1
|
Wolton M, Davey MG, Dietrich S. At early stages of heart development, the first and second heart fields are a continuum of lateral head mesoderm-derived, cardiogenic cells. Dev Biol 2025; 520:200-223. [PMID: 39848483 DOI: 10.1016/j.ydbio.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Pioneering work in the chicken established that the initial development of the heart consists of two stages: the quick assembly of a beating heart, followed by the recruitment of cells from adjacent tissues to deliver the mature in-and outflow tract. Cells to build the primitive heart were dubbed the first heart field (FHF) cells, cells to be recruited later the second heart field (SHF) cells. The current view is that these cells represent distinct, maybe even pre-determined lineages. However, it is still unclear where exactly FHF and SHF are located at different stages of development, and whether there is a sharp boundary or rather an overlap between the two. It is also unclear whether both FHF cells and SHF cells originate from the lateral head mesoderm (LHM), whether the paraxial head mesoderm (PHM) contributes to the SHF, and where the LHM-PHM boundary might be. To investigate this problem, we exploited the size, ease of access and exquisite anatomy of the chicken embryo and used traditional strategies as well as newly developed transgenic lines to trace the location of cardiogenic fields and boundaries from the time the first heart-markers are expressed to the time SHF cell recruitment ceases. Our work shows that both FHF and SHF stem from the LHM. We also found that FHF and SHF lack a distinct anatomical boundary. Rather, FHF and SHF are a continuum, and the recruitment of cells into the heart is a chance event depending on morphogenetic movements, the position of cells within the moving tissues, the separation of the somatic and splanchnic LHM, and the separation of the heart from the splanchnic subpharyngeal mesoderm during heart-looping. Reconciling our and previous studies we propose that first and second heart field precursors are specified but not determined, thus relying on morphogenetic processes and local environments to realise their cardiogenic potential.
Collapse
Affiliation(s)
- Matthew Wolton
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK
| | - Megan G Davey
- Functional Genetics, The Roslin Institute, The Royal (Dick) School of Veterinary Studies, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Susanne Dietrich
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
2
|
Alzamrooni A, Mendes Vieira P, Murciano N, Wolton M, Schubert FR, Robson SC, Dietrich S. Cardiac competence of the paraxial head mesoderm fades concomitant with a shift towards the head skeletal muscle programme. Dev Biol 2023; 501:39-59. [PMID: 37301464 DOI: 10.1016/j.ydbio.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
The vertebrate head mesoderm provides the heart, the great vessels, some smooth and most head skeletal muscle, in addition to parts of the skull. It has been speculated that the ability to generate cardiac and smooth muscle is the evolutionary ground-state of the tissue. However, whether indeed the entire head mesoderm has generic cardiac competence, how long this may last, and what happens as cardiac competence fades, is not clear. Bone morphogenetic proteins (Bmps) are known to promote cardiogenesis. Using 41 different marker genes in the chicken embryo, we show that the paraxial head mesoderm that normally does not engage in cardiogenesis has the ability to respond to Bmp for a long time. However, Bmp signals are interpreted differently at different time points. Up to early head fold stages, the paraxial head mesoderm is able to read Bmps as signal to engage in the cardiac programme; the ability to upregulate smooth muscle markers is retained slightly longer. Notably, as cardiac competence fades, Bmp promotes the head skeletal muscle programme instead. The switch from cardiac to skeletal muscle competence is Wnt-independent as Wnt caudalises the head mesoderm and also suppresses Msc-inducing Bmp provided by the prechordal plate, thus suppressing both the cardiac and the head skeletal muscle programmes. Our study for the first time suggests a specific transition state in the embryo when cardiac competence is replaced by skeletal muscle competence. It sets the stage to unravel the cardiac-skeletal muscle antagonism that is known to partially collapse in heart failure.
Collapse
Affiliation(s)
- Afnan Alzamrooni
- Institute of Biological and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Petra Mendes Vieira
- Institute of Biological and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Nicoletta Murciano
- Institute of Biological and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK; Nanion Technologies GmbH, Ganghoferstr. 70A, DE - 80339, München, Germany; Saarland University, Theoretical Medicine and Biosciences, Kirrbergerstr. 100, DE - 66424, Homburg, Germany
| | - Matthew Wolton
- Institute of Biological and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Frank R Schubert
- Institute of Biological and Biomedical Sciences, School of Biological Sciences, University of Portsmouth, Portsmouth, UK
| | - Samuel C Robson
- Institute of Biological and Biomedical Sciences, Faculty of Science & Health, University of Portsmouth, Portsmouth, UK
| | - Susanne Dietrich
- Institute of Biological and Biomedical Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
3
|
Bragança J, Pinto R, Silva B, Marques N, Leitão HS, Fernandes MT. Charting the Path: Navigating Embryonic Development to Potentially Safeguard against Congenital Heart Defects. J Pers Med 2023; 13:1263. [PMID: 37623513 PMCID: PMC10455635 DOI: 10.3390/jpm13081263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Congenital heart diseases (CHDs) are structural or functional defects present at birth due to improper heart development. Current therapeutic approaches to treating severe CHDs are primarily palliative surgical interventions during the peri- or prenatal stages, when the heart has fully developed from faulty embryogenesis. However, earlier interventions during embryonic development have the potential for better outcomes, as demonstrated by fetal cardiac interventions performed in utero, which have shown improved neonatal and prenatal survival rates, as well as reduced lifelong morbidity. Extensive research on heart development has identified key steps, cellular players, and the intricate network of signaling pathways and transcription factors governing cardiogenesis. Additionally, some reports have indicated that certain adverse genetic and environmental conditions leading to heart malformations and embryonic death may be amendable through the activation of alternative mechanisms. This review first highlights key molecular and cellular processes involved in heart development. Subsequently, it explores the potential for future therapeutic strategies, targeting early embryonic stages, to prevent CHDs, through the delivery of biomolecules or exosomes to compensate for faulty cardiogenic mechanisms. Implementing such non-surgical interventions during early gestation may offer a prophylactic approach toward reducing the occurrence and severity of CHDs.
Collapse
Affiliation(s)
- José Bragança
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Rute Pinto
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Bárbara Silva
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Nuno Marques
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Helena S. Leitão
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center-Research Institute (ABC-RI), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
- School of Health, University of Algarve Campus Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
4
|
Park H, Song G, Hong T, An G, Park S, Lim W. Exposure to the herbicide fluridone induces cardiovascular toxicity in early developmental stages of zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 867:161535. [PMID: 36638995 DOI: 10.1016/j.scitotenv.2023.161535] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 06/17/2023]
Abstract
Fluridone is a systemic herbicide used to control a range of invasive aquatic plants in irrigation systems, lake, and reservoirs. Since aquatic herbicides are more likely to have a hazardous impact on ecosystems than terrestrially applied herbicides, a risk assessment is needed to determine whether to expand or limit their use. The aim of this study was to investigate the developmental toxicity of fluridone using zebrafish. Diverse toxicological results were observed for the sub-lethal endpoints, including lack of hatching, reduced heartbeat and disturbed blood circulation through dysmorphic heart, and edema formation. Abnormal apoptosis was observed in the brain and yolk sac of fluridone-exposed larvae. A computational analysis was used to predict chemical properties in non-target organisms and revealed that fluridone was highly relevant in the cardiovascular system. Double transgenic zebrafish (fli1a:EGFP;cmlc2:dsRed) were used to evaluate the effects of fluridone on the cardiovascular system during embryonic development. Ectopic growth of sub-intestinal vessels and sprouting angiogenesis in the hindbrain region were highly inhibited. Additionally, essential genes involved in the VEGF signaling and heart development were differentially expressed in dose-dependent manner. Collectively, our toxicological findings in fluridone exposure highlight defects in the cardiovascular development causing embryonic lethality that could damage aquatic communities and natural ecosystems.
