1
|
Beckröge T, Jux B, Seifert H, Theobald H, De Domenico E, Paulusch S, Beyer M, Schlitzer A, Mass E, Kolanus W. Impaired primitive erythropoiesis and defective vascular development in Trim71-KO embryos. Life Sci Alliance 2025; 8:e202402956. [PMID: 39909558 DOI: 10.26508/lsa.202402956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
The transition of an embryo from gastrulation to organogenesis requires precisely coordinated changes in gene expression, but the underlying mechanisms remain unclear. The RNA-binding protein Trim71 is essential for development and serves as a potent regulator of post-transcriptional gene expression. Here, we show that global deficiency of Trim71 induces severe defects in mesoderm-derived cells at the onset of organogenesis. Murine Trim71-KO embryos displayed impaired primitive erythropoiesis, yolk sac vasculature, heart function, and circulation, explaining the embryonic lethality of these mice. Tie2 Cre Trim71 conditional knockout did not induce strong defects, showing that Trim71 expression in endothelial cells and their immediate progenitors is dispensable for embryonic survival. scRNA-seq of E7.5 global Trim71-KO embryos revealed that transcriptomic changes arise already at gastrulation, showing a strong up-regulation of the mesodermal pioneer transcription factor Eomes. We identify Eomes as a direct target of Trim71-mediated mRNA repression via the NHL domain, demonstrating a functional link between these important regulatory genes. Taken together, our data suggest that Trim71-dependent control of gene expression at gastrulation establishes a framework for proper development during organogenesis.
Collapse
Affiliation(s)
- Tobias Beckröge
- https://ror.org/041nas322 Molecular Immunology and Cell Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Bettina Jux
- https://ror.org/041nas322 Molecular Immunology and Cell Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Hannah Seifert
- https://ror.org/041nas322 Molecular Immunology and Cell Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Hannah Theobald
- https://ror.org/041nas322 Quantitative Systems Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Elena De Domenico
- https://ror.org/041nas322 Genomics and Immunoregulation, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- https://ror.org/041nas322 PRECISE Platform for Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V. and University of Bonn and West German Genome Center, Bonn, Germany
| | - Stefan Paulusch
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- https://ror.org/041nas322 PRECISE Platform for Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V. and University of Bonn and West German Genome Center, Bonn, Germany
| | - Marc Beyer
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
- https://ror.org/041nas322 PRECISE Platform for Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V. and University of Bonn and West German Genome Center, Bonn, Germany
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., Bonn, Germany
| | - Andreas Schlitzer
- https://ror.org/041nas322 Quantitative Systems Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Elvira Mass
- https://ror.org/041nas322 Developmental Biology of the Immune System, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Waldemar Kolanus
- https://ror.org/041nas322 Molecular Immunology and Cell Biology, Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
2
|
Chen X, Tober J, Dominguez M, Tang AT, Bockman J, Yang J, Mani S, Lee CN, Chen M, Thillaikumaran T, Mericko-Ishizuka P, Mainigi M, Speck NA, Kahn ML. Lineage tracing studies suggest that the placenta is not a de novo source of hematopoietic stem cells. PLoS Biol 2025; 23:e3003003. [PMID: 39874373 PMCID: PMC11774391 DOI: 10.1371/journal.pbio.3003003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Definitive hematopoietic stem and progenitor cells (HSPCs) arise from a small number of hemogenic endothelial cells (HECs) within the developing embryo. Understanding the origin and ontogeny of HSPCs is of considerable interest and potential therapeutic value. It has been proposed that the murine placenta contains HECs that differentiate into HSPCs. However, during human gestation HSPCs arise in the aorta considerably earlier than when they can first be detected in the placenta, suggesting that the placenta may primarily serve as a niche. We found that the Runx1 transcription factor, which is required to generate HSPCs from HECs, is not expressed by mouse placental ECs. To definitively determine whether the mouse placenta is a site of HSPC emergence, we performed lineage tracing experiments with a Hoxa13Cre allele that specifically labels ECs in the placenta and umbilical cord (UC), but not in the yolk sac or embryo. Immunostaining revealed Hoxa13Cre lineage-traced HECs and HSPCs in the UC, a known site of HECs, but not the placenta. Consistent with these findings, ECs harvested from the E10.5 aorta and UC, but not the placenta, gave rise to hematopoietic cells ex vivo, while colony forming assays using E14.5 fetal liver revealed only 2% of HSPCs arose from Hoxa13-expressing precursors. In contrast, the pan-EC Cdh5-CreERT2 allele labeled most HSPCs in the mouse placenta. Lastly, we found that RUNX1 and other HEC genes were not expressed in first-trimester human placenta villous ECs, suggesting that human placenta is not hemogenic. Our findings demonstrate that the placenta functions as a site for expansion of HSPCs that arise within the embryo proper and is not a primary site of HSPC emergence.
Collapse
Affiliation(s)
- Xiaowen Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Martin Dominguez
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alan T. Tang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jenna Bockman
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jisheng Yang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sneha Mani
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Chin Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mei Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Triloshan Thillaikumaran
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Patricia Mericko-Ishizuka
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Monica Mainigi
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nancy A. Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark L. Kahn
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
3
|
Daems M, Ponomarev LC, Simoes-Faria R, Nobis M, Scheele CLGJ, Luttun A, Ghesquière B, Zwijsen A, Jones EAV. Smad1/5 is acetylated in the dorsal aortae of the mouse embryo before the onset of blood flow, driving early arterial gene expression. Cardiovasc Res 2024; 120:2078-2091. [PMID: 39253943 DOI: 10.1093/cvr/cvae201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/22/2024] [Indexed: 09/11/2024] Open
Abstract
AIMS During embryonic development, arteriovenous (AV) differentiation ensures proper blood vessel formation and maturation. Defects in arterial or venous identity cause inappropriate fusion of vessels, resulting in atypical shunts, so-called AV malformations (AVMs). Currently, the mechanism behind AVM formation remains unclear, and treatment options are fairly limited. Mammalian AV differentiation is initiated before the onset of blood flow in the embryo; however, this pre-flow mechanism is poorly understood. Here, we aimed to unravel the role of Smad1/5 signalling in pre-flow arterial identity and, in the process, uncovered an unexpected control mechanism of Smad1/5 signalling. METHODS AND RESULTS We establish that despite Notch1 being expressed in the pre-flow mouse embryo, it is not activated, nor is it necessary for the expression of the earliest arterial genes in the dorsal aortae (i.e. Hey1 and Gja4). Furthermore, interrupting blood flow by using the Ncx1 KO model completely prevents the activation of Notch1 signalling, suggesting a strong role of shear stress in maintaining arterial identity. We demonstrate that early expression of Hey1 and Gja4 requires SMAD1/5 signalling. Using embryo cultures, we show that Smad1/5 signalling is activated through the Alk1/Alk5/transforming growth factor (TGF)βR2 receptor complex, with TGFβ1 as a necessary ligand. Furthermore, our findings demonstrate that early arterial gene expression requires the acetylation of Smad1/5 proteins, rendering them more sensitive to TGFβ1 stimulation. Blocking acetyl-CoA production prevents pre-flow arterial expression of Hey1 and Gja4, while stabilizing acetylation rescues their expression. CONCLUSION Our findings highlight the importance of the acetyl-CoA production in the cell and provide a novel control mechanism of Smad1/5 signalling involving protein acetylation. As disturbed canonical Smad1/5 signalling is involved in several vascular conditions, our results offer new insights in treatment options for circumventing canonical Smad1/5 signalling.
Collapse
MESH Headings
- Animals
- Smad5 Protein/metabolism
- Smad5 Protein/genetics
- Smad1 Protein/metabolism
- Smad1 Protein/genetics
- Acetylation
- Gene Expression Regulation, Developmental
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Mice, Knockout
- Regional Blood Flow
- Signal Transduction
- Aorta/metabolism
- Aorta/physiopathology
- Aorta/embryology
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Protein Processing, Post-Translational
- Transforming Growth Factor beta1/metabolism
- Transforming Growth Factor beta1/genetics
- Mice, Inbred C57BL
- Receptors, Transforming Growth Factor beta/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Mice
- Sodium-Calcium Exchanger
Collapse
Affiliation(s)
- Margo Daems
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Ljuba C Ponomarev
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Rita Simoes-Faria
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - Max Nobis
- Intravital Imaging Expertise Centre, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Colinda L G J Scheele
- Department of Oncology, Laboratory for Intravital Imaging and Dynamics of Tumour Progression, VIB Centre for Cancer Biology, KU Leuven, 3000 Leuven, Belgium
| | - Aernout Luttun
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Expertise Centre, VIB Centre for Cancer Biology, 3000 Leuven, Belgium
- Department of Cellular and Molecular Medicine, Laboratory of Applied Mass Spectrometry, KU Leuven, 3000 Leuven, Belgium
| | - An Zwijsen
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, KU Leuven, Herestraat 49, bus 911, 3000 Leuven, Belgium
- Department of Cardiology, Maastricht University, CARIM School for Cardiovascular Diseases, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
4
|
Home P, Ghosh A, Kumar RP, Ray S, Gunewardena S, Kumar R, Dasgupta P, Roy N, Saha A, Ouseph MM, Leone GW, Paul S. A Single Trophoblast Layer Acts as the Gatekeeper at the Endothelial-Hematopoietic Crossroad in the Placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603303. [PMID: 39071312 PMCID: PMC11275844 DOI: 10.1101/2024.07.12.603303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
During embryonic development the placental vasculature acts as a major hematopoietic niche, where endothelial to hematopoietic transition ensures emergence of hematopoietic stem cells (HSCs). However, the molecular mechanisms that regulate the placental hematoendothelial niche are poorly understood. Using a parietal trophoblast giant cell (TGC)-specific knockout mouse model and single-cell RNA-sequencing, we show that the paracrine factors secreted by the TGCs are critical in the development of this niche. Disruptions in the TGC-specific paracrine signaling leads to the loss of HSC population and the concomitant expansion of a KDR+/DLL4+/PROM1+ hematoendothelial cell-population in the placenta. Combining single-cell transcriptomics and receptor-ligand pair analyses, we also define the parietal TGC-dependent paracrine signaling network and identify Integrin signaling as a fundamental regulator of this process. Our study elucidates novel mechanisms by which non-autonomous signaling from the primary parietal TGCs maintain the delicate placental hematopoietic-angiogenic balance and ensures embryonic and extraembryonic development.
