1
|
Medina-Jover F, Riera-Mestre A, Viñals F. Rethinking growth factors: the case of BMP9 during vessel maturation. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R1-R14. [PMID: 35350597 PMCID: PMC8942324 DOI: 10.1530/vb-21-0019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/07/2022] [Indexed: 12/21/2022]
Abstract
Angiogenesis is an essential process for correct development and physiology. This mechanism is tightly regulated by many signals that activate several pathways, which are constantly interacting with each other. There is mounting evidence that BMP9/ALK1 pathway is essential for a correct vessel maturation. Alterations in this pathway lead to the development of hereditary haemorrhagic telangiectasias. However, little was known about the BMP9 signalling cascade until the last years. Recent reports have shown that while BMP9 arrests cell cycle, it promotes the activation of anabolic pathways to enhance endothelial maturation. In light of this evidence, a new criterion for the classification of cytokines is proposed here, based on the physiological objective of the activation of anabolic routes. Whether this activation by a growth factor is needed to sustain mitosis or to promote a specific function such as matrix formation is a critical characteristic that needs to be considered to classify growth factors. Hence, the state-of-the-art of BMP9/ALK1 signalling is reviewed here, as well as its implications in normal and pathogenic angiogenesis.
Collapse
Affiliation(s)
- Ferran Medina-Jover
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d’Oncologia, Hospital Duran i Reynals, L’Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut (Campus de Bellvitge), Universitat de Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Antoni Riera-Mestre
- Hereditary Hemorrhagic Telangiectasia Unit, Internal Medicine Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat, Barcelona, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Universitat de Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Viñals
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d’Oncologia, Hospital Duran i Reynals, L’Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut (Campus de Bellvitge), Universitat de Barcelona, L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
2
|
Kulikauskas MR, X S, Bautch VL. The versatility and paradox of BMP signaling in endothelial cell behaviors and blood vessel function. Cell Mol Life Sci 2022; 79:77. [PMID: 35044529 PMCID: PMC8770421 DOI: 10.1007/s00018-021-04033-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Blood vessels expand via sprouting angiogenesis, and this process involves numerous endothelial cell behaviors, such as collective migration, proliferation, cell–cell junction rearrangements, and anastomosis and lumen formation. Subsequently, blood vessels remodel to form a hierarchical network that circulates blood and delivers oxygen and nutrients to tissue. During this time, endothelial cells become quiescent and form a barrier between blood and tissues that regulates transport of liquids and solutes. Bone morphogenetic protein (BMP) signaling regulates both proangiogenic and homeostatic endothelial cell behaviors as blood vessels form and mature. Almost 30 years ago, human pedigrees linked BMP signaling to diseases associated with blood vessel hemorrhage and shunts, and recent work greatly expanded our knowledge of the players and the effects of vascular BMP signaling. Despite these gains, there remain paradoxes and questions, especially with respect to how and where the different and opposing BMP signaling outputs are regulated. This review examines endothelial cell BMP signaling in vitro and in vivo and discusses the paradox of BMP signals that both destabilize and stabilize endothelial cell behaviors.
Collapse
Affiliation(s)
- Molly R Kulikauskas
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shaka X
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
3
|
Della Verde G, Mochizuki M, Lorenz V, Roux J, Xu L, Ramin-Wright L, Pfister O, Kuster GM. Fms-like tyrosine kinase 3 is a regulator of the cardiac side population in mice. Life Sci Alliance 2021; 5:5/3/e202101112. [PMID: 34903561 PMCID: PMC8711848 DOI: 10.26508/lsa.202101112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Fms-like tyrosine kinase 3 (Flt3) is a regulator of hematopoietic progenitor cells and a target of tyrosine kinase inhibitors. Flt3-targeting tyrosine kinase inhibitors can have cardiovascular side effects. Flt3 and its ligand (Flt3L) are expressed in the heart, but little is known about their physiological functions. Here, we show that cardiac side population progenitor cells (SP-CPCs) from mice produce and are responsive to Flt3L. Compared with wild-type, flt3L-/- mice have less SP-CPCs with less contribution of CD45-CD34+ cells and lower expression of genes related to epithelial-to-mesenchymal transition, cardiovascular development and stem cell differentiation. Upon culturing, flt3L-/- SP-CPCs show increased proliferation and less vasculogenic commitment, whereas Akt phosphorylation is lower. Notably, proliferation and differentiation can be partially restored towards wild-type levels in the presence of alternative receptor tyrosine kinase-activating growth factors signaling through Akt. The lower vasculogenic potential of flt3L-/- SP-CPCs reflects in decreased microvascularisation and lower systolic function of flt3L-/- hearts. Thus, Flt3 regulates phenotype and function of murine SP-CPCs and contributes to cellular and molecular properties that are relevant for their cardiovasculogenic potential.
Collapse
Affiliation(s)
- Giacomo Della Verde
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Michika Mochizuki
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Vera Lorenz
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julien Roux
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lifen Xu
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Leandra Ramin-Wright
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Otmar Pfister
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland.,Department of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Gabriela M Kuster
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland .,Department of Cardiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
4
|
Wang Z, Wang H, Zhang Y, Yu F, Yu L, Zhang C. Single-cell RNA sequencing analysis to characterize cells and gene expression landscapes in atrial septal defect. J Cell Mol Med 2021; 25:9660-9673. [PMID: 34514716 PMCID: PMC8505850 DOI: 10.1111/jcmm.16914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to characterize the cells and gene expression landscape in atrial septal defect (ASD). We performed single-cell RNA sequencing of cells derived from cardiac tissue of an ASD patient. Unsupervised clustering analysis was performed to identify different cell populations, followed by the investigation of the cellular crosstalk by analysing ligand-receptor interactions across cell types. Finally, differences between ASD and normal samples for all cell types were further investigated. An expression matrix of 18,411 genes in 6487 cells was obtained and used in this analysis. Five cell types, including cardiomyocytes, endothelial cells, smooth muscle cells, fibroblasts and macrophages were identified. ASD showed a decreased proportion of cardiomyocytes and an increased proportion of fibroblasts. There was more cellular crosstalk among cardiomyocytes, fibroblasts and macrophages, especially between fibroblast and macrophage. For all cell types, the majority of the DEGs were downregulated in ASD samples. For cardiomyocytes, there were 199 DEGs (42 upregulated and 157 downregulated) between ASD and normal samples. PPI analysis showed that cardiomyocyte marker gene FABP4 interacted with FOS, while FOS showed interaction with NPPA. Cell trajectory analysis showed that FABP4, FOS, and NPPA showed different expression changes along the pseudotime trajectory. Our results showed that single-cell RNA sequencing provides a powerful tool to study DEG profiles in the cell subpopulations of interest at the single-cell level. These findings enhance the understanding of the underlying mechanisms of ASD at both the cellular and molecular level and highlight potential targets for the treatment of ASD.
Collapse
Affiliation(s)
- Zunzhe Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huating Wang
- Department of Cardiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Ya Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Fangpu Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Liwen Yu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| |
Collapse
|
5
|
Melchert J, Henningfeld KA, Richts S, Lingner T, Jonigk D, Pieler T. The secreted BMP antagonist ERFE is required for the development of a functional circulatory system in Xenopus. Dev Biol 2019; 459:138-148. [PMID: 31846624 DOI: 10.1016/j.ydbio.2019.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 01/24/2023]
Abstract
The hormone Erythroferrone (ERFE) is a member of the C1q/TNF-related protein family that regulates iron homeostasis through the suppression of hamp. In a gain of function screen in Xenopus embryos, we identified ERFE as a potent secondary axis-inducing agent. Experiments in Xenopus embryos and ectodermal explants revealed that ERFE functions as a selective inhibitor of the BMP pathway and the conserved C1q domain is not required for this activity. Inhibition occurs at the extracelluar level, through the interaction of ERFE with the BMP ligand. During early Xenopus embryogenesis, erfe is first expressed in the ventral blood islands where initial erythropoiesis occurs and later in circulating blood cells. ERFE knockdown does not alter the expression of etv.2, aplnr and flt1 in tailbud stage embryos indicating endothelial cell specification is independent of ERFE. However, in tadpole embryos, defects of the vascular network and primitive blood circulation are observed as well as edema formation. RNAseq analysis of ERFE morphant embryos also revealed the inhibition of gja4 indicating disruption of dorsal aorta formation.
Collapse
Affiliation(s)
- Juliane Melchert
- Institute of Developmental Biochemistry, University Medical Center Göttingen, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Justus-von-Liebig-Weg 11, 37077, Goettingen, Germany.
| | - Kristine A Henningfeld
- Institute of Developmental Biochemistry, University Medical Center Göttingen, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Justus-von-Liebig-Weg 11, 37077, Goettingen, Germany
| | - Sven Richts
- Institute of Developmental Biochemistry, University Medical Center Göttingen, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Justus-von-Liebig-Weg 11, 37077, Goettingen, Germany
| | - Thomas Lingner
- Transcriptome and Genome Analysis Laboratory, University Medical Center Göttingen, Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Danny Jonigk
- Institut für Pathologie, Medizinische Hochschule Hannover (MHH) Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Tomas Pieler
- Institute of Developmental Biochemistry, University Medical Center Göttingen, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Justus-von-Liebig-Weg 11, 37077, Goettingen, Germany
| |
Collapse
|
6
|
Russell BE, Rigueur D, Weaver KN, Sund K, Basil JS, Hufnagel RB, Prows CA, Oestreich A, Al-Gazali L, Hopkin RJ, Saal HM, Lyons K, Dauber A. Homozygous missense variant in BMPR1A resulting in BMPR signaling disruption and syndromic features. Mol Genet Genomic Med 2019; 7:e969. [PMID: 31493347 PMCID: PMC6825850 DOI: 10.1002/mgg3.969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/12/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background The bone morphogenetic protein (BMP) pathway is known to play an imperative role in bone, cartilage, and cardiac tissue formation. Truncating, heterozygous variants, and deletions of one of the essential receptors in this pathway, Bone Morphogenetic Protein Receptor Type1A (BMPR1A), have been associated with autosomal dominant juvenile polyposis. Heterozygous deletions have also been associated with cardiac and minor skeletal anomalies. Populations with atrioventricular septal defects are enriched for rare missense BMPR1A variants. Methods We report on a patient with a homozygous missense variant in BMPR1A causing skeletal abnormalities, growth failure a large atrial septal defect, severe subglottic stenosis, laryngomalacia, facial dysmorphisms, and developmental delays. Results Functional analysis of this variant shows increased chondrocyte death for cells with the mutated receptor, increased phosphorylated R‐Smads1/5/8, and loss of Sox9 expression mediated by decreased phosphorylation of p38. Conclusion This homozygous missense variant in BMPR1A appears to cause a distinct clinical phenotype.