Collapse
Affiliation(s)
- Hahyun Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Sunwoo Park
- Department of Plant & Biomaterials Science, Gyeongsang National University, Jinju-si, Gyeongnam 52725, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
5
|
Hack JM, Anwar NZ, Jackson JG, Furth ME, Varner VD. Quantifying endodermal strains during heart tube formation in the developing chicken embryo. J Biomech 2023; 149:111481. [PMID: 36787674 PMCID: PMC10163833 DOI: 10.1016/j.jbiomech.2023.111481] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/17/2022] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
Abstract
In the early avian embryo, the developing heart forms when bilateral fields of cardiac progenitor cells, which reside in the lateral plate mesoderm, move toward the embryonic midline, and fuse above the anterior intestinal portal (AIP) to form a straight, muscle-wrapped tube. During this process, the precardiac mesoderm remains in close contact with the underlying endoderm. Previous work has shown that the endoderm around the AIP actively contracts to pull the cardiac progenitors toward the midline. The morphogenetic deformations associated with this endodermal convergence, however, remain unclear, as do the signaling pathways that might regulate this process. Here, we fluorescently labeled populations of endodermal cells in early chicken embryos and tracked their motion during heart tube formation to compute time-varying strains along the anterior endoderm. We then determined how the computed endodermal strain distributions are affected by the pharmacological inhibition of either myosin II or fibroblast growth factor (FGF) signaling. Our data indicate that a mediolateral gradient in endodermal shortening is present around the AIP, as well as substantial convergence and extension movements both anterior and lateral to the AIP. These active endodermal deformations are disrupted if either actomyosin contractility or FGF signaling are inhibited pharmacologically. Taken together, these results demonstrate how active deformations along the anterior endoderm contribute to heart tube formation within the developing embryo.
Collapse
Affiliation(s)
- Joshua M Hack
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Nareen Z Anwar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - John G Jackson
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Meagan E Furth
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Victor D Varner
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States; Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
6
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
7
|
Huo Q, Li Z, Chen S, Wang J, Li J, Xie N. VWCE as a potential biomarker associated with immune infiltrates in breast cancer. Cancer Cell Int 2021; 21:272. [PMID: 34020650 PMCID: PMC8140436 DOI: 10.1186/s12935-021-01955-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/27/2021] [Indexed: 11/10/2022] Open
Abstract
PURPOSE Von Willebrand Factor C and EGF Domains (VWCE) is an important gene that regulates cell adhesion, migration, and interaction. However, the correlation between VWCE expression and immune infiltrating in breast cancer remain unclear. In this study, we investigated the correlation between VWCE expression and immune infiltration levels in breast cancer. METHODS The expression of VWCE was analyzed by the tumor immune estimation resource (TIMER) and DriverDB databases. Furthermore, genes co-expressed with VWCE and gene ontology (GO) enrichment analysis were investigated by the STRING and Enrichr web servers. Also, we performed the single nucleotide variation (SNV), copy number variation (CNV), and pathway activity analysis through GSCALite. Subsequently, the relationship between VWCE expression and tumor immunity was analyzed by TIMER and TISIDB databases, and further verified the results using Quantitative Real-Time PCR (RT-PCR), Western blotting, and immunohistochemistry. RESULTS The results showed that the expression of VWCE mRNA in breast cancer tissue was significantly lower than that in normal tissues. We found that the expression level of VWCE was associated with subtypes, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) status of breast cancer patients, but there was no significant difference in the expression of VWCE was found in age and nodal status. Further analyses indicated that VWCE was correlated with the activation or inhibition of multiple oncogenic pathways. Additionally, VWCE expression was negatively correlated with the expression of STAT1 (Th1 marker, r = - 0.12, p = 6e-05), but positively correlated with the expression of MS4A4A (r = 0.28, p = 0). These results suggested that the expression of VWCE was correlated with immune infiltration levels of Th1 and M2 macrophage in breast cancer. CONCLUSIONS In our study, VWCE expression was associated with a better prognosis and was immune infiltration in breast cancer. These findings demonstrate that VWCE is a potential prognostic biomarker and correlated with tumor immune cell infiltration, and maybe a promising therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Qin Huo
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen, 518035, China
| | - Zhenwei Li
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen, 518035, China
| | - Siqi Chen
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen, 518035, China
| | - Juan Wang
- Department of Clinical Medicine , University of South China , Hengyang , 421001 , China
| | - Jiaying Li
- Department of Clinical Medicine , University of South China , Hengyang , 421001 , China
| | - Ni Xie
- Biobank, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University , Shenzhen, 518035, China.
| |
Collapse
|
8
|
Characterising open chromatin in chick embryos identifies cis-regulatory elements important for paraxial mesoderm formation and axis extension. Nat Commun 2021; 12:1157. [PMID: 33608545 PMCID: PMC7895974 DOI: 10.1038/s41467-021-21426-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 01/25/2021] [Indexed: 01/31/2023] Open
Abstract
Somites arising from paraxial mesoderm are a hallmark of the segmented vertebrate body plan. They form sequentially during axis extension and generate musculoskeletal cell lineages. How paraxial mesoderm becomes regionalised along the axis and how this correlates with dynamic changes of chromatin accessibility and the transcriptome remains unknown. Here, we report a spatiotemporal series of ATAC-seq and RNA-seq along the chick embryonic axis. Footprint analysis shows differential coverage of binding sites for several key transcription factors, including CDX2, LEF1 and members of HOX clusters. Associating accessible chromatin with nearby expressed genes identifies cis-regulatory elements (CRE) for TCF15 and MEOX1. We determine their spatiotemporal activity and evolutionary conservation in Xenopus and human. Epigenome silencing of endogenous CREs disrupts TCF15 and MEOX1 gene expression and recapitulates phenotypic abnormalities of anterior-posterior axis extension. Our integrated approach allows dissection of paraxial mesoderm regulatory circuits in vivo and has implications for investigating gene regulatory networks.
Collapse
|
9
|
Wittig JG, Münsterberg A. The Chicken as a Model Organism to Study Heart Development. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037218. [PMID: 31767650 DOI: 10.1101/cshperspect.a037218] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heart development is a complex process and begins with the long-range migration of cardiac progenitor cells during gastrulation. This culminates in the formation of a simple contractile tube with multiple layers, which undergoes remodeling into a four-chambered heart. During this morphogenesis, additional cell populations become incorporated. It is important to unravel the underlying genetic and cellular mechanisms to be able to identify the embryonic origin of diseases, including congenital malformations, which impair cardiac function and may affect life expectancy or quality. Owing to the evolutionary conservation of development, observations made in nonamniote and amniote vertebrate species allow us to extrapolate to human. This review will focus on the contributions made to a better understanding of heart development through studying avian embryos-mainly the chicken but also quail embryos. We will illustrate the classic and recent approaches used in the avian system, give an overview of the important discoveries made, and summarize the early stages of cardiac development up to the establishment of the four-chambered heart.