Collapse
Affiliation(s)
- Pratik Home
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: XenoTech, A BioIVT Company, 1101 W Cambridge Cir Dr, Kansas City, KS 66103
| | - Ananya Ghosh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Present address: Department of Urology, University of California San Francisco, 35, Medical 12 Center Way, San Francisco, CA 94143
| | - Ram Parikshan Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soma Ray
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Rajnish Kumar
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Namrata Roy
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Abhik Saha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Madhu M. Ouseph
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065
| | - Gustavo W. Leone
- Department of Biochemistry, Medical College of Wisconsin, WI 53226, USA
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Institute for Reproductive Health and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Berg K, Gorham J, Lundt F, Seidman J, Brueckner M. Endocardial primary cilia and blood flow are required for regulation of EndoMT during endocardial cushion development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594405. [PMID: 38798559 PMCID: PMC11118576 DOI: 10.1101/2024.05.15.594405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Blood flow is critical for heart valve formation, and cellular mechanosensors are essential to translate flow into transcriptional regulation of development. Here, we identify a role for primary cilia in vivo in the spatial regulation of cushion formation, the first stage of valve development, by regionally controlling endothelial to mesenchymal transition (EndoMT) via modulation of Kruppel-like Factor 4 (Klf4) . We find that high shear stress intracardiac regions decrease endocardial ciliation over cushion development, correlating with KLF4 downregulation and EndoMT progression. Mouse embryos constitutively lacking cilia exhibit a blood-flow dependent accumulation of KLF4 in these regions, independent of upstream left-right abnormalities, resulting in impaired cushion cellularization. snRNA-seq revealed that cilia KO endocardium fails to progress to late-EndoMT, retains endothelial markers and has reduced EndoMT/mesenchymal genes that KLF4 antagonizes. Together, these data identify a mechanosensory role for endocardial primary cilia in cushion development through regional regulation of KLF4.
Collapse
|
6
|
Sun C, Zheng S, Perry JSA, Norris GT, Cheng M, Kong F, Skyberg R, Cang J, Erisir A, Kipnis J, Hill DL. Maternal diet during early gestation influences postnatal taste activity-dependent pruning by microglia. J Exp Med 2023; 220:e20212476. [PMID: 37733279 PMCID: PMC10512853 DOI: 10.1084/jem.20212476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/08/2023] [Accepted: 08/02/2023] [Indexed: 09/22/2023] Open
Abstract
A key process in central sensory circuit development involves activity-dependent pruning of exuberant terminals. Here, we studied gustatory terminal field maturation in the postnatal mouse nucleus of the solitary tract (NST) during normal development and in mice where their mothers were fed a low NaCl diet for a limited period soon after conception. Pruning of terminal fields of gustatory nerves in controls involved the complement system and is likely driven by NaCl-elicited taste activity. In contrast, offspring of mothers with an early dietary manipulation failed to prune gustatory terminal fields even though peripheral taste activity developed normally. The ability to prune in these mice was rescued by activating myeloid cells postnatally, and conversely, pruning was arrested in controls with the loss of myeloid cell function. The altered pruning and myeloid cell function appear to be programmed before the peripheral gustatory system is assembled and corresponds to the embryonic period when microglia progenitors derived from the yolk sac migrate to and colonize the brain.
Collapse
Affiliation(s)
- Chengsan Sun
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Shuqiu Zheng
- Division of Nephrology, University School of Medicine, Charlottesville, VA, USA
| | - Justin S A Perry
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Geoffrey T Norris
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Mei Cheng
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Fanzhen Kong
- Department of Anatomy, Binzhou Medical University, Yantai, China
| | - Rolf Skyberg
- Institute of Neuroscience, University of Oregon , Eugene, OR, USA
| | - Jianhua Cang
- Departments of Psychology and Biology, University of Virginia, Charlottesville, VA, USA
| | - Alev Erisir
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - David L Hill
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
7
|
Dimitrov AG. Resting membrane state as an interplay of electrogenic transporters with various pumps. Pflugers Arch 2023; 475:1113-1128. [PMID: 37468808 DOI: 10.1007/s00424-023-02838-4] [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: 05/05/2023] [Revised: 06/26/2023] [Accepted: 07/06/2023] [Indexed: 07/21/2023]
Abstract
In this study, a new idea that electrogenic transporters determine cell resting state is presented. The previous assumption was that pumps, especially the sodium one, determine it. The latter meets difficulties, because it violates the law of conservation of energy; also a significant deficit of pump activity is reported. The amount of energy carried by a single ATP molecule reflects the potential of the inner mitochondrial membrane, which is about -200 mV. If pumps enforce a resting membrane potential that is more than twice smaller, then the majority of energy stored in ATP would be dissipated by each pump turning. However, this problem could be solved if control is transferred from pumps to something else, e.g., electrogenic transporters. Then pumps would transfer the energy to the ionic gradient without losses, while the cell surface membrane potential would be associated with the reversal potential of some electrogenic transporters. A minimal scheme of this type would include a sodium-calcium exchanger as well as sodium and calcium pumps. However, note that calcium channels and pumps are positioned along both intracellular organelles and the surface membrane. Therefore, the above-mentioned scheme would involve them as well as possible intercellular communications. Such schemes where various kinds of pumps are assumed to work in parallel may explain, to a great extent, the slow turning rate of the individual members. Interaction of pumps and transporters positioned at distant biological membranes with various forms of energy transfer between them may thus result in hypoxic/reperfusion injury, different kinds of muscle fatigue, and nerve-glia interactions.
Collapse
Affiliation(s)
- A G Dimitrov
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113, Sofia, Bulgaria.
| |
Collapse
|
8
|
Kobayashi M, Yoshimoto M. Multiple waves of fetal-derived immune cells constitute adult immune system. Immunol Rev 2023; 315:11-30. [PMID: 36929134 PMCID: PMC10754384 DOI: 10.1111/imr.13192] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
It has been over three decades since Drs. Herzenberg and Herzenberg proposed the layered immune system hypothesis, suggesting that different types of stem cells with distinct hematopoietic potential produce specific immune cells. This layering of immune system development is now supported by recent studies showing the presence of fetal-derived immune cells that function in adults. It has been shown that various immune cells arise at different embryonic ages via multiple waves of hematopoiesis from special endothelial cells (ECs), referred to as hemogenic ECs. However, it remains unknown whether these fetal-derived immune cells are produced by hematopoietic stem cells (HSCs) during the fetal to neonatal period. To address this question, many advanced tools have been used, including lineage-tracing mouse models, cellular barcoding techniques, clonal assays, and transplantation assays at the single-cell level. In this review, we will review the history of the search for the origins of HSCs, B-1a progenitors, and mast cells in the mouse embryo. HSCs can produce both B-1a and mast cells within a very limited time window, and this ability declines after embryonic day (E) 14.5. Furthermore, the latest data have revealed that HSC-independent adaptive immune cells exist in adult mice, which implies more complicated developmental pathways of immune cells. We propose revised road maps of immune cell development.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
9
|
Dominguez MH, Krup AL, Muncie JM, Bruneau BG. Graded mesoderm assembly governs cell fate and morphogenesis of the early mammalian heart. Cell 2023; 186:479-496.e23. [PMID: 36736300 PMCID: PMC10091855 DOI: 10.1016/j.cell.2023.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/07/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023]
Abstract
Using four-dimensional whole-embryo light sheet imaging with improved and accessible computational tools, we longitudinally reconstruct early murine cardiac development at single-cell resolution. Nascent mesoderm progenitors form opposing density and motility gradients, converting the temporal birth sequence of gastrulation into a spatial anterolateral-to-posteromedial arrangement. Migrating precardiac mesoderm does not strictly preserve cellular neighbor relationships, and spatial patterns only become solidified as the cardiac crescent emerges. Progenitors undergo a mesenchymal-to-epithelial transition, with a first heart field (FHF) ridge apposing a motile juxta-cardiac field (JCF). Anchored along the ridge, the FHF epithelium rotates the JCF forward to form the initial heart tube, along with push-pull morphodynamics of the second heart field. In Mesp1 mutants that fail to make a cardiac crescent, mesoderm remains highly motile but directionally incoherent, resulting in density gradient inversion. Our practicable live embryo imaging approach defines spatial origins and behaviors of cardiac progenitors and identifies their unanticipated morphological transitions.