Collapse
Affiliation(s)
- Bianca E Russell
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Diana Rigueur
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA, USA
| | - Kathryn N Weaver
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Kristen Sund
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Janet S Basil
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Robert B Hufnagel
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Cynthia A Prows
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Alan Oestreich
- Department of Radiology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Lihadh Al-Gazali
- Department of Pediatrics, United Arab Emirates University, Abu Dhabi, United Arab Emirates
| | - Robert J Hopkin
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Howard M Saal
- Division of Human Genetics, Cincinnati Children's Hospital and University of Cincinnati College of Medicine Department of Pediatrics, Cincinnati, OH, USA
| | - Karen Lyons
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA, USA.,Department of Orthopaedic Surgery, UCLA, Los Angeles, CA, USA
| | - Andrew Dauber
- Division of Endocrinology, Cincinnati Children's Hospital and University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
7
|
Watterston C, Zeng L, Onabadejo A, Childs SJ. MicroRNA26 attenuates vascular smooth muscle maturation via endothelial BMP signalling. PLoS Genet 2019; 15:e1008163. [PMID: 31091229 PMCID: PMC6538191 DOI: 10.1371/journal.pgen.1008163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 05/28/2019] [Accepted: 04/27/2019] [Indexed: 12/23/2022] Open
Abstract
As small regulatory transcripts, microRNAs (miRs) act as genetic ‘fine tuners’ of posttranscriptional events, and as genetic switches to promote phenotypic switching. The miR miR26a targets the BMP signalling effector, smad1. We show that loss of miR26a leads to hemorrhage (a loss of vascular stability) in vivo, suggesting altered vascular differentiation. Reduction in miR26a levels increases smad1 mRNA and phospho-Smad1 (pSmad1) levels. We show that increasing BMP signalling by overexpression of smad1 also leads to hemorrhage. Normalization of Smad1 levels through double knockdown of miR26a and smad1 rescues hemorrhage, suggesting a direct relationship between miR26a, smad1 and vascular stability. Using an in vivo BMP genetic reporter and pSmad1 staining, we show that the effect of miR26a on smooth muscle differentiation is non-autonomous; BMP signalling is active in embryonic endothelial cells, but not in smooth muscle cells. Nonetheless, increased BMP signalling due to loss of miR26a results in an increase in acta2-expressing smooth muscle cell numbers and promotes a differentiated smooth muscle morphology. Similarly, forced expression of smad1 in endothelial cells leads to an increase in smooth muscle cell number and coverage. Furthermore, smooth muscle phenotypes caused by inhibition of the BMP pathway are rescued by loss of miR26a. Taken together, our data suggest that miR26a modulates BMP signalling in endothelial cells and indirectly promotes a differentiated smooth muscle phenotype. Our data highlights how crosstalk from BMP-responsive endothelium to smooth muscle is important for smooth muscle differentiation. The structural integrity of a blood vessel is critical to ensure proper vessel support and vascular tone. Vascular smooth cells (vSMCs) are a key component of the vessel wall and, in their mature state, express contractile proteins that help to constrict and relax the vessel in response to blood flow changes. vSMCs differentiate from immature vascular mural cells that lack contractile function. Here, we use a zebrafish model to identify a small microRNA that regulates vascular stabilization. We show that a small regulatory RNA, microRNA26a is enriched in the endothelial lining of the blood vessel wall and, through signalling, communicates to the smooth muscle cell to control its maturation. Providing a mechanistic insight into vSMC differentiation may help develop and produce feasible miR-based pharmaceutical to promote SMC differentiation.
Collapse
Affiliation(s)
- Charlene Watterston
- Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Lei Zeng
- Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Abidemi Onabadejo
- Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
| | - Sarah J. Childs
- Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary AB, Canada
- * E-mail:
| |
Collapse
|
8
|
Abstract
Venous endothelial cells are molecularly and functionally distinct from their arterial counterparts. Although veins are often considered the default endothelial state, genetic manipulations can modulate both acquisition and loss of venous fate, suggesting that venous identity is the result of active transcriptional regulation. However, little is known about this process. Here we show that BMP signalling controls venous identity via the ALK3/BMPR1A receptor and SMAD1/SMAD5. Perturbations to TGF-β and BMP signalling in mice and zebrafish result in aberrant vein formation and loss of expression of the venous-specific gene Ephb4, with no effect on arterial identity. Analysis of a venous endothelium-specific enhancer for Ephb4 shows enriched binding of SMAD1/5 and a requirement for SMAD binding motifs. Further, our results demonstrate that BMP/SMAD-mediated Ephb4 expression requires the venous-enriched BMP type I receptor ALK3/BMPR1A. Together, our analysis demonstrates a requirement for BMP signalling in the establishment of Ephb4 expression and the venous vasculature. The establishment of functional vasculatures requires the specification of newly formed vessels into veins and arteries. Here, Neal et al. use a combination of genetic approaches in mice and zebrafish to show that BMP signalling, via ALK3 and SMAD1/5, is required for venous specification during blood vessel development.
Collapse
|
9
|
Yang J, Mishina Y. Generation and Identification of Genetically Modified Mice for BMP Receptors. Methods Mol Biol 2019; 1891:165-177. [PMID: 30414132 DOI: 10.1007/978-1-4939-8904-1_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BMP signaling is critical in embryogenesis and in the development of numerous tissues. Many genetically modified (knockout and transgenic) mice have been established to study BMP function in development and disease. Mice with altered BMP receptor genes (including global knockout, conditional knockout, and conditional constitutively active transgenic mouse lines) have been particularly informative. In this chapter, we describe how the genetically modified mice were generated and introduce genotyping methods. These methods include regular PCR and genomic real-time PCR using specific primers based on different constructs in different mice strains.
Collapse
Affiliation(s)
- Jingwen Yang
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.,State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Dissecting BMP signaling input into the gene regulatory networks driving specification of the blood stem cell lineage. Proc Natl Acad Sci U S A 2018; 114:5814-5821. [PMID: 28584091 DOI: 10.1073/pnas.1610615114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) that sustain lifelong blood production are created during embryogenesis. They emerge from a specialized endothelial population, termed hemogenic endothelium (HE), located in the ventral wall of the dorsal aorta (DA). In Xenopus, we have been studying the gene regulatory networks (GRNs) required for the formation of HSCs, and critically found that the hemogenic potential is defined at an earlier time point when precursors to the DA express hematopoietic as well as endothelial genes, in the definitive hemangioblasts (DHs). The GRN for DH programming has been constructed and, here, we show that bone morphogenetic protein (BMP) signaling is essential for the initiation of this GRN. BMP2, -4, and -7 are the principal ligands expressed in the lineage forming the HE. To investigate the requirement and timing of all BMP signaling in HSC ontogeny, we have used a transgenic line, which inducibly expresses an inhibitor of BMP signaling, Noggin, as well as a chemical inhibitor of BMP receptors, DMH1, and described the inputs from BMP signaling into the DH GRN and the HE, as well as into primitive hematopoiesis. BMP signaling is required in at least three points in DH programming: first to initiate the DH GRN through gata2 expression, then for kdr expression to enable the DH to respond to vascular endothelial growth factor A (VEGFA) ligand from the somites, and finally for gata2 expression in the DA, but is dispensable for HE specification after hemangioblasts have been formed.
Collapse
|
11
|
Peng Y, Song L, Li D, Kesterson R, Wang J, Wang L, Rokosh G, Wu B, Wang Q, Jiao K. Sema6D acts downstream of bone morphogenetic protein signalling to promote atrioventricular cushion development in mice. Cardiovasc Res 2018; 112:532-542. [PMID: 28172500 DOI: 10.1093/cvr/cvw200] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 08/10/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yin Peng
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lanying Song
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ding Li
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert Kesterson
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jianbo Wang
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lizhong Wang
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gregg Rokosh
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kai Jiao
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
BMP2 expression in the endocardial lineage is required for AV endocardial cushion maturation and remodeling. Dev Biol 2017; 430:113-128. [PMID: 28790014 DOI: 10.1016/j.ydbio.2017.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 07/16/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
Distal outgrowth, maturation and remodeling of the endocardial cushion mesenchyme in the atrioventricular (AV) canal are the essential morphogenetic events during four-chambered heart formation. Mesenchymalized AV endocardial cushions give rise to the AV valves and the membranous ventricular septum (VS). Failure of these processes results in several human congenital heart defects. Despite this clinical relevance, the mechanisms governing how mesenchymalized AV endocardial cushions mature and remodel into the membranous VS and AV valves have only begun to be elucidated. The role of BMP signaling in the myocardial and secondary heart forming lineage has been well studied; however, little is known about the role of BMP2 expression in the endocardial lineage. To fill this knowledge gap, we generated Bmp2 endocardial lineage-specific conditional knockouts (referred to as Bmp2 cKOEndo) by crossing conditionally-targeted Bmp2flox/flox mice with a Cre-driver line, Nfatc1Cre, wherein Cre-mediated recombination was restricted to the endocardial cells and their mesenchymal progeny. Bmp2 cKOEndo mouse embryos did not exhibit failure or delay in the initial AV endocardial cushion formation at embryonic day (ED) 9.5-11.5; however, significant reductions in AV cushion size were detected in Bmp2 cKOEndo mouse embryos when compared to control embryos at ED13.5 and ED16.5. Moreover, deletion of Bmp2 from the endocardial lineage consistently resulted in membranous ventricular septal defects (VSDs), and mitral valve deficiencies, as evidenced by the absence of stratification of mitral valves at birth. Muscular VSDs were not found in Bmp2 cKOEndo mouse hearts. To understand the underlying morphogenetic mechanisms leading to a decrease in cushion size, cell proliferation and cell death were examined for AV endocardial cushions. Phospho-histone H3 analyses for cell proliferation and TUNEL assays for apoptotic cell death did not reveal significant differences between control and Bmp2 cKOEndo in AV endocardial cushions. However, mRNA expression of the extracellular matrix components, versican, Has2, collagen 9a1, and periostin was significantly reduced in Bmp2 cKOEndo AV cushions. Expression of transcription factors implicated in the cardiac valvulogenesis, Snail2, Twist1 and Sox9, was also significantly reduced in Bmp2 cKOEndo AV cushions. These data provide evidence that BMP2 expression in the endocardial lineage is essential for the distal outgrowth, maturation and remodeling of AV endocardial cushions into the normal membranous VS and the stratified AV valves.