Collapse
Affiliation(s)
- Johannes G Wittig
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| |
Collapse
|
10
|
Vonica A, Bhat N, Phan K, Guo J, Iancu L, Weber JA, Karger A, Cain JW, Wang ECE, DeStefano GM, O'Donnell-Luria AH, Christiano AM, Riley B, Butler SJ, Luria V. Apcdd1 is a dual BMP/Wnt inhibitor in the developing nervous system and skin. Dev Biol 2020; 464:71-87. [PMID: 32320685 PMCID: PMC7307705 DOI: 10.1016/j.ydbio.2020.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/02/2023]
Abstract
Animal development and homeostasis depend on precise temporal and spatial intercellular signaling. Components shared between signaling pathways, generally thought to decrease specificity, paradoxically can also provide a solution to pathway coordination. Here we show that the Bone Morphogenetic Protein (BMP) and Wnt signaling pathways share Apcdd1 as a common inhibitor and that Apcdd1 is a taxon-restricted gene with novel domains and signaling functions. Previously, we showed that Apcdd1 inhibits Wnt signaling (Shimomura et al., 2010), here we find that Apcdd1 potently inhibits BMP signaling in body axis formation and neural differentiation in chicken, frog, zebrafish. Furthermore, we find that Apcdd1 has an evolutionarily novel protein domain. Our results from experiments and modeling suggest that Apcdd1 may coordinate the outputs of two signaling pathways that are central to animal development and human disease.
Collapse
Affiliation(s)
- Alin Vonica
- Departments of Genetics and Development, and Dermatology, Columbia University Medical Center, New York, NY, 10032, USA; Department of Biology, The Nazareth College, Rochester, NY, 14618, USA
| | - Neha Bhat
- Department of Biology, Texas A&M University, College Station, TX, 7783-3258, USA; Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, 06511, USA
| | - Keith Phan
- Department of Neurobiology, University of California, Los Angeles, CA, 90095-7239, USA
| | - Jinbai Guo
- Department of Biology, Texas A&M University, College Station, TX, 7783-3258, USA
| | - Lăcrimioara Iancu
- Institut für Algebra und Zahlentheorie, Universität Stuttgart, D-70569, Stuttgart, Germany; Institute of Mathematics, University of Aberdeen, Aberdeen, AB24 3UE, Scotland, UK
| | - Jessica A Weber
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Amir Karger
- IT-Research Computing, Harvard Medical School, Boston, MA, 02115, USA
| | - John W Cain
- Department of Mathematics, Harvard University, Cambridge, MA, 02138, USA
| | - Etienne C E Wang
- Departments of Genetics and Development, and Dermatology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Gina M DeStefano
- Departments of Genetics and Development, and Dermatology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Anne H O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Angela M Christiano
- Departments of Genetics and Development, and Dermatology, Columbia University Medical Center, New York, NY, 10032, USA.
| | - Bruce Riley
- Department of Biology, Texas A&M University, College Station, TX, 7783-3258, USA.
| | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, 90095-7239, USA.
| | - Victor Luria
- Departments of Genetics and Development, and Dermatology, Columbia University Medical Center, New York, NY, 10032, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
11
|
Serrano Nájera G, Weijer CJ. Cellular processes driving gastrulation in the avian embryo. Mech Dev 2020; 163:103624. [PMID: 32562871 PMCID: PMC7511600 DOI: 10.1016/j.mod.2020.103624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 01/18/2023]
Abstract
Gastrulation consists in the dramatic reorganisation of the epiblast, a one-cell thick epithelial sheet, into a multilayered embryo. In chick, the formation of the internal layers requires the generation of a macroscopic convection-like flow, which involves up to 50,000 epithelial cells in the epiblast. These cell movements locate the mesendoderm precursors into the midline of the epiblast to form the primitive streak. There they acquire a mesenchymal phenotype, ingress into the embryo and migrate outward to populate the inner embryonic layers. This review covers what is currently understood about how cell behaviours ultimately cause these morphogenetic events and how they are regulated. We discuss 1) how the biochemical patterning of the embryo before gastrulation creates compartments of differential cell behaviours, 2) how the global epithelial flows arise from the coordinated actions of individual cells, 3) how the cells delaminate individually from the epiblast during the ingression, and 4) how cells move after the ingression following stereotypical migration routes. We conclude by exploring new technical advances that will facilitate future research in the chick model system.
Collapse
Affiliation(s)
- Guillermo Serrano Nájera
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Cornelis J Weijer
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| |
Collapse
|
12
|
Abstract
In birds as in all amniotes, the site of gastrulation is a midline structure, the primitive streak. This appears as cells in the one cell-thick epiblast undergo epithelial-to-mesenchymal transition to ingress and form definitive mesoderm and endoderm. Global movements involving tens of thousands of cells in the embryonic epiblast precede gastrulation. They position the primitive streak precursors from a marginal position (equivalent to the situation in anamniotes) along the future antero-posterior axis (typical for amniotes). These epithelial movements continue in modified form during gastrulation, when they are accompanied by collective movements of different class in the forming mesoderm and endoderm. Here I discuss the nature of these collective cell movements shaping the embryo, their interplay with signaling events controlling fate specification and significance in an evolutionary perspective.
Collapse
|
13
|
He L, Zhou J, Chen M, Lin CS, Kim SG, Zhou Y, Xiang L, Xie M, Bai H, Yao H, Shi C, Coelho PG, Bromage TG, Hu B, Tovar N, Witek L, Wu J, Chen K, Gu W, Zheng J, Sheu TJ, Zhong J, Wen J, Niu Y, Cheng B, Gong Q, Owens DM, Stanislauskas M, Pei J, Chotkowski G, Wang S, Yang G, Zegarelli DJ, Shi X, Finkel M, Zhang W, Li J, Cheng J, Tarnow DP, Zhou X, Wang Z, Jiang X, Romanov A, Rowe DW, Wang S, Ye L, Ling J, Mao J. Parenchymal and stromal tissue regeneration of tooth organ by pivotal signals reinstated in decellularized matrix. NATURE MATERIALS 2019; 18:627-637. [PMID: 31114073 PMCID: PMC7362336 DOI: 10.1038/s41563-019-0368-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 04/09/2019] [Indexed: 02/05/2023]
Abstract
Cells are transplanted to regenerate an organs' parenchyma, but how transplanted parenchymal cells induce stromal regeneration is elusive. Despite the common use of a decellularized matrix, little is known as to the pivotal signals that must be restored for tissue or organ regeneration. We report that Alx3, a developmentally important gene, orchestrated adult parenchymal and stromal regeneration by directly transactivating Wnt3a and vascular endothelial growth factor. In contrast to the modest parenchyma formed by native adult progenitors, Alx3-restored cells in decellularized scaffolds not only produced vascularized stroma that involved vascular endothelial growth factor signalling, but also parenchymal dentin via the Wnt/β-catenin pathway. In an orthotopic large-animal model following parenchyma and stroma ablation, Wnt3a-recruited endogenous cells regenerated neurovascular stroma and differentiated into parenchymal odontoblast-like cells that extended the processes into newly formed dentin with a structure-mechanical equivalency to native dentin. Thus, the Alx3-Wnt3a axis enables postnatal progenitors with a modest innate regenerative capacity to regenerate adult tissues. Depleted signals in the decellularized matrix may be reinstated by a developmentally pivotal gene or corresponding protein.