Collapse
Affiliation(s)
- Martin H Dominguez
- Gladstone Institutes, San Francisco, CA, USA; Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA; Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA.
| | - Alexis Leigh Krup
- Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | | | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA, USA; Roddenberry Center for Stem Cell Biology and Medicine at Gladstone, San Francisco, CA, USA; Department of Pediatrics, Cardiovascular Research Institute, Institute for Human Genetics, and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
10
|
Fan W, Sun X, Yang C, Wan J, Luo H, Liao B. Pacemaker activity and ion channels in the sinoatrial node cells: MicroRNAs and arrhythmia. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:151-167. [PMID: 36450332 DOI: 10.1016/j.pbiomolbio.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/13/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The primary pacemaking activity of the heart is determined by a spontaneous action potential (AP) within sinoatrial node (SAN) cells. This unique AP generation relies on two mechanisms: membrane clocks and calcium clocks. Nonhomologous arrhythmias are caused by several functional and structural changes in the myocardium. MicroRNAs (miRNAs) are essential regulators of gene expression in cardiomyocytes. These miRNAs play a vital role in regulating the stability of cardiac conduction and in the remodeling process that leads to arrhythmias. Although it remains unclear how miRNAs regulate the expression and function of ion channels in the heart, these regulatory mechanisms may support the development of emerging therapies. This study discusses the spread and generation of AP in the SAN as well as the regulation of miRNAs and individual ion channels. Arrhythmogenicity studies on ion channels will provide a research basis for miRNA modulation as a new therapeutic target.
Collapse
Affiliation(s)
- Wei Fan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Xuemei Sun
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Chao Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China
| | - Juyi Wan
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Hongli Luo
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| | - Bin Liao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Jiangyang District, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
11
|
McAndry C, Collins M, Tills O, Spicer JI, Truebano M. Regulation of gene expression during ontogeny of physiological function in the brackishwater amphipod Gammarus chevreuxi. Mar Genomics 2022; 63:100948. [PMID: 35427917 DOI: 10.1016/j.margen.2022.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Abstract
Embryonic development is a complex process involving the co-ordinated onset and integration of multiple morphological features and physiological functions. While the molecular basis of morphological development in embryos is relatively well known for traditional model species, the molecular underpinning of the development of physiological functions is not. Here, we used global gene expression profiling to investigate the transcriptional changes associated with the development of morphological and physiological function in the amphipod crustacean Gammarus chevreuxi. We compared the transcriptomes at three timepoints during the latter half of development, characterised by different stages of the development of heart form and function: 10 days post fertilisation (dpf, Early: no heart structure visible), 15 dpf (Middle: heart present but not fully functional), and 18 dpf (Late: regular heartbeat). Gene expression profiles differed markedly between developmental stages, likely representing a change in the activity of different processes throughout the latter period of G. chevreuxi embryonic development. Differentially expressed genes belonged to one of three distinct clusters based on their expression patterns across development. One of these clusters, which included key genes relating to cardiac contractile machinery and calcium handling, displayed a pattern of sequential up-regulation throughout the developmental period studied. Further analyses of these transcripts could reveal genes that may influence the onset of a regular heartbeat. We also identified morphological and physiological processes that may occur alongside heart development, such as development of digestive caeca and the cuticle. Elucidating the mechanisms underpinning morphological and physiological development of non-model organisms will support improved understanding of conserved mechanisms, addressing the current phylogenetic gap between relatively well known model species.
Collapse
Affiliation(s)
- C McAndry
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - M Collins
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - O Tills
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - J I Spicer
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK
| | - M Truebano
- Marine Biology and Ecology Research Centre, School of Biological and Marine Sciences, University of Plymouth, Drake Circus, Plymouth PL4 8AA, UK.
| |
Collapse
|
12
|
Bone marrow-independent adventitial macrophage progenitor cells contribute to angiogenesis. Cell Death Dis 2022; 13:220. [PMID: 35264563 PMCID: PMC8907187 DOI: 10.1038/s41419-022-04605-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 12/21/2022]
Abstract
Pathological angiogenesis promotes tumor growth, metastasis, and atherosclerotic plaque rupture. Macrophages are key players in these processes. However, whether these macrophages differentiate from bone marrow-derived monocytes or from local vascular wall-resident stem and progenitor cells (VW-SCs) is an unresolved issue of angiogenesis. To answer this question, we analyzed vascular sprouting and alterations in aortic cell populations in mouse aortic ring assays (ARA). ARA culture leads to the generation of large numbers of macrophages, especially within the aortic adventitia. Using immunohistochemical fate-mapping and genetic in vivo-labeling approaches we show that 60% of these macrophages differentiate from bone marrow-independent Ly6c+/Sca-1+ adventitial progenitor cells. Analysis of the NCX−/− mouse model that genetically lacks embryonic circulation and yolk sac perfusion indicates that at least some of those progenitor cells arise yolk sac-independent. Macrophages represent the main source of VEGF in ARA that vice versa promotes the generation of additional macrophages thereby creating a pro-angiogenetic feedforward loop. Additionally, macrophage-derived VEGF activates CD34+ progenitor cells within the adventitial vasculogenic zone to differentiate into CD31+ endothelial cells. Consequently, depletion of macrophages and VEGFR2 antagonism drastically reduce vascular sprouting activity in ARA. In summary, we show that angiogenic activation induces differentiation of macrophages from bone marrow-derived as well as from bone marrow-independent VW-SCs. The latter ones are at least partially yolk sac-independent, too. Those VW-SC-derived macrophages critically contribute to angiogenesis, making them an attractive target to interfere with pathological angiogenesis in cancer and atherosclerosis as well as with regenerative angiogenesis in ischemic cardiovascular disorders.
Collapse
|
13
|
Ottolia M, John S, Hazan A, Goldhaber JI. The Cardiac Na + -Ca 2+ Exchanger: From Structure to Function. Compr Physiol 2021; 12:2681-2717. [PMID: 34964124 DOI: 10.1002/cphy.c200031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Ca2+ homeostasis is essential for cell function and survival. As such, the cytosolic Ca2+ concentration is tightly controlled by a wide number of specialized Ca2+ handling proteins. One among them is the Na+ -Ca2+ exchanger (NCX), a ubiquitous plasma membrane transporter that exploits the electrochemical gradient of Na+ to drive Ca2+ out of the cell, against its concentration gradient. In this critical role, this secondary transporter guides vital physiological processes such as Ca2+ homeostasis, muscle contraction, bone formation, and memory to name a few. Herein, we review the progress made in recent years about the structure of the mammalian NCX and how it relates to function. Particular emphasis will be given to the mammalian cardiac isoform, NCX1.1, due to the extensive studies conducted on this protein. Given the degree of conservation among the eukaryotic exchangers, the information highlighted herein will provide a foundation for our understanding of this transporter family. We will discuss gene structure, alternative splicing, topology, regulatory mechanisms, and NCX's functional role on cardiac physiology. Throughout this article, we will attempt to highlight important milestones in the field and controversial topics where future studies are required. © 2021 American Physiological Society. Compr Physiol 12:1-37, 2021.
Collapse
Affiliation(s)
- Michela Ottolia
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Scott John
- Department of Medicine (Cardiology), UCLA, Los Angeles, California, USA
| | - Adina Hazan
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Joshua I Goldhaber
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
14
|
Azzoni E, Frontera V, Anselmi G, Rode C, James C, Deltcheva EM, Demian AS, Brown J, Barone C, Patelli A, Harman JR, Nicholls M, Conway SJ, Morrissey E, Jacobsen SEW, Sparrow DB, Harris AL, Enver T, de Bruijn MFTR. The onset of circulation triggers a metabolic switch required for endothelial to hematopoietic transition. Cell Rep 2021; 37:110103. [PMID: 34910918 PMCID: PMC8692754 DOI: 10.1016/j.celrep.2021.110103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 12/14/2022] Open
Abstract
Hematopoietic stem cells (HSCs) emerge during development from the vascular wall of the main embryonic arteries. The onset of circulation triggers several processes that provide critical external factors for HSC generation. Nevertheless, it is not fully understood how and when the onset of circulation affects HSC emergence. Here we show that in Ncx1-/- mouse embryos devoid of circulation the HSC lineage develops until the phenotypic pro-HSC stage. However, these cells reside in an abnormal microenvironment, fail to activate the hematopoietic program downstream of Runx1, and are functionally impaired. Single-cell transcriptomics shows that during the endothelial-to-hematopoietic transition, Ncx1-/- cells fail to undergo a glycolysis to oxidative phosphorylation metabolic switch present in wild-type cells. Interestingly, experimental activation of glycolysis results in decreased intraembryonic hematopoiesis. Our results suggest that the onset of circulation triggers metabolic changes that allow HSC generation to proceed.