Collapse
|
13
|
Benn A, Hiepen C, Osterland M, Schütte C, Zwijsen A, Knaus P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence. FASEB J 2017; 31:4720-4733. [PMID: 28733457 PMCID: PMC5636702 DOI: 10.1096/fj.201700193rr] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 01/04/2023]
Abstract
Before the onset of sprouting angiogenesis, the endothelium is prepatterned for the positioning of tip and stalk cells. Both cell identities are not static, as endothelial cells (ECs) constantly compete for the tip cell position in a dynamic fashion. Here, we show that both bone morphogenetic protein 2 (BMP2) and BMP6 are proangiogenic in vitro and ex vivo and that the BMP type I receptors, activin receptor-like kinase 3 (ALK3) and ALK2, play crucial and distinct roles in this process. BMP2 activates the expression of tip cell-associated genes, such as delta-like ligand 4 (DLL4) and kinase insert domain receptor (KDR), and p38-heat shock protein 27 (HSP27)-dependent cell migration, thereby generating tip cell competence. Whereas BMP6 also triggers collective cell migration via the p38-HSP27 signaling axis, BMP6 induces in addition SMAD1/5 signaling, thereby promoting the expression of stalk cell-associated genes, such as hairy and enhancer of split 1 (HES1) and fms-like tyrosine kinase 1 (FLT1). Specifically, ALK3 is required for sprouting from HUVEC spheroids, whereas ALK2 represses sprout formation. We demonstrate that expression levels and respective complex formation of BMP type I receptors in ECs determine stalk vs. tip cell identity, thus contributing to endothelial plasticity during sprouting angiogenesis. As antiangiogenic monotherapies that target the VEGF or ALK1 pathways have not fulfilled efficacy objectives in clinical trials, the selective targeting of the ALK2/3 pathways may be an attractive new approach.-Benn, A., Hiepen, C., Osterland, M., Schütte, C., Zwijsen, A., Knaus, P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.,Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Marc Osterland
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - Christof Schütte
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - An Zwijsen
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; .,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
14
|
Beets K, Staring MW, Criem N, Maas E, Schellinx N, de Sousa Lopes SMC, Umans L, Zwijsen A. BMP-SMAD signalling output is highly regionalized in cardiovascular and lymphatic endothelial networks. BMC DEVELOPMENTAL BIOLOGY 2016; 16:34. [PMID: 27724845 PMCID: PMC5057272 DOI: 10.1186/s12861-016-0133-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/12/2016] [Indexed: 11/13/2022]
Abstract
BACKGROUND Bone morphogenetic protein (BMP) signalling has emerged as a fundamental pathway in endothelial cell biology and deregulation of this pathway is implicated in several vascular disorders. BMP signalling output in endothelial cells is highly context- and dose-dependent. Phosphorylation of the BMP intracellular effectors, SMAD1/5/9, is routinely used to monitor BMP signalling activity. To better understand the in vivo context-dependency of BMP-SMAD signalling, we investigated differences in BMP-SMAD transcriptional activity in different vascular beds during mouse embryonic and postnatal stages. For this, we used the BRE::gfp BMP signalling reporter mouse in which the BMP response element (BRE) from the ID1-promotor, a SMAD1/5/9 target gene, drives the expression of GFP. RESULTS A mosaic pattern of GFP was present in various angiogenic sprouting plexuses and in endocardium of cardiac cushions and trabeculae in the heart. High calibre veins seemed to be more BRE::gfp transcriptionally active than arteries, and ubiquitous activity was present in embryonic lymphatic vasculature. Postnatal lymphatic vessels showed however only discrete micro-domains of transcriptional activity. Dynamic shifts in transcriptional activity were also observed in the endocardium of the developing heart, with a general decrease in activity over time. Surprisingly, proliferative endothelial cells were almost never GFP-positive. Patches of transcriptional activity seemed to correlate with vasculature undergoing hemodynamic alterations. CONCLUSION The BRE::gfp mouse allows to investigate selective context-dependent aspects of BMP-SMAD signalling. Our data reveals the highly dynamic nature of BMP-SMAD mediated transcriptional regulation in time and space throughout the vascular tree, supporting that BMP-SMAD signalling can be a source of phenotypic diversity in some, but not all, healthy endothelium. This knowledge can provide insight in vascular bed or organ-specific diseases and phenotypic heterogeneity within an endothelial cell population.
Collapse
Affiliation(s)
- Karen Beets
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Michael W. Staring
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Nathan Criem
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Elke Maas
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Niels Schellinx
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Lieve Umans
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - An Zwijsen
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Lohmann F, Dangeti M, Soni S, Chen X, Planutis A, Baron MH, Choi K, Bieker JJ. The DEK Oncoprotein Is a Critical Component of the EKLF/KLF1 Enhancer in Erythroid Cells. Mol Cell Biol 2015; 35:3726-38. [PMID: 26303528 PMCID: PMC4589598 DOI: 10.1128/mcb.00382-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/06/2015] [Accepted: 08/17/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding how transcriptional regulators are themselves controlled is important in attaining a complete picture of the intracellular effects that follow signaling cascades during early development and cell-restricted differentiation. We have addressed this issue by focusing on the regulation of EKLF/KLF1, a zinc finger transcription factor that plays a necessary role in the global regulation of erythroid gene expression. Using biochemical affinity purification, we have identified the DEK oncoprotein as a critical factor that interacts with an essential upstream enhancer element of the EKLF promoter and exerts a positive effect on EKLF levels. This element also binds a core set of erythroid transcription factors, suggesting that DEK is part of a tissue-restricted enhanceosome that contains BMP4-dependent and -independent components. Together with local enrichment of properly coded histones and an open chromatin domain, optimal transcriptional activation of the EKLF locus can be established.
Collapse
Affiliation(s)
- Felix Lohmann
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Mohan Dangeti
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Shefali Soni
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Xiaoyong Chen
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Antanas Planutis
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Margaret H Baron
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | - Kyunghee Choi
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
16
|
BMPER Promotes Epithelial-Mesenchymal Transition in the Developing Cardiac Cushions. PLoS One 2015; 10:e0139209. [PMID: 26418455 PMCID: PMC4587915 DOI: 10.1371/journal.pone.0139209] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/10/2015] [Indexed: 01/25/2023] Open
Abstract
Formation of the cardiac valves is an essential component of cardiovascular development. Consistent with the role of the bone morphogenetic protein (BMP) signaling pathway in cardiac valve formation, embryos that are deficient for the BMP regulator BMPER (BMP-binding endothelial regulator) display the cardiac valve anomaly mitral valve prolapse. However, how BMPER deficiency leads to this defect is unknown. Based on its expression pattern in the developing cardiac cushions, we hypothesized that BMPER regulates BMP2-mediated signaling, leading to fine-tuned epithelial-mesenchymal transition (EMT) and extracellular matrix deposition. In the BMPER-/- embryo, EMT is dysregulated in the atrioventricular and outflow tract cushions compared with their wild-type counterparts, as indicated by a significant increase of Sox9-positive cells during cushion formation. However, proliferation is not impaired in the developing BMPER-/- valves. In vitro data show that BMPER directly binds BMP2. In cultured endothelial cells, BMPER blocks BMP2-induced Smad activation in a dose-dependent manner. In addition, BMP2 increases the Sox9 protein level, and this increase is inhibited by co-treatment with BMPER. Consistently, in the BMPER-/- embryos, semi-quantitative analysis of Smad activation shows that the canonical BMP pathway is significantly more active in the atrioventricular cushions during EMT. These results indicate that BMPER negatively regulates BMP-induced Smad and Sox9 activity during valve development. Together, these results identify BMPER as a regulator of BMP2-induced cardiac valve development and will contribute to our understanding of valvular defects.
Collapse
|
17
|
McLennan IS, Pankhurst MW. Anti-Müllerian hormone is a gonadal cytokine with two circulating forms and cryptic actions. J Endocrinol 2015; 226:R45-57. [PMID: 26163524 DOI: 10.1530/joe-15-0206] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2015] [Indexed: 12/23/2022]
Abstract
Anti-Müllerian hormone (AMH) is a multi-faceted gonadal cytokine. It is present in all vertebrates with its original function in phylogeny being as a regulator of germ cells in both sexes, and as a prime inducer of the male phenotype. Its ancient functions appear to be broadly conserved in mammals, but with this being obscured by its overt role in triggering the regression of the Müllerian ducts in male embryos. Sertoli and ovarian follicular cells primarily release AMH as a prohormone (proAMH), which forms a stable complex (AMHN,C) after cleavage by subtilisin/kexin-type proprotein convertases or serine proteinases. Circulating AMH is a mixture of proAMH and AMHN,C, suggesting that proAMH is activated within the gonads and putatively by its endocrine target-cells. The gonadal expression of the cleavage enzymes is subject to complex regulation, and the preliminary data suggest that this influences the relative proportions of proAMH and AMHN,C in the circulation. AMH shares an intracellular pathway with the bone morphogenetic protein (BMP) and growth differentiation factor (GDF) ligands. AMH is male specific during the initial stage of development, and theoretically should produce male biases throughout the body by adding a male-specific amplification of BMP/GDF signalling. Consistent with this, some of the male biases in neuron number and the non-sexual behaviours of mice are dependent on AMH. After puberty, circulating levels of AMH are similar in men and women. Putatively, the function of AMH in adulthood maybe to add a gonadal influence to BMP/GDF-regulated homeostasis.