Collapse
Affiliation(s)
- Ling He
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jian Zhou
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Mo Chen
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Chyuan-Sheng Lin
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Sahng G Kim
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Columbia University College of Dental Medicine, New York, NY, USA
| | - Yue Zhou
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Department of Conservative Dentistry, Laboratory of Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai, China
| | - Lusai Xiang
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ming Xie
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Department of Prosthodontics, Shanghai Jiao Tong University, Shanghai, China
| | - Hanying Bai
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Hai Yao
- Department of Bioengineering, Clemson University, Charleston, SC, USA
| | - Changcheng Shi
- Department of Bioengineering, Clemson University, Charleston, SC, USA
| | - Paulo G Coelho
- Department of Biomaterials and Biomimetics, New York University, New York, NY, USA
| | - Timothy G Bromage
- Department of Biomaterials and Biomimetics, New York University, New York, NY, USA
| | - Bin Hu
- Department of Biomaterials and Biomimetics, New York University, New York, NY, USA
| | - Nick Tovar
- Department of Biomaterials and Biomimetics, New York University, New York, NY, USA
| | - Lukasz Witek
- Department of Biomaterials and Biomimetics, New York University, New York, NY, USA
| | - Jiaqian Wu
- Vivian L. Smith Department of Neurosurgery, Center for Stem Cell and Regenerative Medicine University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Kenian Chen
- Vivian L. Smith Department of Neurosurgery, Center for Stem Cell and Regenerative Medicine University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Wei Gu
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Jinxuan Zheng
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Tzong-Jen Sheu
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, USA
| | - Juan Zhong
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jin Wen
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Department of Prosthodontics, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Niu
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Bin Cheng
- Columbia University Mailman School of Public Health, Department of Biostatistics, New York, NY, USA
| | - Qimei Gong
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - David M Owens
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
- Department of Dermatology, Columbia University, New York, NY, USA
| | | | - Jasmine Pei
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | | | - Sainan Wang
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Guodong Yang
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | | | - Xin Shi
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | | | - Wen Zhang
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Junyuan Li
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Jiayi Cheng
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA
| | - Dennis P Tarnow
- Columbia University College of Dental Medicine, New York, NY, USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Zuolin Wang
- Department of Conservative Dentistry, Laboratory of Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai, China
| | - Xinquan Jiang
- Department of Prosthodontics, Shanghai Jiao Tong University, Shanghai, China
| | - Alexander Romanov
- Institute of Comparative Medicine, Columbia University Medical Center, New York, NY, USA
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Science Center, Farmington, CT, USA
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Junqi Ling
- Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatology Hospital, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Jeremy Mao
- Columbia University, Center for Craniofacial Regeneration, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
- Columbia University College of Dental Medicine, New York, NY, USA.
- Department of Orthopedic Surgery, Columbia University Physician and Surgeons, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Sivakumar A, Kurpios NA. Transcriptional regulation of cell shape during organ morphogenesis. J Cell Biol 2018; 217:2987-3005. [PMID: 30061107 PMCID: PMC6122985 DOI: 10.1083/jcb.201612115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The emerging field of transcriptional regulation of cell shape changes aims to address the critical question of how gene expression programs produce a change in cell shape. Together with cell growth, division, and death, changes in cell shape are essential for organ morphogenesis. Whereas most studies of cell shape focus on posttranslational events involved in protein organization and distribution, cell shape changes can be genetically programmed. This review highlights the essential role of transcriptional regulation of cell shape during morphogenesis of the heart, lungs, gastrointestinal tract, and kidneys. We emphasize the evolutionary conservation of these processes across different model organisms and discuss perspectives on open questions and research avenues that may provide mechanistic insights toward understanding birth defects.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
15
|
HOPX Defines Heterogeneity of Postnatal Subventricular Zone Neural Stem Cells. Stem Cell Reports 2018; 11:770-783. [PMID: 30174314 PMCID: PMC6135899 DOI: 10.1016/j.stemcr.2018.08.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 12/16/2022] Open
Abstract
The largest diversity of neural lineages generated from the subventricular zone (SVZ) occurs early after birth and is regulated in a spatiotemporal manner depending on the expression of specific transcriptional cues. Transcriptomics and fate-mapping approaches were employed to explore the relationship between regional expression of transcription factors by neural stem cells (NSCs) and the specification of distinct neural lineages. Our results support an early priming of NSCs for the genesis of defined cell types depending on their spatial location in the SVZ and identify HOPX as a marker of a subpopulation primed toward astrocytic fates. Manipulation of HOPX expression, however, showed no effect on astrogenesis but resulted in marked changes in the number of NSCs and of their progenies. Taken together, our results highlight transcriptional and spatial heterogeneity of postnatal NSCs and reveal a key role for HOPX in controlling SVZ germinal activity.
Collapse
|
16
|
Gao LR, Wang G, Zhang J, Li S, Chuai M, Bao Y, Hocher B, Yang X. High salt-induced excess reactive oxygen species production resulted in heart tube malformation during gastrulation. J Cell Physiol 2018; 233:7120-7133. [PMID: 29574800 DOI: 10.1002/jcp.26528] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 01/31/2018] [Indexed: 12/30/2022]
Abstract
An association has been proved between high salt consumption and cardiovascular mortality. In vertebrates, the heart is the first functional organ to be formed. However, it is not clear whether high-salt exposure has an adverse impact on cardiogenesis. Here we report high-salt exposure inhibited basement membrane breakdown by affecting RhoA, thus disturbing the expression of Slug/E-cadherin/N-cadherin/Laminin and interfering with mesoderm formation during the epithelial-mesenchymal transition(EMT). Furthermore, the DiI+ cell migration trajectory in vivo and scratch wound assays in vitro indicated that high-salt exposure restricted cell migration of cardiac progenitors, which was caused by the weaker cytoskeleton structure and unaltered corresponding adhesion junctions at HH7. Besides, down-regulation of GATA4/5/6, Nkx2.5, TBX5, and Mef2c and up-regulation of Wnt3a/β-catenin caused aberrant cardiomyocyte differentiation at HH7 and HH10. High-salt exposure also inhibited cell proliferation and promoted apoptosis. Most importantly, our study revealed that excessive reactive oxygen species(ROS)generated by high salt disturbed the expression of cardiac-related genes, detrimentally affecting the above process including EMT, cell migration, differentiation, cell proliferation and apoptosis, which is the major cause of malformation of heart tubes.