Collapse
Affiliation(s)
- Emanuele Azzoni
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Vincent Frontera
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Giorgio Anselmi
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Christina Rode
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Chela James
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Elitza M Deltcheva
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Atanasiu S Demian
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - John Brown
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK
| | - Cristiana Barone
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, 20900, Italy
| | - Arianna Patelli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, 20900, Italy
| | - Joe R Harman
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew Nicholls
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, IN 46033, USA
| | - Edward Morrissey
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Sten Eirik W Jacobsen
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK; Hematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK; Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine and Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Duncan B Sparrow
- Department of Physiology, Anatomy and Genetics, BHF Centre of Research Excellence, University of Oxford, Oxford, OX1 3PT, UK
| | - Adrian L Harris
- Department of Oncology, Molecular Oncology Laboratories, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, WC1E 6DD, UK; Division of Molecular Medicine and Gene Therapy, Lund University, Lund, 22184, Sweden
| | - Marella F T R de Bruijn
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| |
Collapse
|
15
|
Lotteau S, Zhang R, Hazan A, Grabar C, Gonzalez D, Aynaszyan S, Philipson KD, Ottolia M, Goldhaber JI. Acute Genetic Ablation of Cardiac Sodium/Calcium Exchange in Adult Mice: Implications for Cardiomyocyte Calcium Regulation, Cardioprotection, and Arrhythmia. J Am Heart Assoc 2021; 10:e019273. [PMID: 34472363 PMCID: PMC8649274 DOI: 10.1161/jaha.120.019273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Sodium‐calcium (Ca2+) exchanger isoform 1 (NCX1) is the dominant Ca2+ efflux mechanism in cardiomyocytes and is critical to maintaining Ca2+ homeostasis during excitation‐contraction coupling. NCX1 activity has been implicated in the pathogenesis of cardiovascular diseases, but a lack of specific NCX1 blockers complicates experimental interpretation. Our aim was to develop a tamoxifen‐inducible NCX1 knockout (KO) mouse to investigate compensatory adaptations of acute ablation of NCX1 on excitation‐contraction coupling and intracellular Ca2+ regulation, and to examine whether acute KO of NCX1 confers resistance to triggered arrhythmia and ischemia/reperfusion injury. Methods and Results We used the α‐myosin heavy chain promoter (Myh6)‐MerCreMer promoter to create a tamoxifen‐inducible cardiac‐specific NCX1 KO mouse. Within 1 week of tamoxifen injection, NCX1 protein expression and current were dramatically reduced. Diastolic Ca2+ increased despite adaptive reductions in Ca2+ current and action potential duration and compensatory increases in excitation‐contraction coupling gain, sarcoplasmic reticulum Ca2+ ATPase 2 and plasma membrane Ca2+ ATPase. As these adaptations progressed over 4 weeks, diastolic Ca2+ normalized and SR Ca2+ load increased. Left ventricular function remained normal, but mild fibrosis and hypertrophy developed. Transcriptomics revealed modification of cardiovascular‐related gene networks including cell growth and fibrosis. NCX1 KO reduced spontaneous action potentials triggered by delayed afterdepolarizations and reduced scar size in response to ischemia/reperfusion. Conclusions Tamoxifen‐inducible NCX1 KO mice adapt to acute genetic ablation of NCX1 by reducing Ca2+ influx, increasing alternative Ca2+ efflux pathways, and increasing excitation‐contraction coupling gain to maintain contractility at the cost of mild Ca2+‐activated hypertrophy and fibrosis and decreased survival. Nevertheless, KO myocytes are protected against spontaneous action potentials and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sabine Lotteau
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Rui Zhang
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Adina Hazan
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Christina Grabar
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | - Devina Gonzalez
- Smidt Heart Institute Cedars-Sinai Medical Center Los Angeles CA
| | | | - Kenneth D Philipson
- Department of Physiology David Geffen School of Medicine at UCLA Los Angeles CA
| | - Michela Ottolia
- Division of Molecular Medicine Department of Anesthesiology and Perioperative Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| | | |
Collapse
|
16
|
De novo generation of macrophage from placenta-derived hemogenic endothelium. Dev Cell 2021; 56:2121-2133.e6. [PMID: 34197725 DOI: 10.1016/j.devcel.2021.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 01/31/2023]
Abstract
Macrophages play pivotal roles in immunity, hematopoiesis, and tissue homeostasis. In mammals, macrophages have been shown to originate from yolk-sac-derived erythro-myeloid progenitors and aorta-gonad-mesonephros (AGM)-derived hematopoietic stem cells. However, whether macrophages can arise from other embryonic sites remains unclear. Here, using single-cell RNA sequencing, we profile the transcriptional landscape of mouse fetal placental hematopoiesis. We uncover and experimentally validate that a CD44+ subpopulation of placental endothelial cells (ECs) exhibits hemogenic potential. Importantly, lineage tracing using the newly generated Hoxa13 reporter line shows that Hoxa13-labeled ECs can produce placental macrophages, named Hofbauer cell (HBC)-like cells. Furthermore, we identify two subtypes of HBC-like cells, and cell-cell interaction analysis identifies their potential roles in angiogenesis and antigen presentation, separately. Our study provides a comprehensive understanding of placental hematopoiesis and highlights the placenta as a source of macrophages, which has important implications for both basic and translational research.
Collapse
|
17
|
Liang X, Wu H, Colt M, Guo X, Pluimer B, Zeng J, Dong S, Zhao Z. Microglia and its Genetics in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:676-688. [PMID: 34749609 PMCID: PMC9790807 DOI: 10.2174/1567205018666211105140732] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/30/2021] [Accepted: 07/03/2021] [Indexed: 12/27/2022]
Abstract
Alzheimer's Disease (AD) is the most prevalent form of dementia across the world. While its discovery and pathological manifestations are centered on protein aggregations of amyloid- beta (Aβ) and hyperphosphorylated tau protein, neuroinflammation has emerged in the last decade as a main component of the disease in terms of both pathogenesis and progression. As the main innate immune cell type in the central nervous system (CNS), microglia play a very important role in regulating neuroinflammation, which occurs commonly in neurodegenerative conditions, including AD. Under inflammatory response, microglia undergo morphological changes and status transition from homeostatic to activated forms. Different microglia subtypes displaying distinct genetic profiles have been identified in AD, and these signatures often link to AD risk genes identified from the genome-wide association studies (GWAS), such as APOE and TREM2. Furthermore, many AD risk genes are highly enriched in microglia and specifically influence the functions of microglia in pathogenesis, e.g. releasing inflammatory cytokines and clearing Aβ. Therefore, building up a landscape of these risk genes in microglia, based on current preclinical studies and in the context of their pathogenic or protective effects, would largely help us to understand the complex etiology of AD and provide new insight into the unmet need for effective treatment.
Collapse
Affiliation(s)
- Xinyan Liang
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Haijian Wu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Mark Colt
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Xinying Guo
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Brock Pluimer
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Jianxiong Zeng
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Shupeng Dong
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Shanghai Institute of Immunology; Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| |
Collapse
|
18
|
Daems M, Peacock HM, Jones EAV. Fluid flow as a driver of embryonic morphogenesis. Development 2020; 147:147/15/dev185579. [PMID: 32769200 DOI: 10.1242/dev.185579] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fluid flow is a powerful morphogenic force during embryonic development. The physical forces created by flowing fluids can either create morphogen gradients or be translated by mechanosensitive cells into biological changes in gene expression. In this Primer, we describe how fluid flow is created in different systems and highlight the important mechanosensitive signalling pathways involved for sensing and transducing flow during embryogenesis. Specifically, we describe how fluid flow helps establish left-right asymmetry in the early embryo and discuss the role of flow of blood, lymph and cerebrospinal fluid in sculpting the embryonic cardiovascular and nervous system.