Collapse
Affiliation(s)
- Ian S McLennan
- Department of AnatomyUniversity of Otago, PO Box 913, Dunedin 9054, New Zealand
| | - Michael W Pankhurst
- Department of AnatomyUniversity of Otago, PO Box 913, Dunedin 9054, New Zealand
| |
Collapse
|
18
|
Palencia-Desai S, Rost MS, Schumacher JA, Ton QV, Craig MP, Baltrunaite K, Koenig AL, Wang J, Poss KD, Chi NC, Stainier DYR, Sumanas S. Myocardium and BMP signaling are required for endocardial differentiation. Development 2015; 142:2304-15. [PMID: 26092845 DOI: 10.1242/dev.118687] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/12/2015] [Indexed: 01/09/2023]
Abstract
Endocardial and myocardial progenitors originate in distinct regions of the anterior lateral plate mesoderm and migrate to the midline where they coalesce to form the cardiac tube. Endocardial progenitors acquire a molecular identity distinct from other vascular endothelial cells and initiate expression of specific genes such as nfatc1. Yet the molecular pathways and tissue interactions involved in establishing endocardial identity are poorly understood. The endocardium develops in tight association with cardiomyocytes. To test for a potential role of the myocardium in endocardial morphogenesis, we used two different zebrafish models deficient in cardiomyocytes: the hand2 mutant and a myocardial-specific genetic ablation method. We show that in hand2 mutants endocardial progenitors migrate to the midline but fail to assemble into a cardiac cone and do not express markers of differentiated endocardium. Endocardial differentiation defects were rescued by myocardial but not endocardial-specific expression of hand2. In metronidazole-treated myl7:nitroreductase embryos, myocardial cells were targeted for apoptosis, which resulted in the loss of endocardial nfatc1 expression. However, endocardial cells were present and retained expression of general vascular endothelial markers. We further identified bone morphogenetic protein (BMP) as a candidate myocardium-derived signal required for endocardial differentiation. Chemical and genetic inhibition of BMP signaling at the tailbud stage resulted in severe inhibition of endocardial differentiation while there was little effect on myocardial development. Heat-shock-induced bmp2b expression rescued endocardial nfatc1 expression in hand2 mutants and in myocardium-depleted embryos. Our results indicate that the myocardium is crucial for endocardial morphogenesis and differentiation, and identify BMP as a signal involved in endocardial differentiation.
Collapse
Affiliation(s)
- Sharina Palencia-Desai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Megan S Rost
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jennifer A Schumacher
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Quynh V Ton
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael P Craig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kristina Baltrunaite
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew L Koenig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jinhu Wang
- Department of Cell Biology and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology and Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Neil C Chi
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA
| | - Didier Y R Stainier
- Department of Biochemistry and Biophysics, UCSF, San Francisco, CA 94158, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
19
|
D'Alessandro LCA, Al Turki S, Manickaraj AK, Manase D, Mulder BJM, Bergin L, Rosenberg HC, Mondal T, Gordon E, Lougheed J, Smythe J, Devriendt K, Bhattacharya S, Watkins H, Bentham J, Bowdin S, Hurles ME, Mital S. Exome sequencing identifies rare variants in multiple genes in atrioventricular septal defect. Genet Med 2015; 18:189-98. [PMID: 25996639 PMCID: PMC5988035 DOI: 10.1038/gim.2015.60] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/02/2015] [Indexed: 12/30/2022] Open
Abstract
Purpose The genetic etiology of atrioventricular septal defect (AVSD) is unknown in 40% cases. Conventional sequencing and arrays have identified the etiology in only a minority of non-syndromic individuals with AVSD. Methods Whole exome sequencing was performed in 81 unrelated probands with AVSD to identify potentially causal variants in a comprehensive set of 112 genes with strong biological relevance to AVSD. Results A significant enrichment of rare and rare/damaging variants was identified in the gene set, compared with controls (odds ratio 1.52, 95% confidence interval 1.35–1.71, p = 4.8 x 10-11). The enrichment was specific to AVSD probands compared with a non-AVSD cohort with tetralogy of Fallot (odds ratio 2.25, 95% confidence interval 1.84-2.76, p = 2.2 x 10-16). Six genes (NIPBL, CHD7, CEP152, BMPR1a, ZFPM2 and MDM4) were enriched for rare variants in AVSD compared to controls, including three syndrome-associated genes (NIPBL, CHD7, CEP152). The findings were confirmed in a replication cohort of 81 AVSD probands. Conclusion Mutations in genes with strong biological relevance to AVSD, including syndrome-associated genes, can contribute to AVSD even in those with isolated heart disease. The identification of a gene set associated with AVSD will facilitate targeted genetic screening in this cohort.
Collapse
Affiliation(s)
- Lisa C A D'Alessandro
- Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Saeed Al Turki
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK.,Department of Pathology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Ashok Kumar Manickaraj
- Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Dorin Manase
- Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Lynn Bergin
- Division of Cardiology, Department of Medicine, London Health Sciences Centre, London, Ontario, Canada
| | - Herschel C Rosenberg
- Department of Paediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Tapas Mondal
- Department of Pediatrics, Hamilton Health Sciences Centre, Hamilton, Ontario, Canada
| | - Elaine Gordon
- Division of Cardiology, Department of Medicine, Hamilton Health Sciences Centre, Hamilton, Ontario, Canada
| | - Jane Lougheed
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - John Smythe
- Department of Pediatrics, Kingston General Hospital, Kingston, Ontario, Canada
| | - Koen Devriendt
- Centre for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Shoumo Bhattacharya
- Radcliffe Department of Medicine & Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Radcliffe Department of Medicine & Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jamie Bentham
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts USA
| | - Sarah Bowdin
- Division of Genetics, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Seema Mital
- Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Thalgott J, Dos-Santos-Luis D, Lebrin F. Pericytes as targets in hereditary hemorrhagic telangiectasia. Front Genet 2015; 6:37. [PMID: 25763012 PMCID: PMC4327729 DOI: 10.3389/fgene.2015.00037] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/26/2015] [Indexed: 12/04/2022] Open
Abstract
Defective paracrine Transforming Growth Factor-β (TGF-β) signaling between endothelial cells and the neighboring mural cells have been thought to lead to the development of vascular lesions that are characteristic of Hereditary Hemorrhagic Telangiectasia (HHT). This review highlights recent progress in our understanding of TGF-β signaling in mural cell recruitment and vessel stabilization and how perturbed TGF-β signaling might contribute to defective endothelial-mural cell interaction affecting vessel functionalities. Our recent findings have provided exciting insights into the role of thalidomide, a drug that reduces both the frequency and the duration of epistaxis in individuals with HHT by targeting mural cells. These advances provide opportunities for the development of new therapies for vascular malformations.
Collapse
Affiliation(s)
- Jérémy Thalgott
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Damien Dos-Santos-Luis
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Franck Lebrin
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| |
Collapse
|
21
|
Snider P, Simmons O, Wang J, Hoang CQ, Conway SJ. Ectopic Noggin in a Population of Nfatc1 Lineage Endocardial Progenitors Induces Embryonic Lethality. J Cardiovasc Dev Dis 2014; 1:214-236. [PMID: 26090377 PMCID: PMC4469290 DOI: 10.3390/jcdd1030214] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The initial heart is composed of a myocardial tube lined by endocardial cells. The TGFβ superfamily is known to play an important role, as BMPs from the myocardium signal to the overlying endocardium to create an environment for EMT. Subsequently, BMP and TGFβ signaling pathways synergize to form primitive valves and regulate myocardial growth. In this study, we investigated the requirement of BMP activity by transgenic over-expression of extracellular BMP antagonist Noggin. Using Nfatc1Cre to drive lineage-restricted Noggin within the endocardium, we show that ectopic Noggin arrests cardiac development in E10.5-11 embryos, resulting in small hearts which beat poorly and die by E12.5. This is coupled with hypoplastic endocardial cushions, reduced trabeculation and fewer mature contractile fibrils in mutant hearts. Moreover, Nfatc1Cre-mediated diphtheria toxin fragment-A expression in the endocardium resulted in genetic ablation and a more severe phenotype with lethality at E11 and abnormal linear hearts. Molecular analysis demonstrated that endocardial Noggin resulted in a specific alteration of TGFβ/BMP-mediated signal transduction, in that, both Endoglin and ALK1 were downregulated in mutant endocardium. Combined, these results demonstrate the cell-autonomous requirement of the endocardial lineage and function of unaltered BMP levels in facilitating endothelium-cardiomyocyte cross-talk and promoting endocardial cushion formation.
Collapse
Affiliation(s)
| | | | | | | | - Simon J. Conway
- Author to whom correspondence should be addressed; ; Tel.: +317-278-8781; Fax: +317-278-0138
| |
Collapse
|
22
|
Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, Idowu O, Li M, Shen C, Hu A, Haydon RC, Kang R, Mok J, Lee MJ, Luu HL, Shi LL. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis 2014; 1:87-105. [PMID: 25401122 PMCID: PMC4232216 DOI: 10.1016/j.gendis.2014.07.005] [Citation(s) in RCA: 725] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are a group of signaling molecules that belongs to the Transforming Growth Factor-β (TGF-β) superfamily of proteins. Initially discovered for their ability to induce bone formation, BMPs are now known to play crucial roles in all organ systems. BMPs are important in embryogenesis and development, and also in maintenance of adult tissue homeostasis. Mouse knockout models of various components of the BMP signaling pathway result in embryonic lethality or marked defects, highlighting the essential functions of BMPs. In this review, we first outline the basic aspects of BMP signaling and then focus on genetically manipulated mouse knockout models that have helped elucidate the role of BMPs in development. A significant portion of this review is devoted to the prominent human pathologies associated with dysregulated BMP signaling.
