimuli. Compared to mammals, fish hearts show a broader array of remodeling changes in response to environmental influences, providing exceptional models for dissecting the molecular and cellular bases of cardiac remodeling. We recently characterized a form of pathological remodeling in juvenile pink salmon (Oncorhynchus gorbuscha) in response to crude oil exposure during embryonic cardiogenesis. In the absence of overt pathology (cardiomyocyte death or inflammatory infiltrate), cardiac ventricles in exposed fish showed altered shape, reduced thickness of compact myocardium, and hypertrophic changes in spongy, trabeculated myocardium. Here we used RNA sequencing to characterize molecular pathways underlying these defects. In juvenile ventricular cardiomyocytes, antecedent embryonic oil exposure led to dose-dependent up-regulation of genes involved in innate immunity and two NKX homeobox transcription factors not previously associated with cardiomyocytes, nkx2.3 and nkx3.3. Absent from mammalian genomes, the latter is largely uncharacterized. In zebrafish embryos nkx3.3 demonstrated a potent effect on cardiac morphogenesis, equivalent to nkx2.5, the primary transcription factor associated with ventricular cardiomyocyte identity. The role of nkx3.3 in heart growth is potentially linked to the unique regenerative capacity of fish and amphibians. Moreover, these findings support a cardiomyocyte-intrinsic role for innate immune response genes in pathological hypertrophy. This study demonstrates how an expanding mechanistic understanding of environmental pollution impacts – i.e., the chemical perturbation of biological systems – can ultimately yield new insights into fundamental biological processes.
Collapse
Affiliation(s)
- Luke D. Gardner
- Hopkins Marine Station, Department of Biology, Stanford University, Pacific Grove, CA 93950, USA
| | - Karen A. Peck
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| | - Giles W. Goetz
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| | - Tiffany L. Linbo
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| | - James Cameron
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| | - Nathaniel L. Scholz
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| | - Barbara A. Block
- Hopkins Marine Station, Department of Biology, Stanford University, Pacific Grove, CA 93950, USA
| | - John P. Incardona
- Environmental and Fisheries Sciences Division, Northwest Fisheries Science Center, National Oceanic and Atmospheric Administration, 2725 Montlake Blvd. E., Seattle, WA 98112, USA
| |
Collapse
|
63
|
Jiménez-Amilburu V, Stainier DYR. The transmembrane protein Crb2a regulates cardiomyocyte apicobasal polarity and adhesion in zebrafish. Development 2019; 146:dev.171207. [DOI: 10.1242/dev.171207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Tissue morphogenesis requires changes in cell-cell adhesion as well as in cell shape and polarity. Cardiac trabeculation is a morphogenetic process essential to form a functional ventricular wall. Here we show that zebrafish hearts lacking Crb2a, a component of the Crumbs polarity complex, display compact wall integrity defects and fail to form trabeculae. Crb2a localization is very dynamic at a time when other cardiomyocyte junctional proteins also relocalize. Before the initiation of cardiomyocyte delamination to form the trabecular layer, Crb2a is expressed in all ventricular cardiomyocytes and colocalizes with the junctional protein ZO-1. Subsequently, Crb2a becomes localized all along the apical membrane of compact layer cardiomyocytes and is downregulated in the delaminating cardiomyocytes. We show that blood flow and Nrg/ErbB2 signaling regulate Crb2a localization dynamics. crb2a−/− display a multilayered wall with polarized cardiomyocytes, a unique phenotype. Our data further indicate that Crb2a regulates cardiac trabeculation by controlling the localization of tight and adherens junction proteins in cardiomyocytes. Importantly, transplantation data show that Crb2a controls CM behavior in a cell-autonomous manner in the sense that crb2a−/− cardiomyocytes transplanted into wild-type animals were always found in the trabecular layer. Altogether, our study reveals a critical role for Crb2a during cardiac development.
Collapse
Affiliation(s)
- Vanesa Jiménez-Amilburu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
64
|
Rasouli SJ, El-Brolosy M, Tsedeke AT, Bensimon-Brito A, Ghanbari P, Maischein HM, Kuenne C, Stainier DY. The flow responsive transcription factor Klf2 is required for myocardial wall integrity by modulating Fgf signaling. eLife 2018; 7:e38889. [PMID: 30592462 PMCID: PMC6329608 DOI: 10.7554/elife.38889] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Complex interplay between cardiac tissues is crucial for their integrity. The flow responsive transcription factor KLF2, which is expressed in the endocardium, is vital for cardiovascular development but its exact role remains to be defined. To this end, we mutated both klf2 paralogues in zebrafish, and while single mutants exhibit no obvious phenotype, double mutants display a novel phenotype of cardiomyocyte extrusion towards the abluminal side. This extrusion requires cardiac contractility and correlates with the mislocalization of N-cadherin from the lateral to the apical side of cardiomyocytes. Transgenic rescue data show that klf2 expression in endothelium, but not myocardium, prevents this cardiomyocyte extrusion phenotype. Transcriptome analysis of klf2 mutant hearts reveals that Fgf signaling is affected, and accordingly, we find that inhibition of Fgf signaling in wild-type animals can lead to abluminal cardiomyocyte extrusion. These studies provide new insights into how Klf2 regulates cardiovascular development and specifically myocardial wall integrity.
Collapse
Affiliation(s)
- Seyed Javad Rasouli
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Mohamed El-Brolosy
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Ayele Taddese Tsedeke
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Anabela Bensimon-Brito
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Parisa Ghanbari
- Department of Cardiac Development and RemodelingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Hans-Martin Maischein
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Carsten Kuenne
- Bioinformatics Core UnitMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Didier Y Stainier
- Department of Developmental GeneticsMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| |
Collapse
|
65
|
Tekeli I, Garcia-Puig A, Notari M, García-Pastor C, Aujard I, Jullien L, Raya A. Fate predetermination of cardiac myocytes during zebrafish heart regeneration. Open Biol 2018; 7:rsob.170116. [PMID: 28659386 PMCID: PMC5493782 DOI: 10.1098/rsob.170116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Adult zebrafish have the remarkable ability to regenerate their heart upon injury, a process that involves limited dedifferentiation and proliferation of spared cardiomyocytes (CMs), and migration of their progeny. During regeneration, proliferating CMs are detected throughout the myocardium, including areas distant to the injury site, but whether all of them are able to contribute to the regenerated tissue remains unknown. Here, we developed a CM-specific, photoinducible genetic labelling system, and show that CMs labelled in embryonic hearts survive and contribute to all three (primordial, trabecular and cortical) layers of the adult zebrafish heart. Next, using this system to investigate the fate of CMs from different parts of the myocardium during regeneration, we show that only CMs immediately adjacent to the injury site contributed to the regenerated tissue. Finally, our results show an extensive predetermination of CM fate during adult heart regeneration, with cells from each myocardial layer giving rise to cells that retain their layer identity in the regenerated myocardium. Overall, our results indicate that adult heart regeneration in the zebrafish is a rather static process governed by short-range signals, in contrast to the highly dynamic plasticity of CM fates that takes place during embryonic heart regeneration.