Collapse
Affiliation(s)
- Margo Daems
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Hanna M Peacock
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
19
|
Chu L, Yin H, Gao L, Gao L, Xia Y, Zhang C, Chen Y, Liu T, Huang J, Boheler KR, Zhou Y, Yang HT. Cardiac Na +-Ca 2+ exchanger 1 (ncx1h) is critical for the ventricular cardiomyocyte formation via regulating the expression levels of gata4 and hand2 in zebrafish. SCIENCE CHINA-LIFE SCIENCES 2020; 64:255-268. [PMID: 32648190 DOI: 10.1007/s11427-019-1706-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/22/2020] [Indexed: 10/23/2022]
Abstract
Ca2+ signaling is critical for heart development; however, the precise roles and regulatory pathways of Ca2+ transport proteins in cardiogenesis remain largely unknown. Sodium-calcium exchanger 1 (Ncx1) is responsible for Ca2+ efflux in cardiomyocytes. It is involved in cardiogenesis, while the mechanism is unclear. Here, using the forward genetic screening in zebrafish, we identified a novel mutation at a highly-conserved leucine residue in ncx1 gene (mutantLDD353/ncx1hL154P) that led to smaller hearts with reduced heart rate and weak contraction. Mechanistically, the number of ventricular but not atrial cardiomyocytes was reduced in ncx1hL154P zebrafish. These defects were mimicked by knockdown or knockout of ncx1h. Moreover, ncx1hL154P had cytosolic and mitochondrial Ca2+ overloading and Ca2+ transient suppression in cardiomyocytes. Furthermore, ncx1hL154P and ncx1h morphants downregulated cardiac transcription factors hand2 and gata4 in the cardiac regions, while overexpression of hand2 and gata4 partially rescued cardiac defects including the number of ventricular myocytes. These findings demonstrate an essential role of the novel 154th leucine residue in the maintenance of Ncx1 function in zebrafish, and reveal previous unrecognized critical roles of the 154th leucine residue and Ncx1 in the formation of ventricular cardiomyocytes by at least partially regulating the expression levels of gata4 and hand2.
Collapse
Affiliation(s)
- Liming Chu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Huimin Yin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Lei Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Li Gao
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Xia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Chiyuan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Yi Chen
- State Key Laboratory for Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tingxi Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Jijun Huang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China.,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China
| | - Kenneth R Boheler
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yong Zhou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China. .,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China.
| | - Huang-Tian Yang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Laboratory of Molecular Cardiology and Laboratory of Development and Diseases, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences (CAS), CAS, Shanghai, 200031, China. .,Institute for Stem Cell and Regeneration, CAS, Beijing, 100101, China.
| |
Collapse
|
20
|
Tyser RCV, Srinivas S. The First Heartbeat-Origin of Cardiac Contractile Activity. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037135. [PMID: 31767652 DOI: 10.1101/cshperspect.a037135] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The amniote embryonic heart starts as a crescent of mesoderm that transitions through a midline linear heart tube in the course of developing into the four chambered heart. It is unusual in having to contract rhythmically while still undergoing extensive morphogenetic remodeling. Advances in imaging have allowed us to determine when during development this contractile activity starts. In the mouse, focal regions of contractions can be detected as early as the cardiac crescent stage. Calcium transients, required to trigger contraction, can be detected even earlier, prior to contraction. In this review, we outline what is currently known about how this early contractile function is initiated and the impact early contractile function has on cardiac development.
Collapse
Affiliation(s)
- Richard C V Tyser
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, United Kingdom
| |
Collapse
|
21
|
Chin MT, Conway SJ. Role of Tafazzin in Mitochondrial Function, Development and Disease. J Dev Biol 2020; 8:jdb8020010. [PMID: 32456129 PMCID: PMC7344621 DOI: 10.3390/jdb8020010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
Tafazzin, an enzyme associated with the rare inherited x-linked disorder Barth Syndrome, is a nuclear encoded mitochondrial transacylase that is highly conserved across multiple species and plays an important role in mitochondrial function. Numerous studies have elucidated the mechanisms by which Tafazzin affects mitochondrial function, but its effects on development and susceptibility to adult disease are incompletely understood. The purpose of this review is to highlight previous functional studies across a variety of model organisms, introduce recent studies that show an important role in development, and also to provide an update on the role of Tafazzin in human disease. The profound effects of Tafazzin on cardiac development and adult cardiac homeostasis will be emphasized. These studies underscore the importance of mitochondrial function in cardiac development and disease, and also introduce the concept of Tafazzin as a potential therapeutic modality.
Collapse
Affiliation(s)
- Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
- Correspondence: (M.T.C.); (S.J.C.); Tel.: +1-617-636-8776 (M.T.C.); +1-317-278-8780 (S.J.C.)
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: (M.T.C.); (S.J.C.); Tel.: +1-617-636-8776 (M.T.C.); +1-317-278-8780 (S.J.C.)
| |
Collapse
|
22
|
Hong JM, Hu YD, Chai XQ, Tang CL. Role of activin receptor-like kinase 1 in vascular development and cerebrovascular diseases. Neural Regen Res 2020; 15:1807-1813. [PMID: 32246621 PMCID: PMC7513971 DOI: 10.4103/1673-5374.280305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Activin receptor-like kinase 1 (ALK1) is a transmembrane serine/threonine receptor kinase of the transforming growth factor beta (TGFβ) receptor superfamily. ALK1 is specifically expressed in vascular endothelial cells, and its dynamic changes are closely related to the proliferation of endothelial cells, the recruitment of pericytes to blood vessels, and functional differentiation during embryonic vascular development. The pathophysiology of many cerebrovascular diseases is today understood as a disorder of endothelial cell function and an imbalance in the proportion of vascular cells. Indeed, mutations in ALK1 and its co-receptor endoglin are major genetic risk factors for vascular arteriovenous malformation. Many studies have shown that ALK1 is closely related to the development of cerebral aneurysms, arteriovenous malformations, and cerebral atherosclerosis. In this review, we describe the various roles of ALK1 in the regulation of angiogenesis and in the maintenance of cerebral vascular homeostasis, and we discuss its relationship to functional dysregulation in cerebrovascular diseases. This review should provide new perspectives for basic research on cerebrovascular diseases and offer more effective targets and strategies for clinical diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Jun-Mou Hong
- Department of Vascular Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian Province, China
| | - Yi-Da Hu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiao-Qing Chai
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Chao-Liang Tang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
23
|
Molinaro P, Natale S, Serani A, Calabrese L, Secondo A, Tedeschi V, Valsecchi V, Pannaccione A, Scorziello A, Annunziato L. Genetically modified mice to unravel physiological and pathophysiological roles played by NCX isoforms. Cell Calcium 2020; 87:102189. [PMID: 32199207 DOI: 10.1016/j.ceca.2020.102189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 11/30/2022]
Abstract
Since the discovery of the three isoforms of the Na+/Ca2+ exchanger, NCX1, NCX2 and NCX3 in 1990s, many studies have been devoted to identifying their specific roles in different tissues under several physiological or pathophysiological conditions. In particular, several seminal experimental works laid the foundation for better understanding gene and protein structures, tissue distribution, and regulatory functions of each antiporter isoform. On the other hand, despite the efforts in the development of specific compounds selectively targeting NCX1, NCX2 or NCX3 to test their physiological or pathophysiological roles, several drawbacks hampered the achievement of these goals. In fact, at present no isoform-specific compounds have been yet identified. Moreover, these compounds, despite their potency, possess some nonspecific actions against other ion antiporters, ion channels, and channel receptors. As a result, it is difficult to discriminate direct effects of inhibition/activation of NCX isoforms from the inhibitory or stimulatory effects exerted on other antiporters, channels, receptors, or enzymes. To overcome these difficulties, some research groups used transgenic, knock-out and knock-in mice for NCX isoforms as the most straightforward and fruitful strategy to characterize the biological role exerted by each antiporter isoform. The present review will survey the techniques used to study the roles of NCXs and the current knowledge obtained from these genetic modified mice focusing on the advantages obtained with these strategies in understanding the contribution exerted by each isoform.
Collapse
Affiliation(s)
- Pasquale Molinaro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy.
| | - Silvia Natale
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Angelo Serani
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Lucrezia Calabrese
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Valeria Valsecchi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | - Antonella Scorziello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, "Federico II" University of Naples, 80131, Naples, Italy
| | | |
Collapse
|
24
|
Bujko K, Kucia M, Ratajczak J, Ratajczak MZ. Hematopoietic Stem and Progenitor Cells (HSPCs). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:49-77. [PMID: 31898781 DOI: 10.1007/978-3-030-31206-0_3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) isolated from bone marrow have been successfully employed for 50 years in hematological transplantations. Currently, these cells are more frequently isolated from mobilized peripheral blood or umbilical cord blood. In this chapter, we overview several topics related to these cells including their phenotype, methods for isolation, and in vitro and in vivo assays to evaluate their proliferative potential. The successful clinical application of HSPCs is widely understood to have helped establish the rationale for the development of stem cell therapies and regenerative medicine.