Collapse
Affiliation(s)
- Richard N. Wang
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jordan Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Youlin Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Min Qiao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Michael Peabody
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qian Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zhengjian Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Sahitya Denduluri
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Olumuyiwa Idowu
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Melissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Christine Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alan Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - James Mok
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue L. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
23
|
Endothelial cell FGF signaling is required for injury response but not for vascular homeostasis. Proc Natl Acad Sci U S A 2014; 111:13379-84. [PMID: 25139991 DOI: 10.1073/pnas.1324235111] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor receptors (FGFRs) and are exquisitely sensitive to FGF signals. However, whether the EC or another vascular cell type requires FGF signaling during development, homeostasis, and response to injury is not known. Here, we show that Flk1-Cre or Tie2-Cre mediated deletion of FGFR1 and FGFR2 (Fgfr1/2(Flk1-Cre) or Fgfr1/2(Tie2-Cre) mice), which results in deletion in endothelial and hematopoietic cells, is compatible with normal embryonic development. As adults, Fgfr1/2(Flk1-Cre) mice maintain normal blood pressure and vascular reactivity and integrity under homeostatic conditions. However, neovascularization after skin or eye injury was significantly impaired in both Fgfr1/2(Flk1-Cre) and Fgfr1/2(Tie2-Cre) mice, independent of either hematopoietic cell loss of FGFR1/2 or vascular endothelial growth factor receptor 2 (Vegfr2) haploinsufficiency. Also, impaired neovascularization was associated with delayed cutaneous wound healing. These findings reveal a key requirement for cell-autonomous EC FGFR signaling in injury-induced angiogenesis, but not for vascular homeostasis, identifying the EC FGFR signaling pathway as a target for diseases associated with aberrant vascular proliferation, such as age-related macular degeneration, and for modulating wound healing without the potential toxicity associated with direct manipulation of systemic FGF or VEGF activity.
Collapse
|
24
|
Kim JD, Lee HW, Jin SW. Diversity is in my veins: role of bone morphogenetic protein signaling during venous morphogenesis in zebrafish illustrates the heterogeneity within endothelial cells. Arterioscler Thromb Vasc Biol 2014; 34:1838-45. [PMID: 25060789 DOI: 10.1161/atvbaha.114.303219] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Endothelial cells are a highly diverse group of cells which display distinct cellular responses to exogenous stimuli. Although the aptly named vascular endothelial growth factor-A signaling pathway is hailed as the most important signaling input for endothelial cells, additional factors also participate in regulating diverse aspects of endothelial behaviors and functions. Given this heterogeneity, these additional factors seem to play a critical role in creating a custom-tailored environment to regulate behaviors and functions of distinct subgroups of endothelial cells. For instance, molecular cues that modulate morphogenesis of arterial vascular beds can be distinct from those that govern morphogenesis of venous vascular beds. Recently, we have found that bone morphogenetic protein signaling selectively promotes angiogenesis from venous vascular beds without eliciting similar responses from arterial vascular beds in zebrafish, indicating that bone morphogenetic protein signaling functions as a context-dependent regulator during vascular morphogenesis. In this review, we will provide an overview of the molecular mechanisms that underlie proangiogenic effects of bone morphogenetic protein signaling on venous vascular beds in the context of endothelial heterogeneity and suggest a more comprehensive picture of the molecular mechanisms of vascular morphogenesis during development.
Collapse
Affiliation(s)
- Jun-Dae Kim
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Heon-Woo Lee
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.)
| | - Suk-Won Jin
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (J.-D.K., H.W.L., S.-W.J.) and Department of Internal Medicine (J.-D.K., H.W.L., S.-W.J.), Yale University School of Medicine, New Haven, CT; and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea (S.-W.J.).
| |
Collapse
|
25
|
Endothelial Smad4 restrains the transition to hematopoietic progenitors via suppression of ERK activation. Blood 2014; 123:2161-71. [PMID: 24553180 DOI: 10.1182/blood-2013-09-526053] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In mouse mid-gestational embryos, definitive hematopoietic stem progenitor cells are derived directly from a very small proportion of the arterial endothelium. However, the physiological mechanisms restraining excessive endothelial-hematopoietic transition remain elusive. We show here that genetic deletion of Smad4 from the endothelium stage (using Tie2-Cre), but not from embryonic hematopoietic cells (using Vav-Cre), leads to a strikingly augmented emergence of intra-arterial hematopoietic clusters and an enhanced in vitro generation of hematopoietic progenitors, with no increase in the proliferation and survival of hematopoietic cluster cells. This finding indicates a temporally restricted negative effect of Smad4 on the endothelial to hematopoietic progenitor transition. Furthermore, the absence of endothelial Smad4 causes an increased expression of subaortic bone morphogenetic protein 4 and an activation of aortic extracellular signal-regulated kinase, thereby resulting in the excessive generation of blood cells. Collectively, our data for the first time identify a physiological suppressor that functions specifically during the transition of endothelial cells to hematopoietic progenitors and further suggest that endothelial Smad4 is a crucial modulator of the subaortic microenvironment that controls the hematopoietic fate of the aortic endothelium.
Collapse
|
26
|
Inai K, Burnside JL, Hoffman S, Toole BP, Sugi Y. BMP-2 induces versican and hyaluronan that contribute to post-EMT AV cushion cell migration. PLoS One 2013; 8:e77593. [PMID: 24147033 PMCID: PMC3795687 DOI: 10.1371/journal.pone.0077593] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/09/2013] [Indexed: 11/24/2022] Open
Abstract
Distal outgrowth and maturation of mesenchymalized endocardial cushions are critical morphogenetic events during post-EMT atrioventricular (AV) valvuloseptal morphogenesis. We explored the role of BMP-2 in the regulation of valvulogenic extracellular matrix (ECM) components, versican and hyaluronan (HA), and cell migration during post-EMT AV cushion distal outgrowth/expansion. We observed intense staining of versican and HA in AV cushion mesenchyme from the early cushion expansion stage, Hamburger and Hamilton (HH) stage-17 to the cushion maturation stage, HH stage-29 in the chick. Based on this expression pattern we examined the role of BMP-2 in regulating versican and HA using 3D AV cushion mesenchymal cell (CMC) aggregate cultures on hydrated collagen gels. BMP-2 induced versican expression and HA deposition as well as mRNA expression of versican and Has2 by CMCs in a dose dependent manner. Noggin, an antagonist of BMP, abolished BMP-2-induced versican and HA as well as mRNA expression of versican and Has2. We further examined whether BMP-2-promoted cell migration was associated with expression of versican and HA. BMP-2- promoted cell migration was significantly impaired by treatments with versican siRNA and HA oligomer. In conclusion, we provide evidence that BMP-2 induces expression of versican and HA by AV CMCs and that these ECM components contribute to BMP-2-induced CMC migration, indicating critical roles for BMP-2 in distal outgrowth/expansion of mesenchymalized AV cushions.
Collapse
Affiliation(s)
- Kei Inai
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Jessica L. Burnside
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Stanley Hoffman
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Bryan P. Toole
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology and Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
27
|
Jakobsson L, van Meeteren LA. Transforming growth factor β family members in regulation of vascular function: in the light of vascular conditional knockouts. Exp Cell Res 2013; 319:1264-70. [PMID: 23454603 DOI: 10.1016/j.yexcr.2013.02.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/12/2013] [Accepted: 02/13/2013] [Indexed: 10/27/2022]
Abstract
Blood vessels are composed of endothelial cells, mural cells (smooth muscle cells and pericytes) and their shared basement membrane. During embryonic development a multitude of signaling components orchestrate the formation of new vessels. The process is highly dependent on correct dosage, spacing and timing of these signaling molecules. As vessels mature some cascades remain active, albeit at very low levels, and may be reactivated upon demand. Members of the Transforming growth factor β (TGF-β) protein family are strongly engaged in developmental angiogenesis but are also regulators of vascular integrity in the adult. In humans various genetic alterations within this protein family cause vascular disorders, involving disintegration of vascular integrity. Here we summarize and discuss recent data gathered from conditional and endothelial cell specific genetic loss-of-function of members of the TGF-β family in the mouse.
Collapse
Affiliation(s)
- Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| | | |
Collapse
|
28
|
Harmelink C, Peng Y, DeBenedittis P, Chen H, Shou W, Jiao K. Myocardial Mycn is essential for mouse ventricular wall morphogenesis. Dev Biol 2013; 373:53-63. [PMID: 23063798 PMCID: PMC3508168 DOI: 10.1016/j.ydbio.2012.10.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 09/11/2012] [Accepted: 10/04/2012] [Indexed: 01/27/2023]
Abstract
MYCN is a highly conserved transcription factor with multifaceted roles in development and disease. Mutations in MYCN are associated with Feingold syndrome, a developmental disorder characterized in part by congenital heart defects. Mouse models have helped elucidate MYCN functions; however its cardiac-specific roles during development remain unclear. We employed a Cre/loxp strategy to uncover the specific activities of MYCN in the developing mouse myocardium. Myocardial deletion of Mycn resulted in a thin-myocardial wall defect with dramatically reduced trabeculation. The mutant heart defects strongly resemble the phenotype caused by disruption of BMP10 and Neuregulin-1 (NRG1) signaling pathways, two central mediators of myocardial wall development. Our further examination showed that expression of MYCN is regulated by both BMP and NRG1 signaling. The thin-wall defect in mutant hearts is caused by a reduction in both cell proliferation and cell size. MYCN promotes cardiomyocyte proliferation through regulating expression of cell cycle regulators (including CCND1, CCND2, and ID2) and promotes cardiomyocyte growth through regulating expression of p70S6K. In addition, expression of multiple sarcomere proteins is altered in Mycn myocardial-inactivation embryos, indicating its essential role for proper cardiomyocyte differentiation. In summary, Mycn acts downstream of BMP and NRG1 cardiogenic signaling pathways to promote normal myocardial wall morphogenesis.