Collapse
Affiliation(s)
- Isil Tekeli
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), 3rd Floor, Avinguda de la Gran Via 199-203, 08908 Barcelona, Spain
| | - Anna Garcia-Puig
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), 3rd Floor, Avinguda de la Gran Via 199-203, 08908 Barcelona, Spain
| | - Mario Notari
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), 3rd Floor, Avinguda de la Gran Via 199-203, 08908 Barcelona, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Cristina García-Pastor
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), 3rd Floor, Avinguda de la Gran Via 199-203, 08908 Barcelona, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Isabelle Aujard
- Department of Chemistry, École Normale Supérieure-PSL Research University, 24 rue Lhomond, 75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, PASTEUR, 75005 Paris, France.,CNRS, UMR 8640 PASTEUR, 75005 Paris, France
| | - Ludovic Jullien
- Department of Chemistry, École Normale Supérieure-PSL Research University, 24 rue Lhomond, 75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, PASTEUR, 75005 Paris, France.,CNRS, UMR 8640 PASTEUR, 75005 Paris, France
| | - Angel Raya
- Center of Regenerative Medicine in Barcelona (CMRB), Hospital Duran i Reynals, Hospitalet de Llobregat (Barcelona), 3rd Floor, Avinguda de la Gran Via 199-203, 08908 Barcelona, Spain .,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
66
|
Ding Y, Ma J, Langenbacher AD, Baek KI, Lee J, Chang CC, Hsu JJ, Kulkarni RP, Belperio J, Shi W, Ranjbarvaziri S, Ardehali R, Tintut Y, Demer LL, Chen JN, Fei P, Packard RRS, Hsiai TK. Multiscale light-sheet for rapid imaging of cardiopulmonary system. JCI Insight 2018; 3:e121396. [PMID: 30135307 PMCID: PMC6141183 DOI: 10.1172/jci.insight.121396] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The ability to image tissue morphogenesis in real-time and in 3-dimensions (3-D) remains an optical challenge. The advent of light-sheet fluorescence microscopy (LSFM) has advanced developmental biology and tissue regeneration research. In this review, we introduce a LSFM system in which the illumination lens reshapes a thin light-sheet to rapidly scan across a sample of interest while the detection lens orthogonally collects the imaging data. This multiscale strategy provides deep-tissue penetration, high-spatiotemporal resolution, and minimal photobleaching and phototoxicity, allowing in vivo visualization of a variety of tissues and processes, ranging from developing hearts in live zebrafish embryos to ex vivo interrogation of the microarchitecture of optically cleared neonatal hearts. Here, we highlight multiple applications of LSFM and discuss several studies that have allowed better characterization of developmental and pathological processes in multiple models and tissues. These findings demonstrate the capacity of multiscale light-sheet imaging to uncover cardiovascular developmental and regenerative phenomena.
Collapse
Affiliation(s)
- Yichen Ding
- Department of Medicine, David Geffen School of Medicine at UCLA, and
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | - Jianguo Ma
- Department of Medicine, David Geffen School of Medicine at UCLA, and
- School of Instrumentation Science and Opto-electronics Engineering, Beihang University, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beijing, China
| | - Adam D. Langenbacher
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, California, USA
| | - Kyung In Baek
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | - Juhyun Lee
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| | | | - Jeffrey J. Hsu
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - Rajan P. Kulkarni
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - John Belperio
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - Wei Shi
- Developmental Biology and Regenerative Medicine Program, Department of Surgery, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Reza Ardehali
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - Yin Tintut
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - Linda L. Demer
- Department of Medicine, David Geffen School of Medicine at UCLA, and
| | - Jau-Nian Chen
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, California, USA
| | - Peng Fei
- Department of Medicine, David Geffen School of Medicine at UCLA, and
- School of Optical and Electronic Information, Huazhong University of Science and Technology, Wuhan, China
| | | | - Tzung K. Hsiai
- Department of Medicine, David Geffen School of Medicine at UCLA, and
- Department of Bioengineering, UCLA, Los Angeles, California, USA
| |
Collapse
|
67
|
Wu RS, Lam II, Clay H, Duong DN, Deo RC, Coughlin SR. A Rapid Method for Directed Gene Knockout for Screening in G0 Zebrafish. Dev Cell 2018; 46:112-125.e4. [PMID: 29974860 DOI: 10.1016/j.devcel.2018.06.003] [Citation(s) in RCA: 255] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/31/2022]
Abstract
Zebrafish is a powerful model for forward genetics. Reverse genetic approaches are limited by the time required to generate stable mutant lines. We describe a system for gene knockout that consistently produces null phenotypes in G0 zebrafish. Yolk injection of sets of four CRISPR/Cas9 ribonucleoprotein complexes redundantly targeting a single gene recapitulated germline-transmitted knockout phenotypes in >90% of G0 embryos for each of 8 test genes. Early embryonic (6 hpf) and stable adult phenotypes were produced. Simultaneous multi-gene knockout was feasible but associated with toxicity in some cases. To facilitate use, we generated a lookup table of four-guide sets for 21,386 zebrafish genes and validated several. Using this resource, we targeted 50 cardiomyocyte transcriptional regulators and uncovered a role of zbtb16a in cardiac development. This system provides a platform for rapid screening of genes of interest in development, physiology, and disease models in zebrafish.
Collapse
Affiliation(s)
- Roland S Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ian I Lam
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hilary Clay
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel N Duong
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Rahul C Deo
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shaun R Coughlin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
68
|
Wu M. Mechanisms of Trabecular Formation and Specification During Cardiogenesis. Pediatr Cardiol 2018; 39:1082-1089. [PMID: 29594501 PMCID: PMC6164162 DOI: 10.1007/s00246-018-1868-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/14/2018] [Indexed: 01/08/2023]
Abstract
Trabecular morphogenesis is a key morphologic event during cardiogenesis and contributes to the formation of a competent ventricular wall. Lack of trabeculation results in embryonic lethality. The trabecular morphogenesis is a multistep process that includes, but is not limited to, trabecular initiation, proliferation/growth, specification, and compaction. Although a number of signaling molecules have been implicated in regulating trabeculation, the cellular processes underlying mammalian trabecular formation are not fully understood. Recent works show that the myocardium displays polarity, and oriented cell division (OCD) and directional migration of the cardiomyocytes in the monolayer myocardium are required for trabecular initiation and formation. Furthermore, perpendicular OCD is an extrinsic asymmetric cell division that contributes to trabecular specification, and is a mechanism that causes the trabecular cardiomyocytes to be distinct from the cardiomyocytes in compact zone. Once the coronary vasculature system starts to function in the embryonic heart, the trabeculae will coalesce with the compact zone to thicken the heart wall, and abnormal compaction will lead to left ventricular non-compaction (LVNC) and heart failure. There are many reviews about compaction and LVNC. In this review, we will focus on the roles of myocardial polarity and OCD in trabecular initiation, formation, and specification.
Collapse
Affiliation(s)
- Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, 43 New Scotland Ave, Albany, NY, 12208, USA.
| |
Collapse
|
69
|
Uribe V, Ramadass R, Dogra D, Rasouli SJ, Gunawan F, Nakajima H, Chiba A, Reischauer S, Mochizuki N, Stainier DYR. In vivo analysis of cardiomyocyte proliferation during trabeculation. Development 2018; 145:145/14/dev164194. [PMID: 30061167 DOI: 10.1242/dev.164194] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/16/2018] [Indexed: 12/18/2022]
Abstract
Cardiomyocyte proliferation is crucial for cardiac growth, patterning and regeneration; however, few studies have investigated the behavior of dividing cardiomyocytes in vivo Here, we use time-lapse imaging of beating hearts in combination with the FUCCI system to monitor the behavior of proliferating cardiomyocytes in developing zebrafish. Confirming in vitro observations, sarcomere disassembly, as well as changes in cell shape and volume, precede cardiomyocyte cytokinesis. Notably, cardiomyocytes in zebrafish embryos and young larvae mostly divide parallel to the myocardial wall in both the compact and trabecular layers, and cardiomyocyte proliferation is more frequent in the trabecular layer. While analyzing known regulators of cardiomyocyte proliferation, we observed that the Nrg/ErbB2 and TGFβ signaling pathways differentially affect compact and trabecular layer cardiomyocytes, indicating that distinct mechanisms drive proliferation in these two layers. In summary, our data indicate that, in zebrafish, cardiomyocyte proliferation is essential for trabecular growth, but not initiation, and set the stage to further investigate the cellular and molecular mechanisms driving cardiomyocyte proliferation in vivo.
Collapse
Affiliation(s)
- Veronica Uribe
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Radhan Ramadass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Deepika Dogra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - S Javad Rasouli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
70
|
Rapamycin attenuates pathological hypertrophy caused by an absence of trabecular formation. Sci Rep 2018; 8:8584. [PMID: 29872120 PMCID: PMC5988815 DOI: 10.1038/s41598-018-26843-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/15/2018] [Indexed: 12/29/2022] Open
Abstract
Cardiac trabeculae are mesh-like muscular structures within ventricular walls. Subtle perturbations in trabeculation are associated with many congenital heart diseases (CHDs), and complete failure to form trabeculae leads to embryonic lethality. Despite the severe consequence of an absence of trabecular formation, the exact function of trabeculae remains unclear. Since ErbB2 signaling plays a direct and essential role in trabecular initiation, in this study, we utilized the erbb2 zebrafish mutant as a model to address the function of trabeculae in the heart. Intriguingly, we found that the trabeculae-deficient erbb2 mutant develops a hypertrophic-like (HL) phenotype that can be suppressed by inhibition of Target of Rapamycin (TOR) signaling in a similar fashion to adult mammalian hearts subjected to mechanical overload. Further, cell transplantation experiments demonstrated that erbb2 mutant cells in an otherwise wildtype heart did not undergo hypertrophy, indicating that erbb2 mutant HL phenotypes are due to a loss of trabeculae. Together, we propose that trabeculae serve to enhance contractility and that defects in this process lead to wall-stress induced hypertrophic remodeling.