Collapse
Affiliation(s)
- Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Magda Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Janina Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA. .,Department of Regenerative Medicine, Center for Preclinical Research and Technology, Warsaw Medical University, Warsaw, Poland.
| |
Collapse
|
25
|
Li S, Chopra A, Keung W, Chan CWY, Costa KD, Kong CW, Hajjar RJ, Chen CS, Li RA. Sarco/endoplasmic reticulum Ca2+-ATPase is a more effective calcium remover than sodium-calcium exchanger in human embryonic stem cell-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2019; 317:H1105-H1115. [DOI: 10.1152/ajpheart.00540.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human pluripotent stem cell (hPSCs)-derived ventricular (V) cardiomyocytes (CMs) display immature Ca2+–handing properties with smaller transient amplitudes and slower kinetics due to such differences in crucial Ca2+-handling proteins as the poor sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump but robust Na+-Ca2+ exchanger (NCX) activities in human embryonic stem cell (ESC)-derived VCMs compared with adult. Despite their fundamental importance in excitation-contraction coupling, the relative contribution of SERCA and NCX to Ca2+-handling of hPSC-VCMs remains unexplored. We systematically altered the activities of SERCA and NCX in human embryonic stem cell-derived ventricular cardiomyocytes (hESC-VCMs) and their engineered microtissues, followed by examining the resultant phenotypic consequences. SERCA overexpression in hESC-VCMs shortened the decay of Ca2+ transient at low frequencies (0.5 Hz) without affecting the amplitude, SR Ca2+ content and Ca2+ baseline. Interestingly, short hairpin RNA-based NCX suppression did not prolong the transient decay, indicating a compensatory response for Ca2+ removal. Although hESC-VCMs and their derived microtissues exhibited negative frequency-transient/force responses, SERCA overexpression rendered them less negative at high frequencies (>2 Hz) by accelerating Ca2+ sequestration. We conclude that for hESC-VCMs and their microtissues, SERCA, rather than NCX, is the main Ca2+ remover during diastole; poor SERCA expression is the leading cause for immature negative-frequency/force responses, which can be partially reverted by forced expression. Combinatorial approach to mature calcium handling in hESC-VCMs may help shed further mechanistic insights. NEW & NOTEWORTHY In this study of human pluripotent stem cell-derived cardiomyocytes, we studied the role of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) and Na+-Ca2+ exchanger (NCX) in Ca2+ handling. Our data support the notion that SERCA is more effective in cytosolic calcium removal than the NCX.
Collapse
Affiliation(s)
- Sen Li
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Anant Chopra
- Department of Bioengineering, Boston University, Boston, Massachusetts
- Harvard Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts
| | - Wendy Keung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Camie W. Y. Chan
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Kevin D. Costa
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, New York
| | - Chi-Wing Kong
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Roger J. Hajjar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, Manhattan, New York
| | - Christopher S. Chen
- Department of Bioengineering, Boston University, Boston, Massachusetts
- Harvard Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts
| | - Ronald A. Li
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Pokfulam, Hong Kong
- Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
- Ming-Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong
| |
Collapse
|
26
|
Conway SJ, McConnell R, Simmons O, Snider PL. Armadillo-like helical domain containing-4 is dynamically expressed in both the first and second heart fields. Gene Expr Patterns 2019; 34:119077. [PMID: 31655130 DOI: 10.1016/j.gep.2019.119077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/19/2022]
Abstract
Armadillo repeat and Armadillo-like helical domain containing proteins form a large family with diverse and fundamental functions in many eukaryotes. Herein we investigated the spatiotemporal expression pattern of Armadillo-like helical domain containing 4 (or Armh4) as an uncharacterized protein coding mouse gene, within the mouse embryo during the initial stages of heart morphogenesis. We found Armh4 is initially expressed in both first heart field as well as the second heart field progenitors and subsequently within predominantly their cardiomyocyte derivatives. Armh4 expression is initially cardiac-restricted in the developing embryo and is expressed in second heart field subpharyngeal mesoderm prior to cardiomyocyte differentiation, but Armh4 diminishes as the embryonic heart matures into the fetal heart. Armh4 is subsequently expressed in craniofacial structures and neural crest-derived dorsal root and trigeminal ganglia. Whereas lithium chloride-induced stimulation of Wnt/β-catenin signaling elevated Armh4 expression in both second heart field subpharyngeal mesodermal progenitors and outflow tract, right ventricle and atrial cardiomyocytes, neither a systemic loss of Islet-1 nor an absence of cardiac neural crest cells had any effect upon Armh4 expression. These results confirm that Wnt/β-catenin-responsive Armh4 is a useful specific biomarker of the FHF and SHF cardiomyocyte derivatives only.
Collapse
Affiliation(s)
- Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Reagan McConnell
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; School of Biomedical Sciences, University of Ulster, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Olga Simmons
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paige L Snider
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
27
|
An element for development: Calcium signaling in mammalian reproduction and development. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1230-1238. [DOI: 10.1016/j.bbamcr.2019.02.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 11/21/2022]
|
28
|
Shigeta A, Huang V, Zuo J, Besada R, Nakashima Y, Lu Y, Ding Y, Pellegrini M, Kulkarni RP, Hsiai T, Deb A, Zhou B, Nakano H, Nakano A. Endocardially Derived Macrophages Are Essential for Valvular Remodeling. Dev Cell 2019; 48:617-630.e3. [PMID: 30799229 PMCID: PMC6440481 DOI: 10.1016/j.devcel.2019.01.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022]
Abstract
During mammalian embryogenesis, de novo hematopoiesis occurs transiently in multiple anatomical sites including the yolk sac, dorsal aorta, and heart tube. A long-unanswered question is whether these local transient hematopoietic mechanisms are essential for embryonic growth. Here, we show that endocardial hematopoiesis is critical for cardiac valve remodeling as a source of tissue macrophages. Colony formation assay from explanted heart tubes and genetic lineage tracing with the endocardial specific Nfatc1-Cre mouse revealed that hemogenic endocardium is a de novo source of tissue macrophages in the endocardial cushion, the primordium of the cardiac valves. Surface marker characterization, gene expression profiling, and ex vivo phagocytosis assay revealed that the endocardially derived cardiac tissue macrophages play a phagocytic and antigen presenting role. Indeed, genetic ablation of endocardially derived macrophages caused severe valve malformation. Together, these data suggest that transient hemogenic activity in the endocardium is indispensable for the valvular tissue remodeling in the heart.
Collapse
Affiliation(s)
- Ayako Shigeta
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vincent Huang
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan Zuo
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rana Besada
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yasuhiro Nakashima
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yan Lu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajan P Kulkarni
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tzung Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Arjun Deb
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bin Zhou
- Department of Genetics, Pediatrics, and Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Haruko Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Atsushi Nakano
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
29
|
Desgrange A, Le Garrec JF, Meilhac SM. Left-right asymmetry in heart development and disease: forming the right loop. Development 2018; 145:145/22/dev162776. [PMID: 30467108 DOI: 10.1242/dev.162776] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Extensive studies have shown how bilateral symmetry of the vertebrate embryo is broken during early development, resulting in a molecular left-right bias in the mesoderm. However, how this early asymmetry drives the asymmetric morphogenesis of visceral organs remains poorly understood. The heart provides a striking model of left-right asymmetric morphogenesis, undergoing rightward looping to shape an initially linear heart tube and align cardiac chambers. Importantly, abnormal left-right patterning is associated with severe congenital heart defects, as exemplified in heterotaxy syndrome. Here, we compare the mechanisms underlying the rightward looping of the heart tube in fish, chick and mouse embryos. We propose that heart looping is not only a question of direction, but also one of fine-tuning shape. This is discussed in the context of evolutionary and clinical perspectives.
Collapse
Affiliation(s)
- Audrey Desgrange
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Jean-François Le Garrec
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France .,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| |
Collapse
|
30
|
Courchaine K, Rykiel G, Rugonyi S. Influence of blood flow on cardiac development. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 137:95-110. [PMID: 29772208 PMCID: PMC6109420 DOI: 10.1016/j.pbiomolbio.2018.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/06/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022]
Abstract
The role of hemodynamics in cardiovascular development is not well understood. Indeed, it would be remarkable if it were, given the dauntingly complex array of intricately synchronized genetic, molecular, mechanical, and environmental factors at play. However, with congenital heart defects affecting around 1 in 100 human births, and numerous studies pointing to hemodynamics as a factor in cardiovascular morphogenesis, this is not an area in which we can afford to remain in the dark. This review seeks to present the case for the importance of research into the biomechanics of the developing cardiovascular system. This is accomplished by i) illustrating the basics of some of the highly complex processes involved in heart development, and discussing the known influence of hemodynamics on those processes; ii) demonstrating how altered hemodynamic environments have the potential to bring about morphological anomalies, citing studies in multiple animal models with a variety of perturbation methods; iii) providing examples of widely used technological innovations which allow for accurate measurement of hemodynamic parameters in embryos; iv) detailing the results of studies in avian embryos which point to exciting correlations between various hemodynamic manipulations in early development and phenotypic defect incidence in mature hearts; and finally, v) stressing the relevance of uncovering specific biomechanical pathways involved in cardiovascular formation and remodeling under adverse conditions, to the potential treatment of human patients. The time is ripe to unravel the contributions of hemodynamics to cardiac development, and to recognize their frequently neglected role in the occurrence of heart malformation phenotypes.
Collapse
Affiliation(s)
- Katherine Courchaine
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA
| | - Graham Rykiel
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA
| | - Sandra Rugonyi
- Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland OR, USA.
| |
Collapse
|
31
|
Caolo V, Peacock HM, Kasaai B, Swennen G, Gordon E, Claesson-Welsh L, Post MJ, Verhamme P, Jones EA. Shear Stress and VE-Cadherin. Arterioscler Thromb Vasc Biol 2018; 38:2174-2183. [DOI: 10.1161/atvbaha.118.310823] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Vascular fusion represents an important mechanism of vessel enlargement during development; however, its significance in postnatal vessel enlargement is still unknown. During fusion, 2 adjoining vessels merge to share 1 larger lumen. The aim of this research was to identify the molecular mechanism responsible for vascular fusion.