Collapse
Affiliation(s)
- Lin-Rui Gao
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Guang Wang
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Jing Zhang
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Shuai Li
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee, UK
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, Norfolk, UK
| | - Berthold Hocher
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Institute of Nutritional Science, University of Potsdam, Potsdam-Nuthetal, Germany
| | - Xuesong Yang
- Division of Histology and Embryology, Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Coster AD, Thorne CA, Wu LF, Altschuler SJ. Examining Crosstalk among Transforming Growth Factor β, Bone Morphogenetic Protein, and Wnt Pathways. J Biol Chem 2016; 292:244-250. [PMID: 27895117 PMCID: PMC5217683 DOI: 10.1074/jbc.m116.759654] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/15/2016] [Indexed: 11/10/2022] Open
Abstract
The integration of morphogenic signals by cells is not well understood. A growing body of literature suggests increasingly complex coupling among classically defined pathways. Given this apparent complexity, it is difficult to predict where, when, or even whether crosstalk occurs. Here, we investigated pairs of morphogenic pathways, previously reported to have multiple points of crosstalk, which either do not share (TGFβ and Wnt/β-catenin) or share (TGFβ and bone morphogenetic protein (BMP)) core signaling components. Crosstalk was measured by the ability of one morphogenic pathway to cross-activate core transcription factors and/or target genes of another morphogenic pathway. In contrast to previous studies, we found a surprising absence of crosstalk between TGFβ and Wnt/β-catenin. Further, we did not observe expected cross-pathway inhibition in between TGFβ and BMP, despite the fact that both use (or could compete) for the shared component SMAD4. Critical to our assays was a separation of timescales, which helped separate crosstalk due to initial signal transduction from subsequent post-transcriptional feedback events. Our study revealed fewer (and different) inter-morphogenic pathway crosstalk connections than expected; even pathways that share components can be insulated from one another.
Collapse
Affiliation(s)
- Adam D Coster
- From the Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| | - Curtis A Thorne
- From the Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| | - Lani F Wu
- From the Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 and .,the Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158
| | - Steven J Altschuler
- From the Green Center for Systems Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 and .,the Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94158
| |
Collapse
|
18
|
Baik J, Magli A, Tahara N, Swanson SA, Koyano-Nakagawa N, Borges L, Stewart R, Garry DJ, Kawakami Y, Thomson JA, Perlingeiro RCR. Endoglin integrates BMP and Wnt signalling to induce haematopoiesis through JDP2. Nat Commun 2016; 7:13101. [PMID: 27713415 PMCID: PMC5059784 DOI: 10.1038/ncomms13101] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/02/2016] [Indexed: 01/05/2023] Open
Abstract
Mechanisms of haematopoietic and cardiac patterning remain poorly understood. Here we show that the BMP and Wnt signalling pathways are integrated in an endoglin (Eng)-dependent manner in cardiac and haematopoietic lineage specification. Eng is expressed in early mesoderm and marks both haematopoietic and cardiac progenitors. In the absence of Eng, yolk sacs inappropriately express the cardiac marker, Nkx2.5. Conversely, high levels of Eng in vitro and in vivo increase haematopoiesis and inhibit cardiogenesis. Levels of Eng determine the activation of both BMP and Wnt pathways, which are integrated downstream of Eng by phosphorylation of Smad1 by Gsk3. By interrogating Eng-dependent Wnt-mediated transcriptional changes, we identify Jdp2 as a key Eng-dependent Wnt target, sufficient to establish haematopoietic fate in early mesoderm when BMP and Wnt crosstalk is disturbed. These studies provide mechanistic insight into the integration of BMP and Wnt signalling in the establishment of haematopoietic and cardiac progenitors during embryogenesis. How both BMP and Wnt signalling pathways regulate lineage specification early in development is unclear. Here, the authors show that endoglin via Jdp2, an AP-1 family member, modulates BMP and Wnt signalling to commit mesodermal progenitors to a haematopoietic fate at the expense of the cardiac lineage.
Collapse
Affiliation(s)
- June Baik
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Alessandro Magli
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Naoyuki Tahara
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Scott A Swanson
- Regerative Biology, Morgridge Institute for Research, Madison, Wisconsin 53715, USA
| | - Naoko Koyano-Nakagawa
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Luciene Borges
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Ron Stewart
- Regerative Biology, Morgridge Institute for Research, Madison, Wisconsin 53715, USA
| | - Daniel J Garry
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - James A Thomson
- Regerative Biology, Morgridge Institute for Research, Madison, Wisconsin 53715, USA
| | - Rita C R Perlingeiro
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
19
|
Münsterberg A, Hoppler S. WNT and BMP regulate roadblocks toward cardiomyocyte differentiation: lessons learned from embryos inform human stem cell differentiation. Stem Cell Investig 2016; 3:33. [PMID: 27580968 DOI: 10.21037/sci.2016.07.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 07/14/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Andrea Münsterberg
- School of Biological Sciences, Cell and Developmental Biology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Stefan Hoppler
- Institute of Medical Sciences, University of Aberdeen, Foresterhill Health Campus, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
20
|
Chiapparo G, Lin X, Lescroart F, Chabab S, Paulissen C, Pitisci L, Bondue A, Blanpain C. Mesp1 controls the speed, polarity, and directionality of cardiovascular progenitor migration. J Cell Biol 2016; 213:463-77. [PMID: 27185833 PMCID: PMC4878090 DOI: 10.1083/jcb.201505082] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 04/18/2016] [Indexed: 01/09/2023] Open
Abstract
During embryonic development, Mesp1 marks the earliest cardiovascular progenitors (CPs) and promotes their specification, epithelial-mesenchymal transition (EMT), and cardiovascular differentiation. However, Mesp1 deletion in mice does not impair initial CP specification and early cardiac differentiation but induces cardiac malformations thought to arise from a defect of CP migration. Using inducible gain-of-function experiments during embryonic stem cell differentiation, we found that Mesp2, its closest homolog, was as efficient as Mesp1 at promoting CP specification, EMT, and cardiovascular differentiation. However, only Mesp1 stimulated polarity and directional cell migration through a cell-autonomous mechanism. Transcriptional analysis and chromatin immunoprecipitation experiments revealed that Mesp1 and Mesp2 activate common target genes that promote CP specification and differentiation. We identified two direct Mesp1 target genes, Prickle1 and RasGRP3, that are strongly induced by Mesp1 and not by Mesp2 and that control the polarity and the speed of cell migration. Altogether, our results identify the molecular interface controlled by Mesp1 that links CP specification and cell migration.
Collapse
Affiliation(s)
- Giuseppe Chiapparo
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Xionghui Lin
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Fabienne Lescroart
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Samira Chabab
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Catherine Paulissen
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Lorenzo Pitisci
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium
| | - Antoine Bondue
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium Department of Cardiology, Hopital Erasme, Brussels B-1070, Belgium
| | - Cédric Blanpain
- Université Libre de Bruxelles, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Brussels B-1070, Belgium WELBIO, Université Libre de Bruxelles, Brussels B-1070, Belgium
| |
Collapse
|
21
|
O'Neill HL, Cassidy AP, Harris OB, Cassidy JW. BMP2/BMPR1A is linked to tumour progression in dedifferentiated liposarcomas. PeerJ 2016; 4:e1957. [PMID: 27114889 PMCID: PMC4841227 DOI: 10.7717/peerj.1957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/31/2016] [Indexed: 01/30/2023] Open
Abstract
Bone Morphogenic Protein 2 (BMP2) is a multipurpose cytokine, important in the development of bone and cartilage, and with a role in tumour initiation and progression. BMP2 signal transduction is dependent on two distinct classes of serine/threonine kinase known as the type I and type II receptors. Although the type I receptors (BMPR1A and BMPR1B) are largely thought to have overlapping functions, we find tissue and cellular compartment specific patterns of expression, suggesting potential for distinct BMP2 signalling outcomes dependent on tissue type. Herein, we utilise large publicly available datasets from The Cancer Genome Atlas (TCGA) and Protein Atlas to define a novel role for BMP2 in the progression of dedifferentiated liposarcomas. Using disease free survival as our primary endpoint, we find that BMP2 confers poor prognosis only within the context of high BMPR1A expression. Through further annotation of the TCGA sarcoma dataset, we localise this effect to dedifferentiated liposarcomas but find overall BMP2/BMP receptor expression is equal across subsets. Finally, through gene set enrichment analysis we link the BMP2/BMPR1A axis to increased transcriptional activity of the matrisome and general extracellular matrix remodelling. Our study highlights the importance of continued research into the tumorigenic properties of BMP2 and the potential disadvantages of recombinant human BMP2 (rhBMP2) use in orthopaedic surgery. For the first time, we identify high BMP2 expression within the context of high BMPR1A expression as a biomarker of disease relapse in dedifferentiated liposarcomas.