Collapse
|
71
|
Lai JKH, Collins MM, Uribe V, Jiménez-Amilburu V, Günther S, Maischein HM, Stainier DYR. The Hippo pathway effector Wwtr1 regulates cardiac wall maturation in zebrafish. Development 2018; 145:145/10/dev159210. [PMID: 29773645 DOI: 10.1242/dev.159210] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/23/2018] [Indexed: 12/14/2022]
Abstract
Cardiac trabeculation is a highly regulated process that starts with the delamination of compact layer cardiomyocytes. The Hippo signaling pathway has been implicated in cardiac development but many questions remain. We have investigated the role of Wwtr1, a nuclear effector of the Hippo pathway, in zebrafish and find that its loss leads to reduced cardiac trabeculation. However, in mosaic animals, wwtr1-/- cardiomyocytes contribute more frequently than wwtr1+/- cardiomyocytes to the trabecular layer of wild-type hearts. To investigate this paradox, we examined the myocardial wall at early stages and found that compact layer cardiomyocytes in wwtr1-/- hearts exhibit disorganized cortical actin structure and abnormal cell-cell junctions. Accordingly, wild-type cardiomyocytes in mosaic mutant hearts contribute less frequently to the trabecular layer than when present in mosaic wild-type hearts, indicating that wwtr1-/- hearts are not able to support trabeculation. We also found that Nrg/Erbb2 signaling, which is required for trabeculation, could promote Wwtr1 nuclear export in cardiomyocytes. Altogether, these data suggest that Wwtr1 establishes the compact wall architecture necessary for trabeculation, and that Nrg/Erbb2 signaling negatively regulates its nuclear localization and therefore its activity.
Collapse
Affiliation(s)
- Jason K H Lai
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Michelle M Collins
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Veronica Uribe
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Vanesa Jiménez-Amilburu
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, ECCPS Bioinformatics and Deep Sequencing Platform, Bad Nauheim 61231, Germany
| | - Hans-Martin Maischein
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| |
Collapse
|
72
|
Del Monte-Nieto G, Ramialison M, Adam AAS, Wu B, Aharonov A, D'Uva G, Bourke LM, Pitulescu ME, Chen H, de la Pompa JL, Shou W, Adams RH, Harten SK, Tzahor E, Zhou B, Harvey RP. Control of cardiac jelly dynamics by NOTCH1 and NRG1 defines the building plan for trabeculation. Nature 2018; 557:439-445. [PMID: 29743679 DOI: 10.1038/s41586-018-0110-6] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 03/26/2018] [Indexed: 12/20/2022]
Abstract
In vertebrate hearts, the ventricular trabecular myocardium develops as a sponge-like network of cardiomyocytes that is critical for contraction and conduction, ventricular septation, papillary muscle formation and wall thickening through the process of compaction 1 . Defective trabeculation leads to embryonic lethality2-4 or non-compaction cardiomyopathy (NCC) 5 . There are divergent views on when and how trabeculation is initiated in different species. In zebrafish, trabecular cardiomyocytes extrude from compact myocardium 6 , whereas in chicks, chamber wall thickening occurs before overt trabeculation 7 . In mice, the onset of trabeculation has not been described, but is proposed to begin at embryonic day 9.0, when cardiomyocytes form radially oriented ribs 2 . Endocardium-myocardium communication is essential for trabeculation, and numerous signalling pathways have been identified, including Notch2,8 and Neuregulin (NRG) 4 . Late disruption of the Notch pathway causes NCC 5 . Whereas it has been shown that mutations in the extracellular matrix (ECM) genes Has2 and Vcan prevent the formation of trabeculae in mice9,10 and the matrix metalloprotease ADAMTS1 promotes trabecular termination 3 , the pathways involved in ECM dynamics and the molecular regulation of trabeculation during its early phases remain unexplored. Here we present a model of trabeculation in mice that integrates dynamic endocardial and myocardial cell behaviours and ECM remodelling, and reveal new epistatic relationships between the involved signalling pathways. NOTCH1 signalling promotes ECM degradation during the formation of endocardial projections that are critical for individualization of trabecular units, whereas NRG1 promotes myocardial ECM synthesis, which is necessary for trabecular rearrangement and growth. These systems interconnect through NRG1 control of Vegfa, but act antagonistically to establish trabecular architecture. These insights enabled the prediction of persistent ECM and cardiomyocyte growth in a mouse NCC model, providing new insights into the pathophysiology of congenital heart disease.
Collapse
Affiliation(s)
- Gonzalo Del Monte-Nieto
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia. .,St Vincent's Clinical School, University of New South Wales, Kensington, New South Wales, Australia.
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Arne A S Adam
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Bingruo Wu
- Departments of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, NY, USA
| | - Alla Aharonov
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Gabriele D'Uva
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lauren M Bourke
- Epigenetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Hanying Chen
- Departments of Pediatrics and Medical and Molecular Genetics, Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Ciber cardiovascular, ISCIII, Madrid, Spain
| | - Weinian Shou
- Departments of Pediatrics and Medical and Molecular Genetics, Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University, Indianapolis, IN, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Sarah K Harten
- Epigenetics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Bin Zhou
- Departments of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, NY, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia. .,St Vincent's Clinical School, University of New South Wales, Kensington, New South Wales, Australia. .,School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales, Australia.
| |
Collapse
|
73
|
Huang M, Jiao J, Wang J, Xia Z, Zhang Y. Exposure to acrylamide induces cardiac developmental toxicity in zebrafish during cardiogenesis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 234:656-666. [PMID: 29223822 DOI: 10.1016/j.envpol.2017.11.095] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 11/25/2017] [Accepted: 11/28/2017] [Indexed: 06/07/2023]
Abstract
Acrylamide (AA), an environmental pollutant, has been linked to neurotoxicity, genotoxicity and carcinogenicity. AA is widely used to synthesize polymers for industrial applications, is widely found in Western-style carbohydrate-rich foods and cigarette smoke, and can also be detected in human umbilical cord blood and breast milk. This is the first study that demonstrated the cardiac developmental toxicity of AA in zebrafish embryos. Post-fertilization exposure to AA caused a clearly deficient cardiovascular system with a shrunken heart and abortive morphogenesis and function. Disordered expression of the cardiac genes, myl7, vmhc, myh6, bmp4, tbx2b and notch1b, as well as reduced number of myocardial cells and endocardial cells, indicated the collapsed development of ventricle and atrium and failed differentiation of atrioventricular canal (AVC). Although cell apoptosis was not affected, the capacity of cardiomyocyte proliferation was significantly reduced by AA exposure after fertilization. Further investigation showed that treatment with AA specifically reduced the expressions of nkx2.5, myl7 and vmhc in the anterior lateral plate mesoderm (ALPM) during the early cardiogenesis. In addition, AA exposure disturbed the restricted expressions of bmp4, tbx2b and notch1b during atrioventricular (AV) valve development and cardiac chambers maturation. Our results showed that AA-induced cardiotoxicity was related to decreased cardiac progenitor genes expression, reduced myocardium growth, abnormal cardiac chambers morphogenesis and disordered AVC differentiation. Our study demonstrates that AA exposure during a time point analogous to the first trimester in humans has a detrimental effect on early heart development in zebrafish. A high ingestion rate of AA-containing products may be an underlying risk factor for cardiogenesis in fetuses.