Collapse
Affiliation(s)
- Cristina Harmelink
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Yin Peng
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Paige DeBenedittis
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Hanying Chen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Weinian Shou
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Kai Jiao
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
29
|
Marks-Bluth J, Pimanda JE. Cell signalling pathways that mediate haematopoietic stem cell specification. Int J Biochem Cell Biol 2012; 44:2175-84. [DOI: 10.1016/j.biocel.2012.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 01/27/2023]
|
30
|
Poirier O, Ciumas M, Eyries M, Montagne K, Nadaud S, Soubrier F. Inhibition of apelin expression by BMP signaling in endothelial cells. Am J Physiol Cell Physiol 2012; 303:C1139-45. [DOI: 10.1152/ajpcell.00168.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transforming growth factor-β/bone morphogenic protein (BMP) system is a major pathway for angiogenesis and is involved in hereditary vascular diseases. Here we report that the gene encoding the vasoactive and vascular cell growth-regulating peptide apelin is a target of the BMP pathway. We demonstrate that apelin expression is strongly downregulated by BMP in an endothelial cell line as well as in lung endothelial microvascular cells. We show that BMP signals through the BMPR2-SMAD pathway to downregulate apelin expression and that a transcriptional direct and indirect mechanism is required. The BMP-induced downregulation of apelin expression was found to be critical for hypoxia-induced growth of endothelial cells, because the growth inhibitory effect of BMP in this condition is suppressed by enforced expression of apelin. Thus, we describe an important link between a signaling pathway involved in angiogenesis and vascular diseases and a peptide regulating vascular homeostasis.
Collapse
Affiliation(s)
- Odette Poirier
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Mariana Ciumas
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Mélanie Eyries
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Kevin Montagne
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Sophie Nadaud
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Florent Soubrier
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
31
|
Kruithof BPT, Duim SN, Moerkamp AT, Goumans MJ. TGFβ and BMP signaling in cardiac cushion formation: lessons from mice and chicken. Differentiation 2012; 84:89-102. [PMID: 22656450 DOI: 10.1016/j.diff.2012.04.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/28/2012] [Accepted: 04/04/2012] [Indexed: 02/01/2023]
Abstract
Cardiac cushion formation is crucial for both valvular and septal development. Disruption in this process can lead to valvular and septal malformations, which constitute the largest part of congenital heart defects. One of the signaling pathways that is important for cushion formation is the TGFβ superfamily. The involvement of TGFβ and BMP signaling pathways in cardiac cushion formation has been intensively studied using chicken in vitro explant assays and in genetically modified mice. In this review, we will summarize and discuss the role of TGFβ and BMP signaling components in cardiac cushion formation.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | | | | | | |
Collapse
|
32
|
BMP signaling in vascular diseases. FEBS Lett 2012; 586:1993-2002. [DOI: 10.1016/j.febslet.2012.04.030] [Citation(s) in RCA: 217] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/05/2012] [Accepted: 04/17/2012] [Indexed: 12/24/2022]
|
33
|
Abstract
The early blood vessels of the embryo and yolk sac in mammals develop by aggregation of de novo-forming angioblasts into a primitive vascular plexus, which then undergoes a complex remodeling process. Angiogenesis is also important for disease progression in the adult. However, the precise molecular mechanism of vascular development remains unclear. It is therefore of great interest to determine which genes are specifically expressed in developing endothelial cells (ECs). Here, we used Flk1-deficient mouse embryos, which lack ECs, to perform a genome-wide survey for genes related to vascular development. We identified 184 genes that are highly enriched in developing ECs. The human orthologs of most of these genes were also expressed in HUVECs, and small interfering RNA knockdown experiments on 22 human orthologs showed that 6 of these genes play a role in tube formation by HUVECs. In addition, we created Arhgef15 knockout and RhoJ knockout mice by a gene-targeting method and found that Arhgef15 and RhoJ were important for neonatal retinal vascularization. Thus, the genes identified in our survey show high expression in ECs; further analysis of these genes should facilitate our understanding of the molecular mechanisms of vascular development in the mouse.
Collapse
|
34
|
ER71 specifies Flk-1+ hemangiogenic mesoderm by inhibiting cardiac mesoderm and Wnt signaling. Blood 2012; 119:3295-305. [PMID: 22343916 DOI: 10.1182/blood-2012-01-403766] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Two distinct types of Flk-1(+) mesoderm, hemangiogenic and cardiogenic, are thought to contribute to blood, vessel, and cardiac cell lineages. However, our understanding of how Flk-1(+) mesoderm is specified is currently limited. In the present study, we investigated whether ER71, an Ets transcription factor essential for hematopoietic and endothelial cell lineage development, could modulate the hemangiogenic or cardiogenic outcome of the Flk-1(+) mesoderm. We show that Flk-1(+) mesoderm can be divided into Flk-1(+)PDGFRα(-) hemangiogenic and Flk-1(+)PDGFRα(+) cardiogenic mesoderm. ER71-deficient embryonic stem cells produced only the Flk-1(+)PDGFRα(+) cardiogenic mesoderm, which generated SMCs and cardiomyocytes. Enforced ER71 expression in the wild-type embryonic stem cells skewed toward the Flk-1(+)PDGFRα(-) mesoderm formation, which generated hematopoietic and endothelial cells. Whereas hematopoietic and endothelial cell genes were positively regulated by ER71, cardiac and Wnt signaling pathway genes were negatively regulated by ER71. We show that ER71 could inhibit Wnt signaling in VE-cadherin-independent as well as VE-cadherin-dependent VE-cadherin/β-catenin/Flk-1 complex formation. Enforced β-catenin could rescue cardiogenic mesoderm in the context of ER71 overexpression. In contrast, ER71-deficient Flk-1(+) mesoderm displayed enhanced Wnt signaling, which was reduced by ER71 re-introduction. We provide the molecular basis for the antagonistic relationship between hemangiogenic and cardiogenic mesoderm specification by ER71 and Wnt signaling.
Collapse
|
35
|
Sugi Y, Kern MJ, Markwald RR, Burnside JL. Periostin Expression is Altered in Aortic Valves in Smad6 Mutant Mice. ACTA ACUST UNITED AC 2012; 1. [PMID: 25383261 PMCID: PMC4224111 DOI: 10.4172/2167-0897.1000101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Smad6 is known to predominantly inhibit BMP signaling by negatively regulating the BMP signaling process. Therefore, Smad6 mutation potentially provides an important genetic model for investigating the role of BMP signaling in vivo. Periostin is a 90-kDA secreted extracellular matrix (ECM) protein and implicated in cardiac valve progenitor cell differentiation, maturation and adult aortic valve calcification in mice. We have previously reported periostin expression patterns during AV valve development in mice. Because periostin can play critical roles in aortic valve interstitial cell differentiation and can be correlated with adult valve disease pathogenesis, in the present study we specifically focused on periostin expression during outflow tract (OT) development and its expression within the adult mouse valves. We previously reported that periostin expression in valve progenitor cells was altered by exogenously adding BMP-2 in culture. In this study, we investigated whether expression of periostin and other valvulogenic ECM proteins was altered in Smad6-mutant newborn mice in vivo. Periostin protein was localized within OT during embryonic development in mice. At embryonic day (ED) 13.5, robust periostin expression was detected within the developing pulmonary trunk and developing pulmonary and aortic valves. Periostin expression remained intense in pulmonary and aortic valves up to the adult stage. Our immunohistochemical and immunointensity analyses revealed that periostin expression was significantly reduced in the aortic valves in Smad6−/− neonatal hearts. Versican expression was also significantly reduced in Smad6−/− aortic valves, whereas, hyaluronan deposition was not significantly altered in the Smad6−/− neonatal valves. Expression of periostin and versican was less prominently affected in AV valves compared to the aortic valves, suggesting that a cell lineage/origin-dependent response to regulatory molecules may play a critical role in valve interstitial cell development and ECM protein expression.
Collapse
Affiliation(s)
- Yukiko Sugi
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Michael J Kern
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Roger R Markwald
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| | - Jessica L Burnside
- Department of Regenerative Medicine and Cell Biology, Cardiovascular Developmental Biology Center, Medical University of South Carolina, USA
| |
Collapse
|
36
|
Townsend TA, Robinson JY, How T, DeLaughter DM, Blobe GC, Barnett JV. Endocardial cell epithelial-mesenchymal transformation requires Type III TGFβ receptor interaction with GIPC. Cell Signal 2011; 24:247-56. [PMID: 21945156 DOI: 10.1016/j.cellsig.2011.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 09/02/2011] [Accepted: 09/05/2011] [Indexed: 12/19/2022]
Abstract
An early event in heart valve formation is the epithelial-mesenchymal transformation (EMT) of a subpopulation of endothelial cells in specific regions of the heart tube, the endocardial cushions. The Type III TGFβ receptor (TGFβR3) is required for TGFβ2- or BMP-2-stimulated EMT in atrioventricular endocardial cushion (AVC) explants in vitro but the mediators downstream of TGFβR3 are not well described. Using AVC and ventricular explants as an in vitro assay, we found an absolute requirement for specific TGFβR3 cytoplasmic residues, GAIP-interacting protein, C terminus (GIPC), and specific Activin Receptor-Like Kinases (ALK)s for TGFβR3-mediated EMT when stimulated by TGFβ2 or BMP-2. The introduction of TGFβR3 into nontransforming ventricular endocardial cells, followed by the addition of either TGFβ2 or BMP-2, results in EMT. TGFβR3 lacking the entire cytoplasmic domain, or only the 3C-terminal amino acids that are required to bind GIPC, fails to support EMT in response to TGFβ2 or BMP-2. Overexpression of GIPC in AVC endocardial cells enhanced EMT while siRNA-mediated silencing of GIPC in ventricular cells overexpressing TGFβR3 significantly inhibited EMT. Targeting of specific ALKs by siRNA revealed that TGFβR3-mediated EMT requires ALK2 and ALK3, in addition to ALK5, but not ALK4 or ALK6. Taken together, these data identify GIPC, ALK2, ALK3, and ALK5 as signaling components required for TGFβR3-mediated endothelial cell EMT.