Collapse
Affiliation(s)
- Hannah L O'Neill
- Aberdeen Royal Infirmary, University of Aberdeen , Aberdeen , United Kingdom
| | - Amy P Cassidy
- Aberdeen Royal Infirmary, University of Aberdeen, Aberdeen, United Kingdom; Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Olivia B Harris
- Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - John W Cassidy
- Queens' College, University of Cambridge, Cambridge Cambridgeshire, United Kingdom; Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
22
|
Wittig JG, Münsterberg A. The Early Stages of Heart Development: Insights from Chicken Embryos. J Cardiovasc Dev Dis 2016; 3:jcdd3020012. [PMID: 29367563 PMCID: PMC5715676 DOI: 10.3390/jcdd3020012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 03/28/2016] [Accepted: 03/30/2016] [Indexed: 01/01/2023] Open
Abstract
The heart is the first functioning organ in the developing embryo and a detailed understanding of the molecular and cellular mechanisms involved in its formation provides insights into congenital malformations affecting its function and therefore the survival of the organism. Because many developmental mechanisms are highly conserved, it is possible to extrapolate from observations made in invertebrate and vertebrate model organisms to humans. This review will highlight the contributions made through studying heart development in avian embryos, particularly the chicken. The major advantage of chick embryos is their accessibility for surgical manipulation and functional interference approaches, both gain- and loss-of-function. In addition to experiments performed in ovo, the dissection of tissues for ex vivo culture, genomic, or biochemical approaches is straightforward. Furthermore, embryos can be cultured for time-lapse imaging, which enables tracking of fluorescently labeled cells and detailed analysis of tissue morphogenesis. Owing to these features, investigations in chick embryos have led to important discoveries, often complementing genetic studies in mice and zebrafish. As well as including some historical aspects, we cover here some of the crucial advances made in understanding early heart development using the chicken model.
Collapse
Affiliation(s)
- Johannes G Wittig
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| |
Collapse
|
23
|
Fourel L, Valat A, Faurobert E, Guillot R, Bourrin-Reynard I, Ren K, Lafanechère L, Planus E, Picart C, Albiges-Rizo C. β3 integrin-mediated spreading induced by matrix-bound BMP-2 controls Smad signaling in a stiffness-independent manner. J Cell Biol 2016; 212:693-706. [PMID: 26953352 PMCID: PMC4792076 DOI: 10.1083/jcb.201508018] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 02/09/2016] [Indexed: 12/31/2022] Open
Abstract
Matrix-bound BMP-2 is sufficient to induce β3 integrin–dependent, Cdc42/Src/FAK/ILK-mediated cell spreading by overriding the stiffness response through actin and adhesion site dynamics, showing BMP receptors and integrins work together to control signaling and tensional homeostasis, thereby coupling cell adhesion and fate commitment. Understanding how cells integrate multiple signaling pathways to achieve specific cell differentiation is a challenging question in cell biology. We have explored the physiological presentation of BMP-2 by using a biomaterial that harbors tunable mechanical properties to promote localized BMP-2 signaling. We show that matrix-bound BMP-2 is sufficient to induce β3 integrin–dependent C2C12 cell spreading by overriding the soft signal of the biomaterial and impacting actin organization and adhesion site dynamics. In turn, αvβ3 integrin is required to mediate BMP-2–induced Smad signaling through a Cdc42–Src–FAK–ILK pathway. β3 integrin regulates a multistep process to control first BMP-2 receptor activity and second the inhibitory role of GSK3 on Smad signaling. Overall, our results show that BMP receptors and β3 integrin work together to control Smad signaling and tensional homeostasis, thereby coupling cell adhesion and fate commitment, two fundamental aspects of developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Laure Fourel
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France Centre National de la Recherche Scientifique UMR 5628, Laboratoire des Matériaux et du Génie Physique, Institute of Technology, 38016 Grenoble, France
| | - Anne Valat
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France Centre National de la Recherche Scientifique UMR 5628, Laboratoire des Matériaux et du Génie Physique, Institute of Technology, 38016 Grenoble, France
| | - Eva Faurobert
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France
| | - Raphael Guillot
- Centre National de la Recherche Scientifique UMR 5628, Laboratoire des Matériaux et du Génie Physique, Institute of Technology, 38016 Grenoble, France
| | - Ingrid Bourrin-Reynard
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France
| | - Kefeng Ren
- Centre National de la Recherche Scientifique UMR 5628, Laboratoire des Matériaux et du Génie Physique, Institute of Technology, 38016 Grenoble, France
| | - Laurence Lafanechère
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France
| | - Emmanuelle Planus
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France
| | - Catherine Picart
- Centre National de la Recherche Scientifique UMR 5628, Laboratoire des Matériaux et du Génie Physique, Institute of Technology, 38016 Grenoble, France
| | - Corinne Albiges-Rizo
- Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, France Centre National de la Recherche Scientifique, Equipe de Recherche Labellisée 5284, 38042 Grenoble, France Université Grenoble Alpes, 38041 Grenoble, France
| |
Collapse
|
24
|
Pandey MK, DeGrado TR. Glycogen Synthase Kinase-3 (GSK-3)-Targeted Therapy and Imaging. Am J Cancer Res 2016; 6:571-93. [PMID: 26941849 PMCID: PMC4775866 DOI: 10.7150/thno.14334] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is associated with various key biological processes, including glucose regulation, apoptosis, protein synthesis, cell signaling, cellular transport, gene transcription, proliferation, and intracellular communication. Accordingly, GSK-3 has been implicated in a wide variety of diseases and specifically targeted for both therapeutic and imaging applications by a large number of academic laboratories and pharmaceutical companies. Here, we review the structure, function, expression levels, and ligand-binding properties of GSK-3 and its connection to various diseases. A selected list of highly potent GSK-3 inhibitors, with IC50 <20 nM for adenosine triphosphate (ATP)-competitive inhibitors and IC50 <5 μM for non-ATP-competitive inhibitors, were analyzed for structure activity relationships. Furthermore, ubiquitous expression of GSK-3 and its possible impact on therapy and imaging are also highlighted. Finally, a rational perspective and possible route to selective and effective GSK-3 inhibitors is discussed.