Collapse
Affiliation(s)
- Mengmeng Huang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Jingjing Jiao
- Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jun Wang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zhidan Xia
- Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yu Zhang
- National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
74
|
Paun B, Bijnens B, Butakoff C. Relationship between the left ventricular size and the amount of trabeculations. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2018; 34:e2939. [PMID: 29124903 DOI: 10.1002/cnm.2939] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 06/07/2023]
Abstract
Contemporary imaging modalities offer noninvasive quantification of myocardial deformation; however, they make gross assumptions about internal structure of the cardiac walls. Our aim is to study the possible impact of the trabeculations on the stroke volume, strain, and capacity of differently sized ventricles. The cardiac left ventricle is represented by an ellipsoid and the trabeculations by a tissue occupying a fixed volume. The ventricular contraction is modeled by scaling the ellipsoid whereupon the measurements of longitudinal strain, end-diastolic, end-systolic, and stroke volumes are derived and compared. When the trabeculated and nontrabeculated ventricles, having the same geometry and deformation pattern, contain the same amount of blood and contract with the same strain, we observed an increased stroke volume in our model of the trabeculated ventricle. When these ventricles contain and eject the same amount of blood, we observed a reduced strain in the trabeculated case. We identified that a trade-off between the strain and the amount of trabeculations could be reached with a 0.35- to 0.41-cm dense trabeculated layer, without blood filled recesses (for a ventricle with end-diastolic volume of about 150 mL). A trabeculated ventricle can work at lower strains compared to a nontrabeculated ventricle to produce the same stroke volume, which could be a possible explanation why athletes and pregnant women develop reversible signs of left ventricular noncompaction, since the trabeculations could help generating extra cardiac output. This knowledge might help to assess heart failure patients with dilated cardiomyopathies who often show signs of noncompaction.
Collapse
Affiliation(s)
- Bruno Paun
- PhySense, Universitat Pompeu Fabra, Barcelona, Spain
| | - Bart Bijnens
- PhySense, Universitat Pompeu Fabra, Barcelona, Spain
- ICREA, Barcelona, Spain
- KU Leuven, Leuven, Belgium
| | | |
Collapse
|
75
|
Pasiliao CC, Hopyan S. Cell ingression: Relevance to limb development and for adaptive evolution. Genesis 2017; 56. [PMID: 29280270 DOI: 10.1002/dvg.23086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 11/16/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
Cell ingression is an out-of-plane type of cell intercalation that is essential for the formation of multiple embryonic structures including the limbs. In particular, cell ingression underlies epithelial-to-mesenchymal transition of lateral plate cells to initiate limb bud growth, delamination of neural crest cells to generate peripheral nerve sheaths, and emigration of myoblasts from somites to assemble muscles. Individual cells that ingress undergo apical constriction to generate bottle shaped cells, diminish adhesion to their epithelial cell neighbors, and generate protrusive blebs that likely facilitate their ingression into a subepithelial tissue layer. How signaling pathways regulate the progression of delamination is important for understanding numerous developmental events. In this review, we focus on cellular and molecular mechanisms that drive cell ingression and draw comparisons between different morphogenetic contexts in various model organisms. We speculate that cell behaviors that facilitated tissue invagination among diploblasts subsequently drove individual cell ingression and epithelial-to-mesenchymal transition. Future insights that link signalling pathways to biophysical mechanisms will likely advance our comprehension of this phenomenon.
Collapse
Affiliation(s)
- Clarissa C Pasiliao
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, M5S 1A8, Canada
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, M5S 1A8, Canada.,Division of Orthopaedic Surgery, Hospital for Sick Children and University of, Toronto, M5G 1X8, Canada
| |
Collapse
|
76
|
Jiménez-Amilburu V, Rasouli SJ, Staudt DW, Nakajima H, Chiba A, Mochizuki N, Stainier DYR. In Vivo Visualization of Cardiomyocyte Apicobasal Polarity Reveals Epithelial to Mesenchymal-like Transition during Cardiac Trabeculation. Cell Rep 2017; 17:2687-2699. [PMID: 27926871 DOI: 10.1016/j.celrep.2016.11.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/05/2016] [Accepted: 11/03/2016] [Indexed: 12/22/2022] Open
Abstract
Despite great strides in understanding cardiac trabeculation, many mechanistic aspects remain unclear. To elucidate how cardiomyocyte shape changes are regulated during this process, we engineered transgenes to label their apical and basolateral membranes. Using these tools, we observed that compact-layer cardiomyocytes are clearly polarized while delaminating cardiomyocytes have lost their polarity. The apical transgene also enabled the imaging of cardiomyocyte apical constriction in real time. Furthermore, we found that Neuregulin signaling and blood flow/cardiac contractility are required for cardiomyocyte apical constriction and depolarization. Notably, we observed the activation of Notch signaling in cardiomyocytes adjacent to those undergoing apical constriction, and we showed that this activation is positively regulated by Neuregulin signaling. Inhibition of Notch signaling did not increase the percentage of cardiomyocytes undergoing apical constriction or of trabecular cardiomyocytes. These studies provide information about cardiomyocyte polarization and enhance our understanding of the complex mechanisms underlying ventricular morphogenesis and maturation.
Collapse
Affiliation(s)
- Vanesa Jiménez-Amilburu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - S Javad Rasouli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - David W Staudt
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
77
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
78
|
Rasouli SJ, Stainier DYR. Regulation of cardiomyocyte behavior in zebrafish trabeculation by Neuregulin 2a signaling. Nat Commun 2017; 8:15281. [PMID: 28485381 PMCID: PMC5477525 DOI: 10.1038/ncomms15281] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/15/2017] [Indexed: 12/16/2022] Open
Abstract
Trabeculation is crucial for cardiac muscle growth in vertebrates. This process requires the Erbb2/4 ligand Neuregulin (Nrg), secreted by the endocardium, as well as blood flow/cardiac contractility. Here, we address two fundamental, yet unresolved, questions about cardiac trabeculation: why does it initially occur in the ventricle and not the atrium, and how is it modulated by blood flow/contractility. Using loss-of-function approaches, we first show that zebrafish Nrg2a is required for trabeculation, and using a protein-trap line, find that it is expressed in both cardiac chambers albeit with different spatiotemporal patterns. Through gain-of-function experiments, we show that atrial cardiomyocytes can also respond to Nrg2a signalling, suggesting that the cardiac jelly, which remains prominent in the atrium, represents a barrier to Erbb2/4 activation. Furthermore, we find that blood flow/contractility is required for Nrg2a expression, and that while non-contractile hearts fail to trabeculate, non-contractile cardiomyocytes are also competent to respond to Nrg2a/Erbb2 signalling. Cardiac trabeculae (which are sponge-like muscular structures) form mostly as a result of cardiomyocyte (CM) delamination in zebrafish. Here, the authors identify Nrg2a in zebrafish as a key regulator of trabeculation, and atrial and non-contractile CMs also respond to Nrg2a despite not forming trabeculae.
Collapse
Affiliation(s)
- S Javad Rasouli
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
79
|
Schmutz S, Valente M, Cumano A, Novault S. Analysis of Cell Suspensions Isolated from Solid Tissues by Spectral Flow Cytometry. J Vis Exp 2017. [PMID: 28518119 DOI: 10.3791/55578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Flow cytometry has been used for the past 40 years to define and analyze the phenotype of lymphoid and other hematopoietic cells. Initially restricted to the analysis of a few fluorochromes, currently there are dozens of different fluorescent dyes, and up to 14-18 different dyes can be combined at a time. However, several limitations still impair the analytical capabilities. Because of the multiplicity of fluorescent probes, data analysis has become increasingly complex due to the need of large, multi-parametric compensation matrices. Moreover, mutant mouse models carrying fluorescent proteins to detect and trace specific cell types in different tissues have become available, so the analysis (by flow cytometry) of auto-fluorescent cell suspensions obtained from solid organs is required. Spectral flow cytometry, which distinguishes the shapes of emission spectra along a wide range of continuous wavelengths, addresses some of these problems. The data is analyzed with an algorithm that replaces compensation matrices and treats auto-fluorescence as an independent parameter. Thus, spectral flow cytometry should be capable of discriminating fluorochromes with similar emission peaks and can provide a multi-parametric analysis without compensation requirements. This protocol describes the spectral flow cytometry analysis, allowing for a 21-parameter (19 fluorescent probes) characterization and the management of an auto-fluorescent signal, providing high resolution in minor population detection. The results presented here show that spectral flow cytometry presents advantages in the analysis of cell populations from tissues difficult to characterize in conventional flow cytometry, such as the heart and the intestine. Spectral flow cytometry thus demonstrates the multi-parametric analytical capacity of high-performing conventional flow cytometry without the requirement for compensation and enables auto-fluorescence management.