Approach and Results—
We previously showed that both low shear stress and DAPT (
N
-[
N
-(3,5-difluorophenacetyl)-L-alanyl]-
S
-phenylglycine t-butyl ester) treatment in the embryo result in a hyperfused vascular plexus and that increasing shear stress levels could prevent DAPT-induced fusion. We, therefore, investigated vascular endothelial-cadherin (VEC) phosphorylation because this is a common downstream target of low shear stress and DAPT treatment. VEC phosphorylation increases after DAPT treatment and decreased shear stress. The increased phosphorylation occurred independent of the cleavage of the Notch intracellular domain. Increasing shear stress rescues hyperfusion by DAPT treatment by causing the association of the phosphatase vascular endothelial-protein tyrosine phosphatase with VEC, counteracting VEC phosphorylation. Finally, Src (proto-oncogene tyrosine-protein kinase Src) inhibition prevents VEC phosphorylation in endothelial cells and can rescue hyperfusion induced by low shear stress and DAPT treatment. Moesin, a VEC target that was previously reported to mediate endothelial cell rearrangement during lumenization, relocalizes to cell membranes in vascular beds undergoing hyperfusion.
Conclusions—
This study provides the first evidence that VEC phosphorylation, induced by DAPT treatment and low shear stress, is involved in the process of fusion during vascular remodeling.
Collapse
Affiliation(s)
- Vincenza Caolo
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium (V.C., H.M.P., B.K., P.V., E.A.V.J.)
| | - Hanna M. Peacock
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium (V.C., H.M.P., B.K., P.V., E.A.V.J.)
| | - Bahar Kasaai
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium (V.C., H.M.P., B.K., P.V., E.A.V.J.)
| | - Geertje Swennen
- Department of Physiology, CARIM, Maastricht University, The Netherlands (G.S., M.J.P.)
| | - Emma Gordon
- Department of Immunology, Genetics, and Pathology, Uppsala University, Rudbeck Laboratory, Sweden (E.G., L.C.-W.)
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics, and Pathology, Uppsala University, Rudbeck Laboratory, Sweden (E.G., L.C.-W.)
| | - Mark J. Post
- Department of Physiology, CARIM, Maastricht University, The Netherlands (G.S., M.J.P.)
| | - Peter Verhamme
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium (V.C., H.M.P., B.K., P.V., E.A.V.J.)
| | - Elizabeth A.V. Jones
- From the Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Belgium (V.C., H.M.P., B.K., P.V., E.A.V.J.)
| |
Collapse
|
32
|
Xie A, Zhou A, Liu H, Shi G, Liu M, Boheler KR, Dudley SC. Mitochondrial Ca2+ flux modulates spontaneous electrical activity in ventricular cardiomyocytes. PLoS One 2018; 13:e0200448. [PMID: 30001390 PMCID: PMC6042741 DOI: 10.1371/journal.pone.0200448] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/26/2018] [Indexed: 11/23/2022] Open
Abstract
Introduction Ca2+ release from sarcoplasmic reticulum (SR) is known to contribute to automaticity via the cytoplasmic Na+-Ca2+ exchanger (NCX). Mitochondria participate in Ca2+ cycling. We studied the role of mitochondrial Ca2+ flux in ventricular spontaneous electrical activity. Methods Spontaneously contracting mouse embryonic stem cells (ESC)-derived ventricular cardiomyocytes (CMs) were differentiated from wild type and ryanodine receptor type 2 (RYR2) knockout mouse ESCs and differentiated for 19–21 days. Automaticity was also observed in human induced pluripotent stem cell (hiPSC)-derived ventricular CMs differentiated for 30 days, and acute isolated adult mouse ventricular cells in ischemic simulated buffer. Action potentials (APs) were recorded by perforated whole cell current-clamp. Cytoplasmic and mitochondrial Ca2+ transients were determined by fluorescent imaging. Results In mouse ESC-derived ventricular CMs, spontaneous beating was dependent on the L-type Ca2+ channel, cytoplasmic NCX and mitochondrial NCX. Spontaneous beating was modulated by SR Ca2+ release from RYR2 or inositol trisphosphate receptors (IP3R), the pacemaker current (If) and mitochondrial Ca2+ uptake by the mitochondrial Ca2+ uniporter (MCU). In RYR2 knockout mouse ESC-derived ventricular CMs, mitochondrial Ca2+ flux influenced spontaneous beating independently of the SR Ca2+ release from RYR2, and the mitochondrial effect was dependent on IP3R SR Ca2+ release. Depolarization of mitochondria and preservation of ATP could terminate spontaneous beating. A contribution of mitochondrial Ca2+ flux to automaticity was confirmed in hiPSC-derived ventricular CMs and ischemic adult mouse ventricular CMs, confirming the findings across species and cell maturity levels. Conclusions Mitochondrial and sarcolemma NCX fluxes are required for ventricular automaticity. Mitochondrial Ca2+ uptake plays a modulatory role. Mitochondrial Ca2+ uptake through MCU is influenced by IP3R-dependent SR Ca2+ release.
Collapse
Affiliation(s)
- An Xie
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Anyu Zhou
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Hong Liu
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Guangbin Shi
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Man Liu
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
| | - Kenneth R. Boheler
- Stem Cell and Regenerative Medicine Consortium, LKS Faculty of Medicine, Hong Kong University, Hong Kong, P.R. China
| | - Samuel C. Dudley
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, United States of America
- * E-mail:
| |
Collapse
|
33
|
Freyer L, Hsu CW, Nowotschin S, Pauli A, Ishida J, Kuba K, Fukamizu A, Schier AF, Hoodless PA, Dickinson ME, Hadjantonakis AK. Loss of Apela Peptide in Mice Causes Low Penetrance Embryonic Lethality and Defects in Early Mesodermal Derivatives. Cell Rep 2018; 20:2116-2130. [PMID: 28854362 DOI: 10.1016/j.celrep.2017.08.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/22/2017] [Accepted: 08/01/2017] [Indexed: 01/22/2023] Open
Abstract
Apela (also known as Elabela, Ende, and Toddler) is a small signaling peptide that activates the G-protein-coupled receptor Aplnr to stimulate cell migration during zebrafish gastrulation. Here, using CRISPR/Cas9 to generate a null, reporter-expressing allele, we study the role of Apela in the developing mouse embryo. We found that loss of Apela results in low-penetrance cardiovascular defects that manifest after the onset of circulation. Three-dimensional micro-computed tomography revealed a higher penetrance of vascular remodeling defects, from which some mutants recover, and identified extraembryonic anomalies as the earliest morphological distinction in Apela mutant embryos. Transcriptomics at late gastrulation identified aberrant upregulation of erythroid and myeloid markers in mutant embryos prior to the appearance of physical malformations. Double-mutant analyses showed that loss of Apela signaling impacts early Aplnr-expressing mesodermal populations independently of the alternative ligand Apelin, leading to lethal cardiac defects in some Apela null embryos.
Collapse
Affiliation(s)
- Laina Freyer
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Chih-Wei Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sonja Nowotschin
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Andrea Pauli
- The Research Institute of Molecular Pathology, Vienna BioCenter, 1030 Vienna, Austria
| | - Junji Ishida
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Keiji Kuba
- Department of Biochemistry and Metabolic Science, Akita University, Akita 010-8543, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8577, Japan
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Pamela A Hoodless
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Mary E Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
34
|
Lee LK, Ghorbanian Y, Wang W, Wang Y, Kim YJ, Weissman IL, Inlay MA, Mikkola HKA. LYVE1 Marks the Divergence of Yolk Sac Definitive Hemogenic Endothelium from the Primitive Erythroid Lineage. Cell Rep 2017; 17:2286-2298. [PMID: 27880904 PMCID: PMC6940422 DOI: 10.1016/j.celrep.2016.10.080] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 09/01/2016] [Accepted: 10/21/2016] [Indexed: 01/08/2023] Open
Abstract
The contribution of the different waves and sites of developmental hematopoiesis to fetal and adult blood production remains unclear. Here, we identify lymphatic vessel endothelial hyaluronan receptor-1 (LYVE1) as a marker of yolk sac (YS) endothelium and definitive hematopoietic stem and progenitor cells (HSPCs). Endothelium in mid-gestation YS and vitelline vessels, but not the dorsal aorta and placenta, were labeled by Lyve1-Cre. Most YS HSPCs and erythro-myeloid progenitors were Lyve1-Cre lineage traced, but primitive erythroid cells were not, suggesting that they represent distinct lineages. Fetal liver (FL) and adult HSPCs showed 35%-40% Lyve1-Cre marking. Analysis of circulation-deficient Ncx1-/- concepti identified the YS as a major source of Lyve1-Cre labeled HSPCs. FL proerythroblast marking was extensive at embryonic day (E) 11.5-13.5, but decreased to hematopoietic stem cell (HSC) levels by E16.5, suggesting that HSCs from multiple sources became responsible for erythropoiesis. Lyve1-Cre thus marks the divergence between YS primitive and definitive hematopoiesis and provides a tool for targeting YS definitive hematopoiesis and FL colonization.