Collapse
|
25
|
Yang K, Wang X, Zhang H, Wang Z, Nan G, Li Y, Zhang F, Mohammed MK, Haydon RC, Luu HH, Bi Y, He TC. The evolving roles of canonical WNT signaling in stem cells and tumorigenesis: implications in targeted cancer therapies. J Transl Med 2016; 96:116-36. [PMID: 26618721 PMCID: PMC4731283 DOI: 10.1038/labinvest.2015.144] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/06/2015] [Indexed: 02/07/2023] Open
Abstract
The canonical WNT/β-catenin signaling pathway governs a myriad of biological processes underlying the development and maintenance of adult tissue homeostasis, including regulation of stem cell self-renewal, cell proliferation, differentiation, and apoptosis. WNTs are secreted lipid-modified glycoproteins that act as short-range ligands to activate receptor-mediated signaling pathways. The hallmark of the canonical pathway is the activation of β-catenin-mediated transcriptional activity. Canonical WNTs control the β-catenin dynamics as the cytoplasmic level of β-catenin is tightly regulated via phosphorylation by the 'destruction complex', consisting of glycogen synthase kinase 3β (GSK3β), casein kinase 1α (CK1α), the scaffold protein AXIN, and the tumor suppressor adenomatous polyposis coli (APC). Aberrant regulation of this signaling cascade is associated with varieties of human diseases, especially cancers. Over the past decade, significant progress has been made in understanding the mechanisms of canonical WNT signaling. In this review, we focus on the current understanding of WNT signaling at the extracellular, cytoplasmic membrane, and intracellular/nuclear levels, including the emerging knowledge of cross-talk with other pathways. Recent progresses in developing novel WNT pathway-targeted therapies will also be reviewed. Thus, this review is intended to serve as a refresher of the current understanding about the physiologic and pathogenic roles of WNT/β-catenin signaling pathway, and to outline potential therapeutic opportunities by targeting the canonical WNT pathway.
Collapse
Affiliation(s)
- Ke Yang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Xin Wang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Department of Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Zhongliang Wang
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guoxin Nan
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yasha Li
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Fugui Zhang
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Maryam K. Mohammed
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| | - Tong-Chuan He
- Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University; Chongqing, China, Molecular Oncology Laboratory, The University of Chicago Medical Center, Chicago, IL 60637, USA, Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China, Corresponding authors T.-C. He, MD, PhD, Molecular Oncology Laboratory, The University of Chicago Medical Center, 5841 South Maryland Avenue, MC 3079, Chicago, IL 60637, USA, Tel. (773) 702-7169; Fax (773) 834-4598, , Yang Bi, MD, PhD, Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children's Hospital, Chongqing Medical University, Chongqing 400046, China, Tel. 011-86-23-63633113; Fax: 011-86-236362690,
| |
Collapse
|
26
|
Thies RS, Murry CE. The advancement of human pluripotent stem cell-derived therapies into the clinic. Development 2016; 142:3077-84. [PMID: 26395136 DOI: 10.1242/dev.126482] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human pluripotent stem cells (hPSCs) offer many potential applications for drug screening and 'disease in a dish' assay capabilities. However, a more ambitious goal is to develop cell therapeutics using hPSCs to generate and replace somatic cells that are lost as a result of disease or injury. This Spotlight article will describe the state of progress of some of the hPSC-derived therapeutics that offer the most promise for clinical use. Lessons from developmental biology have been instrumental in identifying signaling molecules that can guide these differentiation processes in vitro, and will be described in the context of these cell therapy programs.
Collapse
Affiliation(s)
- R Scott Thies
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Charles E Murry
- Center for Cardiovascular Biology, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA Department of Pathology, University of Washington, Seattle, WA 98195, USA Department of Bioengineering, University of Washington, Seattle, WA 98195, USA Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
27
|
McBride-Gagyi SH, McKenzie JA, Buettmann EG, Gardner MJ, Silva MJ. Bmp2 conditional knockout in osteoblasts and endothelial cells does not impair bone formation after injury or mechanical loading in adult mice. Bone 2015; 81:533-543. [PMID: 26344756 PMCID: PMC4640950 DOI: 10.1016/j.bone.2015.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/20/2015] [Accepted: 09/03/2015] [Indexed: 12/11/2022]
Abstract
Post-natal osteogenesis after mechanical trauma or stimulus occurs through either endochondral healing, intramembranous healing or lamellar bone formation. Bone morphogenetic protein 2 (BMP2) is up-regulated in each of these osteogenic processes and is expressed by a variety of cells including osteoblasts and vascular cells. It is known that genetic knockout of Bmp2 in all cells or in osteo-chondroprogenitor cells completely abrogates endochondral healing after full fracture. However, the importance of BMP2 from differentiated osteoblasts and endothelial cells is not known. Moreover, the importance of BMP2 in non-endochondral bone formation such as intramembranous healing or lamellar bone formation is not known. Using inducible and tissue-specific Cre-lox mediated targeting of Bmp2 in adult (10-24 week old) mice, we assessed the role of BMP2 expression globally, by osteoblasts, and by vascular endothelial cells in endochondral healing, intramembranous healing and lamellar bone formation. These three osteogenic processes were modeled using full femur fracture, ulnar stress fracture, and ulnar non-damaging cyclic loading, respectively. Our results confirmed the requirement of BMP2 for endochondral fracture healing, as mice in which Bmp2 was knocked out in all cells prior to fracture failed to form a callus. Targeted deletion of Bmp2 in osteoblasts (osterix-expressing) or vascular endothelial cells (vascular endothelial cadherin-expressing) did not impact fracture healing in any way. Regarding non-endochondral bone formation, we found that BMP2 is largely dispensable for intramembranous bone formation after stress fracture and also not required for lamellar bone formation induced by mechanical loading. Taken together our results indicate that osteoblasts and endothelial cells are not a critical source of BMP2 in endochondral fracture healing, and that non-endochondral bone formation in the adult mouse is not as critically dependent on BMP2.
Collapse
Affiliation(s)
- Sarah Howe McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall, M176, St. Louis, MO 63104, USA; Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Jennifer A McKenzie
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Evan G Buettmann
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Michael J Gardner
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| | - Matthew J Silva
- Department of Orthopedic Surgery, Washington University in St. Louis School of Medicine, 660 S. Euclid, Campus Box8233, St. Louis, MO 63110, USA.
| |
Collapse
|
28
|
Sánchez-Duffhues G, Hiepen C, Knaus P, Ten Dijke P. Bone morphogenetic protein signaling in bone homeostasis. Bone 2015; 80:43-59. [PMID: 26051467 DOI: 10.1016/j.bone.2015.05.025] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/11/2015] [Accepted: 05/20/2015] [Indexed: 01/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are cytokines belonging to the transforming growth factor-β (TGF-β) superfamily. They play multiple functions during development and tissue homeostasis, including regulation of the bone homeostasis. The BMP signaling pathway consists in a well-orchestrated manner of ligands, membrane receptors, co-receptors and intracellular mediators, that regulate the expression of genes controlling the normal functioning of the bone tissues. Interestingly, BMP signaling perturbation is associated to a variety of low and high bone mass diseases, including osteoporosis, bone fracture disorders and heterotopic ossification. Consistent with these findings, in vitro and in vivo studies have shown that BMPs have potent effects on the activity of cells regulating bone function, suggesting that manipulation of the BMP signaling pathway may be employed as a therapeutic approach to treat bone diseases. Here we review the recent advances on BMP signaling and bone homeostasis, and how this knowledge may be used towards improved diagnosis and development of novel treatment modalities. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany.