Collapse
Affiliation(s)
- Sandrine Schmutz
- Flow Cytometry Core Facility, Center for Translational Research-Technical Core, Institut Pasteur
| | - Mariana Valente
- Unit for Lymphopoiesis, Immunology Department, INSERM U1223, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur; Stem-Cell Microenvironments in Repair/Regeneration Team, Instituto de Investigação e Inovação em Saúde (i3s), INEB - Instituto de Engenharia Biomédica; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto; Stem Cells and Regenerative Medicine Team, UMRS 1166, ICAN - Institute of Cardiometabolism And Nutrition, UPMC - Université Pierre et Marie Curie - Paris 6, INSERM
| | - Ana Cumano
- Unit for Lymphopoiesis, Immunology Department, INSERM U1223, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur;
| | - Sophie Novault
- Flow Cytometry Core Facility, Center for Translational Research-Technical Core, Institut Pasteur;
| |
Collapse
|
80
|
Galdos FX, Guo Y, Paige SL, VanDusen NJ, Wu SM, Pu WT. Cardiac Regeneration: Lessons From Development. Circ Res 2017; 120:941-959. [PMID: 28302741 DOI: 10.1161/circresaha.116.309040] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/14/2016] [Accepted: 12/15/2016] [Indexed: 02/06/2023]
Abstract
Palliative surgery for congenital heart disease has allowed patients with previously lethal heart malformations to survive and, in most cases, to thrive. However, these procedures often place pressure and volume loads on the heart, and over time, these chronic loads can cause heart failure. Current therapeutic options for initial surgery and chronic heart failure that results from failed palliation are limited, in part, by the mammalian heart's low inherent capacity to form new cardiomyocytes. Surmounting the heart regeneration barrier would transform the treatment of congenital, as well as acquired, heart disease and likewise would enable development of personalized, in vitro cardiac disease models. Although these remain distant goals, studies of heart development are illuminating the path forward and suggest unique opportunities for heart regeneration, particularly in fetal and neonatal periods. Here, we review major lessons from heart development that inform current and future studies directed at enhancing cardiac regeneration.
Collapse
Affiliation(s)
- Francisco X Galdos
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Yuxuan Guo
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sharon L Paige
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Nathan J VanDusen
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.)
| | - Sean M Wu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| | - William T Pu
- From the Cardiovascular Institute, School of Medicine, Stanford University, CA (F.X.G., S.L.P., S.M.W.); Department of Cardiology, Boston Children's Hospital, MA (Y.G., N.J.V., W.T.P.); Division of Pediatric Cardiology, Department of Pediatrics (S.L.P.), Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), and Institute of Stem Cell and Regenerative Biology, School of Medicine, Stanford, CA (F.X.G., S.L.P., S.M.W.); and Harvard Stem Cell Institute, Harvard University, Cambridge, MA (W.T.P.).
| |
Collapse
|
81
|
Schmutz S, Valente M, Cumano A, Novault S. Spectral Cytometry Has Unique Properties Allowing Multicolor Analysis of Cell Suspensions Isolated from Solid Tissues. PLoS One 2016; 11:e0159961. [PMID: 27500930 PMCID: PMC4976887 DOI: 10.1371/journal.pone.0159961] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/19/2016] [Indexed: 12/17/2022] Open
Abstract
Flow cytometry, initially developed to analyze surface protein expression in hematopoietic cells, has increased in analytical complexity and is now widely used to identify cells from different tissues and organisms. As a consequence, data analysis became increasingly difficult due the need of large multi-parametric compensation matrices and to the eventual auto-fluorescence frequently found in cell suspensions obtained from solid organs. In contrast with conventional flow cytometry that detects the emission peak of fluorochromes, spectral flow cytometry distinguishes the shapes of emission spectra along a large range of continuous wave lengths. The data is analyzed with an algorithm that replaces compensation matrices and treats auto-fluorescence as an independent parameter. Thus, spectral flow cytometry should be capable to discriminate fluorochromes with similar emission peaks and provide multi-parametric analysis without compensation requirements. Here we show that spectral flow cytometry achieves a 21-parametric (19 fluorescent probes) characterization and deals with auto-fluorescent cells, providing high resolution of specifically fluorescence-labeled populations. Our results showed that spectral flow cytometry has advantages in the analysis of cell populations of tissues difficult to characterize in conventional flow cytometry, such as heart and intestine. Spectral flow cytometry thus combines the multi-parametric analytical capacity of the highest performing conventional flow cytometry without the requirement for compensation and enabling auto-fluorescence management.
Collapse
Affiliation(s)
| | - Mariana Valente
- Institut Pasteur, Immunology Department, Lymphopoiesis Unit, Paris, France
- Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
- Inserm U1223, Paris, France
- Instituto de Investigação e Inovação em Saúde (i3s) & INEB–Instituto de Engenharia Biomédica, Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironment for Newtherapies Group, Porto, Portugal
- ICBAS–Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Cumano
- Institut Pasteur, Immunology Department, Lymphopoiesis Unit, Paris, France
- Univ. Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
- Inserm U1223, Paris, France
- * E-mail:
| | - Sophie Novault
- Institut Pasteur, Flow Cytometry Core Facility, Paris, France
| |
Collapse
|
82
|
Foglia MJ, Poss KD. Building and re-building the heart by cardiomyocyte proliferation. Development 2016; 143:729-40. [PMID: 26932668 DOI: 10.1242/dev.132910] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adult human heart does not regenerate significant amounts of lost tissue after injury. Rather than making new, functional muscle, human hearts are prone to scarring and hypertrophy, which can often lead to fatal arrhythmias and heart failure. The most-cited basis of this ineffective cardiac regeneration in mammals is the low proliferative capacity of adult cardiomyocytes. However, mammalian cardiomyocytes can avidly proliferate during fetal and neonatal development, and both adult zebrafish and neonatal mice can regenerate cardiac muscle after injury, suggesting that latent regenerative potential exists. Dissecting the cellular and molecular mechanisms that promote cardiomyocyte proliferation throughout life, deciphering why proliferative capacity normally dissipates in adult mammals, and deriving means to boost this capacity are primary goals in cardiovascular research. Here, we review our current understanding of how cardiomyocyte proliferation is regulated during heart development and regeneration.
Collapse
Affiliation(s)
- Matthew J Foglia
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
83
|
Captur G, Wilson R, Bennett MF, Luxán G, Nasis A, de la Pompa JL, Moon JC, Mohun TJ. Morphogenesis of myocardial trabeculae in the mouse embryo. J Anat 2016; 229:314-25. [PMID: 27020702 PMCID: PMC4948049 DOI: 10.1111/joa.12465] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2016] [Indexed: 01/26/2023] Open
Abstract
Formation of trabeculae in the embryonic heart and the remodelling that occurs prior to birth is a conspicuous, but poorly understood, feature of vertebrate cardiogenesis. Mutations disrupting trabecular development in the mouse are frequently embryonic lethal, testifying to the importance of the trabeculae, and aberrant trabecular structure is associated with several human cardiac pathologies. Here, trabecular architecture in the developing mouse embryo has been analysed using high-resolution episcopic microscopy (HREM) and three-dimensional (3D) modelling. This study shows that at all stages from mid-gestation to birth, the ventricular trabeculae comprise a complex meshwork of myocardial strands. Such an arrangement defies conventional methods of measurement, and an approach based upon fractal algorithms has been used to provide an objective measure of trabecular complexity. The extent of trabeculation as it changes along the length of left and right ventricles has been quantified, and the changes that occur from formation of the four-chambered heart until shortly before birth have been mapped. This approach not only measures qualitative features evident from visual inspection of 3D models, but also detects subtle, consistent and regionally localised differences that distinguish each ventricle and its developmental stage. Finally, the combination of HREM imaging and fractal analysis has been applied to analyse changes in embryonic heart structure in a genetic mouse model in which trabeculation is deranged. It is shown that myocardial deletion of the Notch pathway component Mib1 (Mib1(flox/flox) ; cTnT-cre) results in a complex array of abnormalities affecting trabeculae and other parts of the heart.