Collapse
Affiliation(s)
- Todd A Townsend
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Lopes M, Goupille O, Saint Cloment C, Lallemand Y, Cumano A, Robert B. Msx genes define a population of mural cell precursors required for head blood vessel maturation. Development 2011; 138:3055-66. [PMID: 21693521 DOI: 10.1242/dev.063214] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Vessels are primarily formed from an inner endothelial layer that is secondarily covered by mural cells, namely vascular smooth muscle cells (VSMCs) in arteries and veins and pericytes in capillaries and veinules. We previously showed that, in the mouse embryo, Msx1(lacZ) and Msx2(lacZ) are expressed in mural cells and in a few endothelial cells. To unravel the role of Msx genes in vascular development, we have inactivated the two Msx genes specifically in mural cells by combining the Msx1(lacZ), Msx2(lox) and Sm22α-Cre alleles. Optical projection tomography demonstrated abnormal branching of the cephalic vessels in E11.5 mutant embryos. The carotid and vertebral arteries showed an increase in caliber that was related to reduced vascular smooth muscle coverage. Taking advantage of a newly constructed Msx1(CreERT2) allele, we demonstrated by lineage tracing that the primary defect lies in a population of VSMC precursors. The abnormal phenotype that ensues is a consequence of impaired BMP signaling in the VSMC precursors that leads to downregulation of the metalloprotease 2 (Mmp2) and Mmp9 genes, which are essential for cell migration and integration into the mural layer. Improper coverage by VSMCs secondarily leads to incomplete maturation of the endothelial layer. Our results demonstrate that both Msx1 and Msx2 are required for the recruitment of a population of neural crest-derived VSMCs.
Collapse
Affiliation(s)
- Miguel Lopes
- Institut Pasteur, Génétique Moléculaire de la Morphogenèse, CNRS URA 2578, Paris, France
| | | | | | | | | | | |
Collapse
|
38
|
Song L, Zhao M, Wu B, Zhou B, Wang Q, Jiao K. Cell autonomous requirement of endocardial Smad4 during atrioventricular cushion development in mouse embryos. Dev Dyn 2011; 240:211-20. [PMID: 21089072 DOI: 10.1002/dvdy.22493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrioventricular (AV) cushions are the precursors of AV septum and valves. In this study, we examined roles of Smad4 during AV cushion development using a conditional gene inactivation approach. We found that endothelial/endocardial inactivation of Smad4 led to the hypocellular AV cushion defect and that both reduced cell proliferation and increased apoptosis contributed to the defect. Expression of multiple genes critical for cushion development was down-regulated in mutant embryos. In collagen gel assays, the number of mesenchymal cells formed is significantly reduced in mutant AV explants compared to that in control explants, suggesting that the reduction of cushion mesenchyme formation in mutants is unlikely secondary to their gross vasculature abnormalities. Using a previously developed immortal endocardial cell line, we showed that Smad4 is required for BMP signaling- stimulated upregulation of Tbx20 and Gata4. Therefore, our data collectively support the cell-autonomous requirement of endocardial Smad4 in regulating AV cushion development.
Collapse
Affiliation(s)
- Langying Song
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
39
|
Transcription factor genes Smad4 and Gata4 cooperatively regulate cardiac valve development. [corrected]. Proc Natl Acad Sci U S A 2011; 108:4006-11. [PMID: 21330551 DOI: 10.1073/pnas.1019025108] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We report that the dominant human missense mutations G303E and G296S in GATA4, a cardiac-specific transcription factor gene, cause atrioventricular septal defects and valve abnormalities by disrupting a signaling cascade involved in endocardial cushion development. These GATA4 missense mutations, but not a mutation causing secundum atrial septal defects (S52F), demonstrated impaired protein interactions with SMAD4, a transcription factor required for canonical bone morphogenetic protein/transforming growth factor-β (BMP/TGF-β) signaling. Gata4 and Smad4 genetically interact in vivo: atrioventricular septal defects result from endothelial-specific Gata4 and Smad4 compound haploinsufficiency. Endothelial-specific knockout of Smad4 caused an absence of valve-forming activity: Smad4-deficient endocardium was associated with acellular endocardial cushions, absent epithelial-to-mesenchymal transformation, reduced endocardial proliferation, and loss of Id2 expression in valve-forming regions. We show that Gata4 and Smad4 cooperatively activated the Id2 promoter, that human GATA4 mutations abrogated this activity, and that Id2 deficiency in mice could cause atrioventricular septal defects. We suggest that one determinant of the phenotypic spectrum caused by human GATA4 mutations is differential effects on GATA4/SMAD4 interactions required for endocardial cushion development.
Collapse
|
40
|
Abstract
The myocardium of the heart is composed of multiple highly specialized myocardial lineages, including those of the ventricular and atrial myocardium, and the specialized conduction system. Specification and maturation of each of these lineages during heart development is a highly ordered, ongoing process involving multiple signaling pathways and their intersection with transcriptional regulatory networks. Here, we attempt to summarize and compare much of what we know about specification and maturation of myocardial lineages from studies in several different vertebrate model systems. To date, most research has focused on early specification, and although there is still more to learn about early specification, less is known about factors that promote subsequent maturation of myocardial lineages required to build the functioning adult heart.
Collapse
Affiliation(s)
- Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla CA 92093, USA.
| | | | | | | |
Collapse
|
41
|
Nie X, Brown CB, Wang Q, Jiao K. Inactivation of Bmp4 from the Tbx1 expression domain causes abnormal pharyngeal arch artery and cardiac outflow tract remodeling. Cells Tissues Organs 2010; 193:393-403. [PMID: 21123999 DOI: 10.1159/000321170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2010] [Indexed: 11/19/2022] Open
Abstract
Maldevelopment of outflow tract and aortic arch arteries is among the most common forms of human congenital heart diseases. Both Bmp4 and Tbx1 are known to play critical roles during cardiovascular development. Expression of these two genes partially overlaps in pharyngeal arch areas in mouse embryos. In this study, we applied a conditional gene inactivation approach to test the hypothesis that Bmp4 expressed from the Tbx1 expression domain plays a critical role for normal development of outflow tract and pharyngeal arch arteries. We showed that inactivation of Bmp4 from Tbx1-expressing cells leads to the spectrum of deformities resembling the cardiovascular defects observed in human DiGeorge syndrome patients. Inactivation of Bmp4 from the Tbx1 expression domain did not cause patterning defects, but affected remodeling of outflow tract and pharyngeal arch arteries. Our further examination revealed that Bmp4 is required for normal recruitment/differentiation of smooth muscle cells surrounding the PAA4 and survival of outflow tract cushion mesenchymal cells.
Collapse
Affiliation(s)
- Xuguang Nie
- Division of Research, Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
42
|
Bone Morphogenic Protein-4 Impairs Endothelial Function Through Oxidative Stress–Dependent Cyclooxygenase-2 Upregulation. Circ Res 2010; 107:984-91. [DOI: 10.1161/circresaha.110.222794] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
Bone morphogenic protein (BMP)4 can stimulate superoxide production and exert proinflammatory effects on the endothelium. The underlying mechanisms of how BMP4 mediates endothelial dysfunction and hypertension remain elusive.
Objective:
To elucidate the cellular pathways by which BMP4-induced endothelial dysfunction is mediated through oxidative stress–dependent upregulation of cyclooxygenase (COX)-2.
Methods and Results:
Impaired endothelium-dependent relaxations, exaggerated endothelium-dependent contractions, and reactive oxygen species (ROS) production were observed in BMP4-treated mouse aortae, which were prevented by the BMP4 antagonist noggin. Pharmacological inhibition with thromboxane prostanoid receptor antagonist or COX-2 but not COX-1 inhibitor prevented BMP4-induced endothelial dysfunction, which was further confirmed with the use of
COX-1
−/−
or
COX-2
−/−
mice. Noggin and knockdown of BMP receptor 1A abolished endothelium-dependent contractions and COX-2 upregulation in BMP4-treated aortae. Apocynin and tempol treatment were effective in restoring endothelium-dependent relaxations, preventing endothelium-dependent contractions and eliminating ROS overproduction and COX-2 overexpression in BMP4-treated aortae. BMP4 increased p38 mitogen-activated protein kinase (MAPK) activity through a ROS-sensitive mechanism and p38 MAPK inhibitor prevented BMP4-induced endothelial dysfunction. COX-2 inhibition blocked the effect of BMP4 without affecting BMP4-induced ROS overproduction and COX-2 upregulation. Importantly, renal arteries from hypertensive rats and humans showed higher levels of COX-2 and BMP4 accompanied by endothelial dysfunction.
Conclusions:
We show for the first time that ROS serve as a pathological link between BMP4 stimulation and the downstream COX-2 upregulation in endothelial cells, leading to endothelial dysfunction through ROS-dependent p38 MAPK activation. This BMP4/ROS/COX-2 cascade is important in the maintenance of endothelial dysfunction in hypertension.
Collapse
|
43
|
Cannon JE, Upton PD, Smith JC, Morrell NW. Intersegmental vessel formation in zebrafish: requirement for VEGF but not BMP signalling revealed by selective and non-selective BMP antagonists. Br J Pharmacol 2010; 161:140-9. [PMID: 20718746 PMCID: PMC2962823 DOI: 10.1111/j.1476-5381.2010.00871.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 03/26/2010] [Accepted: 04/11/2010] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Bone morphogenetic proteins (BMPs) were first identified through their role in inducing bone and cartilage formation, but many other important functions have since been ascribed to BMPs, including dorsoventral patterning, angiogenesis and tissue homeostasis. Using dorsomorphin and LDN193189, selective small molecule inhibitors of BMP signalling, we investigated the role of BMP signalling in early vascular patterning in zebrafish. EXPERIMENTAL APPROACH The effects of dorsomorphin and LDN193189 on vascular endothelial growth factor-a (VEGF) and BMP signalling in developing zebrafish and in human pulmonary artery endothelial cells were determined using confocal microscopy, Western blotting and quantitative PCR. KEY RESULTS We showed that dorsomorphin, similar to the VEGF inhibitor SU5416, strongly inhibits intersegmental vessel formation in zebrafish and that this is due to inhibition of VEGF activation of VEGF receptor 2 (VEGFR2), leading to reduced VEGF-induced phospho-ERK (extracellular regulated kinase) 1/2 and VEGF target gene transcription. These effects occurred at concentrations of dorsomorphin that block BMP signalling. We also showed that LDN193189, an analogue of dorsomorphin, more potently blocks BMP signalling but has no effect on VEGF signalling in zebrafish and does not disrupt early vascular patterning. CONCLUSIONS AND IMPLICATIONS Dorsomorphin inhibits both BMP and VEGF signalling, whereas LDN193189 is a more selective BMP antagonist. Results obtained in cardiovascular studies using dorsomorphin need to be interpreted with caution, and use of LDN193189 would be preferable due to its selectivity. Our data also suggest that BMP signalling is dispensable for early patterning of intersegmental vessels in zebrafish.