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, The Netherlands.
| |
Collapse
|
29
|
Jain R, Li D, Gupta M, Manderfield LJ, Ifkovits JL, Wang Q, Liu F, Liu Y, Poleshko A, Padmanabhan A, Raum JC, Li L, Morrisey EE, Lu MM, Won KJ, Epstein JA. HEART DEVELOPMENT. Integration of Bmp and Wnt signaling by Hopx specifies commitment of cardiomyoblasts. Science 2015; 348:aaa6071. [PMID: 26113728 DOI: 10.1126/science.aaa6071] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cardiac progenitor cells are multipotent and give rise to cardiac endothelium, smooth muscle, and cardiomyocytes. Here, we define and characterize the cardiomyoblast intermediate that is committed to the cardiomyocyte fate, and we characterize the niche signals that regulate commitment. Cardiomyoblasts express Hopx, which functions to coordinate local Bmp signals to inhibit the Wnt pathway, thus promoting cardiomyogenesis. Hopx integrates Bmp and Wnt signaling by physically interacting with activated Smads and repressing Wnt genes. The identification of the committed cardiomyoblast that retains proliferative potential will inform cardiac regenerative therapeutics. In addition, Bmp signals characterize adult stem cell niches in other tissues where Hopx-mediated inhibition of Wnt is likely to contribute to stem cell quiescence and to explain the role of Hopx as a tumor suppressor.
Collapse
Affiliation(s)
- Rajan Jain
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deqiang Li
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mudit Gupta
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren J Manderfield
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamie L Ifkovits
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qiaohong Wang
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feiyan Liu
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ying Liu
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrey Poleshko
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arun Padmanabhan
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey C Raum
- Department of Genetics, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Li Li
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward E Morrisey
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Min Lu
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kyoung-Jae Won
- Department of Genetics, Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, Penn Cardiovascular Institute, Institute of Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
30
|
Luo JY, Zhang Y, Wang L, Huang Y. Regulators and effectors of bone morphogenetic protein signalling in the cardiovascular system. J Physiol 2015; 593:2995-3011. [PMID: 25952563 DOI: 10.1113/jp270207] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/27/2015] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) play key roles in the regulation of cell proliferation, differentiation and apoptosis in various tissues and organs, including the cardiovascular system. BMPs signal through both Smad-dependent and -independent cascades to exert a wide spectrum of biological activities. Cardiovascular disorders such as abnormal angiogenesis, atherosclerosis, pulmonary hypertension and cardiac hypertrophy have been linked to aberrant BMP signalling. To correct the dysregulated BMP signalling in cardiovascular pathogenesis, it is essential to get a better understanding of how the regulators and effectors of BMP signalling control cardiovascular function and how the dysregulated BMP signalling contributes to cardiovascular dysfunction. We hence highlight several key regulators of BMP signalling such as extracellular regulators of ligands, mechanical forces, microRNAs and small molecule drugs as well as typical BMP effectors like direct downstream target genes, mitogen-activated protein kinases, reactive oxygen species and microRNAs. The insights into these molecular processes will help target both the regulators and important effectors to reverse BMP-associated cardiovascular pathogenesis.
Collapse
Affiliation(s)
- Jiang-Yun Luo
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Li Wang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Huang
- Shenzhen Research Institute, Institute of Vascular Medicine, and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
31
|
Furtado J, Bento M, Correia E, Inácio JM, Belo JA. Expression and function of Ccbe1 in the chick early cardiogenic regions are required for correct heart development. PLoS One 2014; 9:e115481. [PMID: 25545279 PMCID: PMC4278723 DOI: 10.1371/journal.pone.0115481] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/24/2014] [Indexed: 11/25/2022] Open
Abstract
During the course of a differential screen to identify transcripts specific for chick heart/hemangioblast precursor cells, we have identified Ccbe1 (Collagen and calcium-binding EGF-like domain 1). While the importance of Ccbe1 for the development of the lymphatic system is now well demonstrated, its role in cardiac formation remained unknown. Here we show by whole-mount in situ hybridization analysis that cCcbe1 mRNA is initially detected in early cardiac progenitors of the two bilateral cardiogenic fields (HH4), and at later stages on the second heart field (HH9-18). Furthermore, cCcbe1 is expressed in multipotent and highly proliferative cardiac progenitors. We characterized the role of cCcbe1 during early cardiogenesis by performing functional studies. Upon morpholino-induced cCcbe1 knockdown, the chick embryos displayed heart malformations, which include aberrant fusion of the heart fields, leading to incomplete terminal differentiation of the cardiomyocytes. cCcbe1 overexpression also resulted in severe heart defects, including cardia bifida. Altogether, our data demonstrate that although cardiac progenitors cells are specified in cCcbe1 morphants, the migration and proliferation of cardiac precursors cells are impaired, suggesting that cCcbe1 is a key gene during early heart development.
Collapse
Affiliation(s)
- João Furtado
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular. e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-135 Faro, Portugal
| | - Margaret Bento
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular. e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-135 Faro, Portugal
| | - Elizabeth Correia
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular. e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-135 Faro, Portugal
| | - José Manuel Inácio
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular. e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-135 Faro, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal
| | - José António Belo
- Regenerative Medicine Program, Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular. e Estrutural, Universidade do Algarve, Campus de Gambelas, 8005-135 Faro, Portugal
- CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal
- * E-mail:
| |
Collapse
|
32
|
Derynck R, Muthusamy BP, Saeteurn KY. Signaling pathway cooperation in TGF-β-induced epithelial-mesenchymal transition. Curr Opin Cell Biol 2014; 31:56-66. [PMID: 25240174 DOI: 10.1016/j.ceb.2014.09.001] [Citation(s) in RCA: 304] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/01/2014] [Accepted: 09/01/2014] [Indexed: 12/28/2022]
Abstract
Transdifferentiation of epithelial cells into cells with mesenchymal properties and appearance, that is, epithelial-mesenchymal transition (EMT), is essential during development, and occurs in pathological contexts, such as in fibrosis and cancer progression. Although EMT can be induced by many extracellular ligands, TGF-β and TGF-β-related proteins have emerged as major inducers of this transdifferentiation process in development and cancer. Additionally, it is increasingly apparent that signaling pathways cooperate in the execution of EMT. This update summarizes the current knowledge of the coordination of TGF-β-induced Smad and non-Smad signaling pathways in EMT, and the remarkable ability of Smads to cooperate with other transcription-directed signaling pathways in the control of gene reprogramming during EMT.
Collapse
Affiliation(s)
- Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA.
| | - Baby Periyanayaki Muthusamy
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| | - Koy Y Saeteurn
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143-0669, USA
| |
Collapse
|
33
|
CardioPulse ArticlesThe Russian National Congress of Cardiology 2014Moving from academia to industryEACPR country of the month initiative: NetherlandsHow hearts are formed in embryosDifferential clinical outcomes after 1 year vs. 5 years in the SORT OUT III randomized comparison of zotarolimus- and sirolimus-eluting coronary stents:. Eur Heart J 2014; 35:2335. [DOI: 10.1093/eurheartj/ehu273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|