Collapse
Affiliation(s)
- Gabriella Captur
- Institute of Cardiovascular ScienceUniversity College LondonLondonUK
- The Barts Heart CentreBarts Health NHS TrustLondonUK
| | - Robert Wilson
- The Francis Crick Institute Mill Hill LaboratoryThe RidgewayLondonUK
| | - Michael F Bennett
- The Francis Crick Institute Mill Hill LaboratoryThe RidgewayLondonUK
| | - Guillermo Luxán
- Intercellular Signalling in Cardiovascular Development & Disease LaboratoryCentro Nacional de Investigaciones Cardiovasculares (CNIC)Melchor Fernández AlmagroMadridSpain
| | - Arthur Nasis
- Monash Cardiovascular Research CentreMonashHEARTMonash UniversityClaytonAustralia
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development & Disease LaboratoryCentro Nacional de Investigaciones Cardiovasculares (CNIC)Melchor Fernández AlmagroMadridSpain
| | - James C Moon
- Institute of Cardiovascular ScienceUniversity College LondonLondonUK
- The Barts Heart CentreBarts Health NHS TrustLondonUK
| | - Timothy J Mohun
- The Francis Crick Institute Mill Hill LaboratoryThe RidgewayLondonUK
| |
Collapse
|
84
|
Han P, Bloomekatz J, Ren J, Zhang R, Grinstein JD, Zhao L, Burns CG, Burns CE, Anderson RM, Chi NC. Coordinating cardiomyocyte interactions to direct ventricular chamber morphogenesis. Nature 2016; 534:700-4. [PMID: 27357797 PMCID: PMC5330678 DOI: 10.1038/nature18310] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 05/05/2016] [Indexed: 12/17/2022]
Abstract
Many organs are composed of complex tissue walls that are structurally organized to optimize organ function. In particular, the ventricular myocardial wall of the heart is comprised of an outer compact layer that concentrically encircles the ridge-like inner trabecular layer. Although disruption in the morphogenesis of this myocardial wall can lead to various forms of congenital heart disease (CHD)1 and non-compaction cardiomyopathies2, it remains unclear how embryonic cardiomyocytes assemble to form ventricular wall layers of appropriate spatial dimensions and myocardial mass. Here, we utilize advanced genetic and imaging tools in zebrafish to reveal an interplay between myocardial Notch and Erbb2 signaling that directs the spatial allocation of myocardial cells to their proper morphologic positions in the ventricular wall. Although previous studies have shown that endocardial Notch signaling non-cell-autonomously promotes myocardial trabeculation through Erbb2 and BMP signaling3, we discover that distinct ventricular cardiomyocyte clusters exhibit myocardial Notch activity that cell-autonomously inhibits Erbb2 signaling and prevents cardiomyocyte sprouting and trabeculation. Myocardial-specific Notch inactivation leads to ventricles of reduced size and increased wall thickness due to excessive trabeculae, whereas widespread myocardial Notch activity results in ventricles of increased size with a single-cell thick wall but no trabeculae. Notably, this myocardial Notch signaling is activated non-cell-autonomously by neighboring Erbb2-activated cardiomyocytes that sprout and form nascent trabeculae. Thus, these findings support an interactive cellular feedback process that guides the assembly of cardiomyocytes to morphologically create the ventricular myocardial wall and more broadly provides insight into the cellular dynamics of how diverse cell lineages organize to create form.
Collapse
Affiliation(s)
- Peidong Han
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA
| | - Joshua Bloomekatz
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA
| | - Jie Ren
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA
| | - Ruilin Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA
| | - Jonathan D Grinstein
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA
| | - Long Zhao
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - C Geoffrey Burns
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Caroline E Burns
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Ryan M Anderson
- Center for Diabetes and Metabolic Diseases, Department of Pediatrics and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, California 92093, USA.,Institute of Genomic Medicine, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
85
|
Haack T, Abdelilah-Seyfried S. The force within: endocardial development, mechanotransduction and signalling during cardiac morphogenesis. Development 2016; 143:373-86. [PMID: 26839341 DOI: 10.1242/dev.131425] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.
Collapse
Affiliation(s)
- Timm Haack
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, Hannover D-30625, Germany Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, Potsdam D-14476, Germany
| |
Collapse
|
86
|
Abstract
During cardiac trabeculation, cardiomyocytes delaminate from the outermost (compact) layer to form complex muscular structures known as trabeculae. As these cardiomyocytes delaminate, the remodeling of adhesion junctions must be tightly coordinated so cells can extrude from the compact layer while remaining in tight contact with their neighbors. In this study, we examined the distribution of N-cadherin (Cdh2) during cardiac trabeculation in zebrafish. By analyzing the localization of a Cdh2-EGFP fusion protein expressed under the control of the zebrafish cdh2 promoter, we initially observed Cdh2-EGFP expression along the lateral sides of embryonic cardiomyocytes, in an evenly distributed pattern, and with the occasional appearance of punctae. Within a few hours, Cdh2-EGFP distribution on the lateral sides of cardiomyocytes evolves into a clear punctate pattern as Cdh2-EGFP molecules outside the punctae cluster to increase the size of these aggregates. In addition, Cdh2-EGFP molecules also appear on the basal side of cardiomyocytes that remain in the compact layer. Delaminating cardiomyocytes accumulate Cdh2-EGFP on the surface facing the basal side of compact layer cardiomyocytes, thereby allowing tight adhesion between these layers. Importantly, we find that blood flow/cardiac contractility is required for the transition from an even distribution of Cdh2-EGFP to the formation of punctae. Furthermore, using time-lapse imaging of beating hearts in conjunction with a Cdh2 tandem fluorescent protein timer transgenic line, we observed that Cdh2-EGFP molecules appear to move from the lateral to the basal side of cardiomyocytes along the cell membrane, and that Erb-b2 receptor tyrosine kinase 2 (Erbb2) function is required for this relocalization.
Collapse
|
87
|
D'Amato G, Luxán G, de la Pompa JL. Notch signalling in ventricular chamber development and cardiomyopathy. FEBS J 2016; 283:4223-4237. [DOI: 10.1111/febs.13773] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/12/2016] [Accepted: 06/03/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Gaetano D'Amato
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC); Madrid Spain
| | - Guillermo Luxán
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC); Madrid Spain
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC); Madrid Spain
| |
Collapse
|
88
|
Samsa LA, Givens C, Tzima E, Stainier DYR, Qian L, Liu J. Cardiac contraction activates endocardial Notch signaling to modulate chamber maturation in zebrafish. Development 2016; 142:4080-91. [PMID: 26628092 DOI: 10.1242/dev.125724] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Congenital heart disease often features structural abnormalities that emerge during development. Accumulating evidence indicates a crucial role for cardiac contraction and the resulting fluid forces in shaping the heart, yet the molecular basis of this function is largely unknown. Using the zebrafish as a model of early heart development, we investigated the role of cardiac contraction in chamber maturation, focusing on the formation of muscular protrusions called trabeculae. By genetic and pharmacological ablation of cardiac contraction, we showed that cardiac contraction is required for trabeculation through its role in regulating notch1b transcription in the ventricular endocardium. We also showed that Notch1 activation induces expression of ephrin b2a (efnb2a) and neuregulin 1 (nrg1) in the endocardium to promote trabeculation and that forced Notch activation in the absence of cardiac contraction rescues efnb2a and nrg1 expression. Using in vitro and in vivo systems, we showed that primary cilia are important mediators of fluid flow to stimulate Notch expression. Together, our findings describe an essential role for cardiac contraction-responsive transcriptional changes in endocardial cells to regulate cardiac chamber maturation.
Collapse
Affiliation(s)
- Leigh Ann Samsa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chris Givens
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eleni Tzima
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Li Qian
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
89
|
Brown DR, Samsa LA, Qian L, Liu J. Advances in the Study of Heart Development and Disease Using Zebrafish. J Cardiovasc Dev Dis 2016; 3. [PMID: 27335817 PMCID: PMC4913704 DOI: 10.3390/jcdd3020013] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Animal models of cardiovascular disease are key players in the translational medicine pipeline used to define the conserved genetic and molecular basis of disease. Congenital heart diseases (CHDs) are the most common type of human birth defect and feature structural abnormalities that arise during cardiac development and maturation. The zebrafish, Danio rerio, is a valuable vertebrate model organism, offering advantages over traditional mammalian models. These advantages include the rapid, stereotyped and external development of transparent embryos produced in large numbers from inexpensively housed adults, vast capacity for genetic manipulation, and amenability to high-throughput screening. With the help of modern genetics and a sequenced genome, zebrafish have led to insights in cardiovascular diseases ranging from CHDs to arrhythmia and cardiomyopathy. Here, we discuss the utility of zebrafish as a model system and summarize zebrafish cardiac morphogenesis with emphasis on parallels to human heart diseases. Additionally, we discuss the specific tools and experimental platforms utilized in the zebrafish model including forward screens, functional characterization of candidate genes, and high throughput applications.