Collapse
Affiliation(s)
- JE Cannon
- Wellcome Trust/CR-UK Gurdon Institute and Department of Zoology, The University of CambridgeCambridge, UK
- University of Cambridge, Department of MedicineCambridge, UK
| | - PD Upton
- University of Cambridge, Department of MedicineCambridge, UK
| | - JC Smith
- MRC National Institute for Medical ResearchThe Ridgeway, Mill Hill, London, UK
| | - NW Morrell
- University of Cambridge, Department of MedicineCambridge, UK
| |
Collapse
|
44
|
Sun M, Forsman C, Sergi C, Gopalakrishnan R, O’Connor MB, Petryk A. The expression of twisted gastrulation in postnatal mouse brain and functional implications. Neuroscience 2010; 169:920-931. [PMID: 20493240 PMCID: PMC2971674 DOI: 10.1016/j.neuroscience.2010.05.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 05/11/2010] [Accepted: 05/12/2010] [Indexed: 12/28/2022]
Abstract
Twisted gastrulation (TWSG1), an extracellular regulator of bone morphogenetic protein (BMP) signaling, is critical for embryonic brain development. Mice deficient in TWSG1 have abnormal forebrain development manifesting as holoprosencephaly. The expression and potential roles of TWSG1 in postnatal brain development are less well understood. We show that Twsg1 is expressed in the adult mouse brain in the choroid plexus (CP), hippocampus, and other regions, with the strongest expression observed in CP. TWSG1 was also detected in a human fetal brain at mid-gestation, with highest levels in the epithelium of CP. Bmp1, Bmp2, Bmp4-Bmp7 as well as BmprIA and BmprII, but not BmprIB, were expressed in CP. BMP antagonists Chordin (Chrd) and Noggin were not detected in CP, however Chrd-like 1 and brain-specific Chrd-like (Brorin) were expressed. Electrophysiological study of synaptic plasticity revealed normal paired-pulse facilitation and long-term potentiation in the CA1 region of hippocampus in Twsg1(-/-) mice. Among the homozygous mutants that survive beyond the first 2 weeks, the prevalence of hydrocephalus was 4.3%, compared to 1.5% in a wild type colony (P=0.0133) between 3 and 10 weeks of life. We detected a high level of BMP signaling in CP in wild type adult mice that was 17-fold higher than in the hippocampus (P=0.005). In contrast, transforming growth factor beta (TGFbeta) signaling was predominant in the hippocampus. Both BMP signaling and the expression of BMP downstream targets Msx1 and Msx2 were reduced in CP in Twsg1(-/-) mice. In summary, we show that Twsg1 is expressed in the adult mouse and human fetal CP. We also show that BMP is a branch of TGFbeta superfamily that is dominant in CP. This presents an interesting avenue for future research in light of the novel roles of CP in neural progenitor differentiation and neuronal repair, especially since TWSG1 appears to be the main regulator of BMP present in CP.
Collapse
Affiliation(s)
- Mu Sun
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455-0356, USA
- Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455-0356, USA
| | - Cynthia Forsman
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455-0356, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455-0356, USA
| | - Consolato Sergi
- Department of Laboratory Medicine & Pathology, University of Alberta, Alberta, Canada T6G 2B7
- Institute of Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Rajaram Gopalakrishnan
- Diagnostic/Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455-0356, USA
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455-0356, USA
- Howard Hughes Medical Institute, University of Minnesota, Minneapolis, MN 55455-0356, USA
| | - Anna Petryk
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455-0356, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455-0356, USA
| |
Collapse
|
45
|
Abstract
Genetic and functional studies indicate that common components of the bone morphogenetic protein (BMP) signaling pathway play critical roles in regulating vascular development in the embryo and in promoting vascular homeostasis and disease in the adult. However, discrepancies between in vitro and in vivo findings and distinct functional properties of the BMP signaling pathway in different vascular beds, have led to controversies in the field that have been difficult to reconcile. This review attempts to clarify some of these issues by providing an up to date overview of the biology and genetics of BMP signaling relevant to the intact vasculature.
Collapse
|
46
|
Lan Y, Yang X. The role of Smad signaling in vascular and hematopoietic development revealed by studies using genetic mouse models. SCIENCE CHINA-LIFE SCIENCES 2010; 53:485-9. [DOI: 10.1007/s11427-010-0087-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 11/01/2009] [Indexed: 01/12/2023]
|
47
|
|
48
|
Uchimura T, Komatsu Y, Tanaka M, McCann KL, Mishina Y. Bmp2 and Bmp4 genetically interact to support multiple aspects of mouse development including functional heart development. Genesis 2009; 47:374-84. [PMID: 19391114 DOI: 10.1002/dvg.20511] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Bone morphogenetic proteins (BMPs) have multiple roles during embryogenesis. Current data indicate that the dosage of BMPs is tightly regulated for normal development in mice. Since Bmp2 or Bmp4 homozygous mutant mice show early embryonic lethality, we generated compound heterozygous mice for Bmp2 and Bmp4 to explore the impact of lowered dosage of these BMP ligands. Genotyping pups bred between Bmp2 and Bmp4 heterozygous mice revealed that the ratio of adult compound heterozygous mice for Bmp2 and Bmp4 is much lower than expected. During embryogenesis, the compound heterozygous embryos showed several abnormalities, including defects in eye formation, body wall closure defects, and ventricular septal defects (VSD) in the heart. However, the ratio of the compound heterozygous embryos was the same as expected. Caesarean sections at E18.5 revealed that half of the compound heterozygotes died soon after birth, and the majority of the dead individuals exhibited VSD. Survivors were able to grow to adults, but their body weight was significantly lower than control littermates. They demonstrated progressive abnormalities in the heart, eventually showing a branched leaflet in atrioventricular valves. These results suggest that the dosage of both BMP2 and 4 is critical for functional heart formation during embryogenesis and after birth.
Collapse
Affiliation(s)
- Takashi Uchimura
- Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | | | | | | | | |
Collapse
|
49
|
Nomura-Kitabayashi A, Phoon CKL, Kishigami S, Rosenthal J, Yamauchi Y, Abe K, Yamamura KI, Samtani R, Lo CW, Mishina Y. Outflow tract cushions perform a critical valve-like function in the early embryonic heart requiring BMPRIA-mediated signaling in cardiac neural crest. Am J Physiol Heart Circ Physiol 2009; 297:H1617-28. [PMID: 19717734 DOI: 10.1152/ajpheart.00304.2009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Neural crest-specific ablation of BMP type IA receptor (BMPRIA) causes embryonic lethality by embryonic day (E) 12.5, and this was previously postulated to arise from a myocardial defect related to signaling by a small population of cardiac neural crest cells (cNCC) in the epicardium. However, as BMP signaling via cNCC is also required for proper development of the outflow tract cushions, precursors to the semilunar valves, a plausible alternate or additional hypothesis is that heart failure may result from an outflow tract cushion defect. To investigate whether the outflow tract cushions may serve as dynamic valves in regulating hemodynamic function in the early embryo, in this study we used noninvasive ultrasound biomicroscopy-Doppler imaging to quantitatively assess hemodynamic function in mouse embryos with P0-Cre transgene mediated neural crest ablation of Bmpr1a (P0 mutants). Similar to previous studies, the neural crest-deleted Bmpr1a P0 mutants died at approximately E12.5, exhibiting persistent truncus arteriosus, thinned myocardium, and congestive heart failure. Surprisingly, our ultrasound analyses showed normal contractile indices, heart rate, and atrioventricular conduction in the P0 mutants. However, reversed diastolic arterial blood flow was detected as early as E11.5, with cardiovascular insufficiency and death rapidly ensuing by E12.5. Quantitative computed tomography showed thinning of the outflow cushions, and this was associated with a marked reduction in cell proliferation. These results suggest BMP signaling to cNCC is required for growth of the outflow tract cushions. This study provides definitive evidence that the outflow cushions perform a valve-like function critical for survival of the early mouse embryo.
Collapse
Affiliation(s)
- Aya Nomura-Kitabayashi
- Laboratory of Reproductive and Developmental Toxicology, National Institutes of Environmental Health Science, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Nomura-Kitabayashi A, Anderson GA, Sleep G, Mena J, Karabegovic A, Karamath S, Letarte M, Puri MC. Endoglin is dispensable for angiogenesis, but required for endocardial cushion formation in the midgestation mouse embryo. Dev Biol 2009; 335:66-77. [PMID: 19703439 DOI: 10.1016/j.ydbio.2009.08.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 07/27/2009] [Accepted: 08/15/2009] [Indexed: 11/16/2022]
Abstract
Vascular patterning depends on precisely coordinated timing of endothelial cell differentiation and onset of cardiac function. Endoglin is a transmembrane receptor for members of the TGF-beta superfamily that is expressed on endothelial cells from early embryonic gestation to adult life. Heterozygous loss of function mutations in human ENDOGLIN cause Hereditary Hemorrhagic Telangiectasia Type 1, a vascular disorder characterized by arteriovenous malformations that lead to hemorrhage and stroke. Endoglin null mice die in embryogenesis with numerous lesions in the cardiovascular tree including incomplete yolk sac vessel branching and remodeling, vessel dilation, hemorrhage and abnormal cardiac morphogenesis. Since defects in multiple cardiovascular tissues confound interpretations of these observations, we performed in vivo chimeric rescue analysis using Endoglin null embryonic stem cells. We demonstrate that Endoglin is required cell autonomously for endocardial to mesenchymal transition during formation of the endocardial cushions. Endoglin null cells contribute widely to endothelium in chimeric embryos rescued from cardiac development defects, indicating that Endoglin is dispensable for angiogenesis and vascular remodeling in the midgestation embryo, but is required for early patterning of the heart.
Collapse
Affiliation(s)
- Aya Nomura-Kitabayashi
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada M4N-3M5
| | | | | | | | | | | | | | | |
Collapse
|