Collapse
Affiliation(s)
- Daniel R. Brown
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (D.R.B.); (L.Q.)
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Leigh Ann Samsa
- Department of Cell Biology and Physiology; University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (D.R.B.); (L.Q.)
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (D.R.B.); (L.Q.)
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +1-919-962-0326; Fax: +1-919- 843-2063
| |
Collapse
|
90
|
Abstract
The molecular mechanisms underlying cardiogenesis are of critical biomedical importance due to the high prevalence of cardiac birth defects. Over the past two decades, the zebrafish has served as a powerful model organism for investigating heart development, facilitated by its powerful combination of optical access to the embryonic heart and plentiful opportunities for genetic analysis. Work in zebrafish has identified numerous factors that are required for various aspects of heart formation, including the specification and differentiation of cardiac progenitor cells, the morphogenesis of the heart tube, cardiac chambers, and atrioventricular canal, and the establishment of proper cardiac function. However, our current roster of regulators of cardiogenesis is by no means complete. It is therefore valuable for ongoing studies to continue pursuit of additional genes and pathways that control the size, shape, and function of the zebrafish heart. An extensive arsenal of techniques is available to distinguish whether particular mutations, morpholinos, or small molecules disrupt specific processes during heart development. In this chapter, we provide a guide to the experimental strategies that are especially effective for the characterization of cardiac phenotypes in the zebrafish embryo.
Collapse
Affiliation(s)
- A R Houk
- University of California, San Diego, CA, United States
| | - D Yelon
- University of California, San Diego, CA, United States
| |
Collapse
|
91
|
Li J, Miao L, Shieh D, Spiotto E, Li J, Zhou B, Paul A, Schwartz RJ, Firulli AB, Singer HA, Huang G, Wu M. Single-Cell Lineage Tracing Reveals that Oriented Cell Division Contributes to Trabecular Morphogenesis and Regional Specification. Cell Rep 2016; 15:158-170. [PMID: 27052172 DOI: 10.1016/j.celrep.2016.03.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/20/2016] [Accepted: 02/26/2016] [Indexed: 01/07/2023] Open
Abstract
The cardiac trabeculae are sheet-like structures extending from the myocardium that function to increase surface area. A lack of trabeculation causes embryonic lethality due to compromised cardiac function. To understand the cellular and molecular mechanisms of trabecular formation, we genetically labeled individual cardiomyocytes prior to trabeculation via the brainbow multicolor system and traced and analyzed the labeled cells during trabeculation by whole-embryo clearing and imaging. The clones derived from labeled single cells displayed four different geometric patterns that are derived from different patterns of oriented cell division (OCD) and migration. Of the four types of clones, the inner, transmural, and mixed clones contributed to trabecular cardiomyocytes. Further studies showed that perpendicular OCD is an extrinsic asymmetric cell division that putatively contributes to trabecular regional specification. Furthermore, N-Cadherin deletion in labeled clones disrupted the clonal patterns. In summary, our data demonstrate that OCD contributes to trabecular morphogenesis and specification.
Collapse
Affiliation(s)
- Jingjing Li
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Lianjie Miao
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - David Shieh
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Ernest Spiotto
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Jian Li
- Key Laboratory of Molecular Medicine, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | - Antoni Paul
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Robert J Schwartz
- Biology and Biochemistry, University of Houston, Houston, TX 77204-5001, USA
| | - Anthony B Firulli
- Riley Heart Research Center, Indiana University, Indianapolis, IN 46202, USA
| | - Harold A Singer
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Guoying Huang
- Key Laboratory of Molecular Medicine, Ministry of Education, Fudan University, Shanghai 200032, China
| | - Mingfu Wu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
92
|
Abstract
The zebrafish model is the only available high-throughput vertebrate assessment system, and it is uniquely suited for studies of in vivo cell biology. A sequenced and annotated genome has revealed a large degree of evolutionary conservation in comparison to the human genome. Due to our shared evolutionary history, the anatomical and physiological features of fish are highly homologous to humans, which facilitates studies relevant to human health. In addition, zebrafish provide a very unique vertebrate data stream that allows researchers to anchor hypotheses at the biochemical, genetic, and cellular levels to observations at the structural, functional, and behavioral level in a high-throughput format. In this review, we will draw heavily from toxicological studies to highlight advances in zebrafish high-throughput systems. Breakthroughs in transgenic/reporter lines and methods for genetic manipulation, such as the CRISPR-Cas9 system, will be comprised of reports across diverse disciplines.
Collapse
Affiliation(s)
- Gloria R Garcia
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Pamela D Noyes
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA
| | - Robert L Tanguay
- Oregon State University, Department of Environmental and Molecular Toxicology, Environmental Health Sciences Center, Corvallis, OR 97331, USA.
| |
Collapse
|
93
|
Foglia MJ, Cao J, Tornini VA, Poss KD. Multicolor mapping of the cardiomyocyte proliferation dynamics that construct the atrium. Development 2016; 143:1688-96. [PMID: 26989176 DOI: 10.1242/dev.136606] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/08/2016] [Indexed: 01/13/2023]
Abstract
The orchestrated division of cardiomyocytes assembles heart chambers of distinct morphology. To understand the structural divergence of the cardiac chambers, we determined the contributions of individual embryonic cardiomyocytes to the atrium in zebrafish by multicolor fate-mapping and we compare our analysis to the established proliferation dynamics of ventricular cardiomyocytes. We find that most atrial cardiomyocytes become rod-shaped in the second week of life, generating a single-muscle-cell-thick myocardial wall with a striking webbed morphology. Inner pectinate myofibers form mainly by direct branching, unlike delamination events that create ventricular trabeculae. Thus, muscle clones assembling the atrial chamber can extend from wall to lumen. As zebrafish mature, atrial wall cardiomyocytes proliferate laterally to generate cohesive patches of diverse shapes and sizes, frequently with dominant clones that comprise 20-30% of the wall area. A subpopulation of cardiomyocytes that transiently express atrial myosin heavy chain (amhc) contributes substantially to specific regions of the ventricle, suggesting an unappreciated level of plasticity during chamber formation. Our findings reveal proliferation dynamics and fate decisions of cardiomyocytes that produce the distinct architecture of the atrium.
Collapse
Affiliation(s)
- Matthew J Foglia
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jingli Cao
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Valerie A Tornini
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
94
|
Collins MM, Stainier DYR. Organ Function as a Modulator of Organ Formation: Lessons from Zebrafish. Curr Top Dev Biol 2016; 117:417-33. [PMID: 26969993 DOI: 10.1016/bs.ctdb.2015.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Organogenesis requires an intricate balance between cell differentiation and tissue growth to generate a complex and fully functional organ. However, organogenesis is not solely driven by genetic inputs, as the development of several organ systems requires their own functionality. This theme is particularly evident in the developing heart as progression of cardiac development is accompanied by increased and altered hemodynamic forces. In the absence or disruption of these forces, heart development is abnormal, suggesting that the heart must sense these changes and respond appropriately. Here, we discuss concepts of how embryonic heart function contributes to heart development using lessons learned mostly from studies in zebrafish.
Collapse
Affiliation(s)
- Michelle M Collins
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
95
|
Hemodynamics driven cardiac valve morphogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1760-6. [PMID: 26608609 DOI: 10.1016/j.bbamcr.2015.11.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 11/22/2022]
Abstract
Mechanical forces are instrumental to cardiovascular development and physiology. The heart beats approximately 2.6 billion times in a human lifetime and heart valves ensure that these contractions result in an efficient, unidirectional flow of the blood. Composed of endocardial cells (EdCs) and extracellular matrix (ECM), cardiac valves are among the most mechanically challenged structures of the body both during and after their development. Understanding how hemodynamic forces modulate cardiovascular function and morphogenesis is key to unraveling the relationship between normal and pathological cardiovascular development and physiology. Most valve diseases have their origins in embryogenesis, either as signs of abnormal developmental processes or the aberrant re-expression of fetal gene programs normally quiescent in adulthood. Here we review recent discoveries in the mechanobiology of cardiac valve development and introduce the latest technologies being developed in the zebrafish, including live cell imaging and optical technologies, as well as modeling approaches that are currently transforming this field. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
|
96
|
Boselli F, Vermot J. Live imaging and modeling for shear stress quantification in the embryonic zebrafish heart. Methods 2015; 94:129-34. [PMID: 26390811 DOI: 10.1016/j.ymeth.2015.09.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/07/2015] [Accepted: 09/17/2015] [Indexed: 01/17/2023] Open
Abstract
Hemodynamic shear stress is sensed by the endocardial cells composing the inner cell layer of the heart, and plays a major role in cardiac morphogenesis. Yet, the underlying hemodynamics and the associated mechanical stimuli experienced by endocardial cells remains poorly understood. Progress in the field has been hampered by the need for high temporal resolution imaging allowing the flow profiles generated in the beating heart to be resolved. To fill this gap, we propose a method to analyze the wall dynamics, the flow field, and the wall shear stress of the developing zebrafish heart. This method combines live confocal imaging and computational fluid dynamics to overcome difficulties related to live imaging of blood flow in the developing heart. To provide an example of the applicability of the method, we discuss the hemodynamic frequency content sensed by endocardial cells at the onset of valve formation, and how the fundamental frequency of the wall shear stress represents a unique mechanical cue to endocardial, heart-valve precursors.
Collapse
Affiliation(s)
- Francesco Boselli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France; Université de Strasbourg, Illkirch, France.
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR 7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964 Illkirch, France; Université de Strasbourg, Illkirch, France.
| |
Collapse
|
97
|
Matrone G, Wilson KS, Mullins JJ, Tucker CS, Denvir MA. Temporal cohesion of the structural, functional and molecular characteristics of the developing zebrafish heart. Differentiation 2015; 89:117-27. [PMID: 26095446 DOI: 10.1016/j.diff.2015.05.001] [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: 10/19/2014] [Revised: 04/06/2015] [Accepted: 05/10/2015] [Indexed: 11/25/2022]
Abstract
Heart formation is a complex, dynamic and highly coordinated process of molecular, morphogenetic and functional factors with each interacting and contributing to formation of the mature organ. Cardiac abnormalities in early life can be lethal in mammals but not in the zebrafish embryo which has been widely used to study the developing heart. While early cardiac development in the zebrafish has been well characterized, functional changes during development and how these relate to architectural, cellular and molecular aspects of development have not been well described previously. To address this we have carefully characterised cardiac structure, function, cardiomyocyte proliferation and cardiac-specific gene expression between 48 and 120 hpf in the zebrafish. We show that the zebrafish heart increases in volume and changes shape significantly between 48 and 72 hpf accompanied by a 40% increase in cardiomyocyte number. Between 96 and 120 hpf, while external heart expansion slows, there is rapid formation of a mature and extensive trabecular network within the ventricle chamber. While ejection fraction does not change during the course of development other determinants of contractile function increase significantly particularly between 72 and 96 hpf leading to an increase in cardinal vein blood flow. This study has revealed a number of novel aspects of cardiac developmental dynamics with striking temporal orchestration of structure and function within the first few days of development. These changes are associated with changes in expression of developmental and maturational genes. This study provides important insights into the complex temporal relationship between structure and function of the developing zebrafish heart.
Collapse
Affiliation(s)
- Gianfranco Matrone
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.
| | - Kathryn S Wilson
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - John J Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Carl S Tucker
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Martin A Denvir
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
98
|
Trivedi V, Truong TV, Trinh LA, Holland DB, Liebling M, Fraser SE. Dynamic structure and protein expression of the live embryonic heart captured by 2-photon light sheet microscopy and retrospective registration. BIOMEDICAL OPTICS EXPRESS 2015; 6:2056-66. [PMID: 26114028 PMCID: PMC4473743 DOI: 10.1364/boe.6.002056] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/25/2015] [Accepted: 05/01/2015] [Indexed: 05/05/2023]
Abstract
We present an imaging and image reconstruction pipeline that captures the dynamic three-dimensional beating motion of the live embryonic zebrafish heart at subcellular resolution. Live, intact zebrafish embryos were imaged using 2-photon light sheet microscopy, which offers deep and fast imaging at 70 frames per second, and the individual optical sections were assembled into a full 4D reconstruction of the beating heart using an optimized retrospective image registration algorithm. This imaging and reconstruction platform permitted us to visualize protein expression patterns at endogenous concentrations in zebrafish gene trap lines.
Collapse
Affiliation(s)
- Vikas Trivedi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
- Translational Imaging Center, University of Southern California, Molecular and Computational Biology, Los Angeles CA 90089, USA
- Equal contribution
| | - Thai V. Truong
- Translational Imaging Center, University of Southern California, Molecular and Computational Biology, Los Angeles CA 90089, USA
- Equal contribution
| | - Le A. Trinh
- Translational Imaging Center, University of Southern California, Molecular and Computational Biology, Los Angeles CA 90089, USA
| | - Daniel B. Holland
- Translational Imaging Center, University of Southern California, Molecular and Computational Biology, Los Angeles CA 90089, USA
| | - Michael Liebling
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara CA 93111 USA
- Idiap Research Institute, 1920 Martigny, Switzerland
| | - Scott E. Fraser
- Translational Imaging Center, University of Southern California, Molecular and Computational Biology, Los Angeles CA 90089, USA
| |
Collapse
|
99
|
Chaudhry B, Ramsbottom S, Henderson DJ. Genetics of cardiovascular development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 124:19-41. [PMID: 24751425 DOI: 10.1016/b978-0-12-386930-2.00002-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Structural malformations of the heart are the commonest abnormalities found at the time of birth and the incidence is higher in fetuses that are lost during the first trimester. Although the form of the heart has been studied for centuries, it is in the past decades that the genetic pathways that control heart development have been unraveled. Recently, the concept of the second heart field, a population of multipotent cardiac cells that augment the initial simple heart tube, has clarified the development of the heart. Understanding how the second heart field is used in morphogenesis and how genes interact in a subtle and more complex way is moving us closer to understanding how the normal heart forms and why abnormalities occur. In this chapter, we present a description of the morphological processes that create the formed postnatal human heart and emphasize key genetic pathways and genes that control these aspects. Where possible, these are also linked to the common patterns of human cardiac malformation. Undoubtedly, the details will refine or change with further research but emphasis has been placed on areas of greatest certainty and the presentation designed to promote a general understanding.
Collapse
Affiliation(s)
- Bill Chaudhry
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Simon Ramsbottom
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| | - Deborah J Henderson
- Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom
| |
Collapse
|
100
|
ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol 2015; 17:627-38. [PMID: 25848746 DOI: 10.1038/ncb3149] [Citation(s) in RCA: 505] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 03/05/2015] [Indexed: 12/14/2022]
Abstract
The murine neonatal heart can regenerate after injury through cardiomyocyte (CM) proliferation, although this capacity markedly diminishes after the first week of life. Neuregulin-1 (NRG1) administration has been proposed as a strategy to promote cardiac regeneration. Here, using loss- and gain-of-function genetic tools, we explore the role of the NRG1 co-receptor ERBB2 in cardiac regeneration. NRG1-induced CM proliferation diminished one week after birth owing to a reduction in ERBB2 expression. CM-specific Erbb2 knockout revealed that ERBB2 is required for CM proliferation at embryonic/neonatal stages. Induction of a constitutively active ERBB2 (caERBB2) in neonatal, juvenile and adult CMs resulted in cardiomegaly, characterized by extensive CM hypertrophy, dedifferentiation and proliferation, differentially mediated by ERK, AKT and GSK3β/β-catenin signalling pathways. Transient induction of caERBB2 following myocardial infarction triggered CM dedifferentiation and proliferation followed by redifferentiation and regeneration. Thus, ERBB2 is both necessary for CM proliferation and sufficient to reactivate postnatal CM proliferative and regenerative potentials.
Collapse
|