Collapse
Affiliation(s)
- Lydia K Lee
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA 90095, USA; Department of Obstetrics and Gynecology, UCLA, Los Angeles, CA 90095, USA
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, Department of Molecular Biology & Biochemistry at UCI, Irvine, CA 92697, USA
| | - Wenyuan Wang
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Yanling Wang
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Yeon Joo Kim
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA 90095, USA
| | - Irving L Weissman
- Institute of Stem Cell Biology and Regenerative Medicine and Ludwig Center, Stanford University, Stanford, CA 94305, USA
| | - Matthew A Inlay
- Sue and Bill Gross Stem Cell Research Center, Department of Molecular Biology & Biochemistry at UCI, Irvine, CA 92697, USA
| | - Hanna K A Mikkola
- Department of Molecular, Cell & Developmental Biology, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
35
|
Lloyd AF, Davies CL, Miron VE. Microglia: origins, homeostasis, and roles in myelin repair. Curr Opin Neurobiol 2017; 47:113-120. [PMID: 29073528 DOI: 10.1016/j.conb.2017.10.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/26/2017] [Accepted: 10/07/2017] [Indexed: 11/27/2022]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS), implicated in developmental processes, homeostasis, and responses to injury. Derived from the yolk sac during development, microglia self-renew, self-regulate their numbers during homeostatic conditions, and show a robust proliferative capacity even in adulthood. Together with monocyte-derived macrophages (MDM), microglia coordinate the regeneration of CNS myelin around axons, termed remyelination. Gene expression analyses and experimental modelling have identified pro-remyelination roles for microglia/MDM in clearance of myelin debris, secretion of growth factors, and remodelling of the extracellular matrix. Further investigations into the molecular mechanisms controlling these regenerative functions will reveal novel therapeutic strategies to enhance remyelination, by harnessing the beneficial effects of the innate immune response to injury.
Collapse
Affiliation(s)
- Amy F Lloyd
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Claire L Davies
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
36
|
Hwa JJ, Beckouche N, Huang L, Kram Y, Lindskog H, Wang RA. Abnormal arterial-venous fusions and fate specification in mouse embryos lacking blood flow. Sci Rep 2017; 7:11965. [PMID: 28931948 PMCID: PMC5607254 DOI: 10.1038/s41598-017-12353-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
The functions of blood flow in the morphogenesis of mammalian arteries and veins are not well understood. We examined the development of the dorsal aorta (DA) and the cardinal vein (CV) in Ncx1 -/- mutants, which lack blood flow due to a deficiency in a sodium calcium ion exchanger expressed specifically in the heart. The mutant DA and CV were abnormally connected. The endothelium of the Ncx1 -/- mutant DA lacked normal expression of the arterial markers ephrin-B2 and Connexin-40. Notch1 activation, known to promote arterial specification, was decreased in mutant DA endothelial cells (ECs), which ectopically expressed the venous marker Coup-TFII. These findings suggest that flow has essential functions in the DA by promoting arterial and suppressing venous marker expression. In contrast, flow plays a lesser role in the CV, because expression of arterial-venous markers in CV ECs was not as dramatically affected in Ncx1 -/- mutants. We propose a molecular mechanism by which blood flow mediates DA and CV morphogenesis, by regulating arterial-venous specification of DA ECs to ensure proper separation of the developing DA and CV.
Collapse
Affiliation(s)
- Jennifer J Hwa
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Nathan Beckouche
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Lawrence Huang
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Yoseph Kram
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Henrik Lindskog
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Rong A Wang
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
37
|
Shimizu H, Langenbacher AD, Huang J, Wang K, Otto G, Geisler R, Wang Y, Chen JN. The Calcineurin-FoxO-MuRF1 signaling pathway regulates myofibril integrity in cardiomyocytes. eLife 2017; 6:27955. [PMID: 28826496 PMCID: PMC5576919 DOI: 10.7554/elife.27955] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Altered Ca2+ handling is often present in diseased hearts undergoing structural remodeling and functional deterioration. However, whether Ca2+ directly regulates sarcomere structure has remained elusive. Using a zebrafish ncx1 mutant, we explored the impacts of impaired Ca2+ homeostasis on myofibril integrity. We found that the E3 ubiquitin ligase murf1 is upregulated in ncx1-deficient hearts. Intriguingly, knocking down murf1 activity or inhibiting proteasome activity preserved myofibril integrity, revealing a MuRF1-mediated proteasome degradation mechanism that is activated in response to abnormal Ca2+ homeostasis. Furthermore, we detected an accumulation of the murf1 regulator FoxO in the nuclei of ncx1-deficient cardiomyocytes. Overexpression of FoxO in wild type cardiomyocytes induced murf1 expression and caused myofibril disarray, whereas inhibiting Calcineurin activity attenuated FoxO-mediated murf1 expression and protected sarcomeres from degradation in ncx1-deficient hearts. Together, our findings reveal a novel mechanism by which Ca2+ overload disrupts myofibril integrity by activating a Calcineurin-FoxO-MuRF1-proteosome signaling pathway.
Collapse
Affiliation(s)
- Hirohito Shimizu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Adam D Langenbacher
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Jie Huang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Kevin Wang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Georg Otto
- Genetics and Genomic Medicine, UCL Institute of Child Health, London, United Kingdom
| | - Robert Geisler
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Yibin Wang
- Department of Anesthesiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Department of Medicine and Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Jau-Nian Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
38
|
Ratajczak MZ. Why are hematopoietic stem cells so 'sexy'? on a search for developmental explanation. Leukemia 2017; 31:1671-1677. [PMID: 28502982 PMCID: PMC5540746 DOI: 10.1038/leu.2017.148] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/03/2017] [Accepted: 03/07/2017] [Indexed: 02/07/2023]
Abstract
Evidence has accumulated that normal human and murine hematopoietic stem cells express several functional pituitary and gonadal sex hormones, and that, in fact, some sex hormones, such as androgens, have been employed for many years to stimulate hematopoiesis in patients with bone marrow aplasia. Interestingly, sex hormone receptors are also expressed by leukemic cell lines and blasts. In this review, I will discuss the emerging question of why hematopoietic cells express these receptors. A tempting hypothetical explanation for this phenomenon is that hematopoietic stem cells are related to subpopulation of migrating primordial germ cells. To support of this notion, the anatomical sites of origin of primitive and definitive hematopoiesis during embryonic development are tightly connected with the migratory route of primordial germ cells: from the proximal epiblast to the extraembryonic endoderm at the bottom of the yolk sac and then back to the embryo proper via the primitive streak to the aorta-gonado-mesonephros (AGM) region on the way to the genital ridges. The migration of these cells overlaps with the emergence of primitive hematopoiesis in the blood islands at the bottom of the yolk sac, and definitive hematopoiesis that occurs in hemogenic endothelium in the embryonic dorsal aorta in AGM region.
Collapse
Affiliation(s)
- M Z Ratajczak
- Stem Cell Institute, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
39
|
Yamamoto S, Muramatsu M, Azuma E, Ikutani M, Nagai Y, Sagara H, Koo BN, Kita S, O'Donnell E, Osawa T, Takahashi H, Takano KI, Dohmoto M, Sugimori M, Usui I, Watanabe Y, Hatakeyama N, Iwamoto T, Komuro I, Takatsu K, Tobe K, Niida S, Matsuda N, Shibuya M, Sasahara M. A subset of cerebrovascular pericytes originates from mature macrophages in the very early phase of vascular development in CNS. Sci Rep 2017. [PMID: 28634350 PMCID: PMC5478595 DOI: 10.1038/s41598-017-03994-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Pericytes are believed to originate from either mesenchymal or neural crest cells. It has recently been reported that pericytes play important roles in the central nervous system (CNS) by regulating blood-brain barrier homeostasis and blood flow at the capillary level. However, the origin of CNS microvascular pericytes and the mechanism of their recruitment remain unknown. Here, we show a new source of cerebrovascular pericytes during neurogenesis. In the CNS of embryonic day 10.5 mouse embryos, CD31+F4/80+ hematopoietic lineage cells were observed in the avascular region around the dorsal midline of the developing midbrain. These cells expressed additional macrophage markers such as CD206 and CD11b. Moreover, the CD31+F4/80+ cells phagocytosed apoptotic cells as functionally matured macrophages, adhered to the newly formed subventricular vascular plexus, and then divided into daughter cells. Eventually, these CD31+F4/80+ cells transdifferentiated into NG2/PDGFRβ/desmin-expressing cerebrovascular pericytes, enwrapping and associating with vascular endothelial cells. These data indicate that a subset of cerebrovascular pericytes derive from mature macrophages in the very early phase of CNS vascular development, which in turn are recruited from sites of embryonic hematopoiesis such as the yolk sac by way of blood flow.
Collapse
Affiliation(s)
- Seiji Yamamoto
- Department of Pathology, University of Toyama, Toyama, Japan.
| | - Masashi Muramatsu
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Erika Azuma
- Department of Pathology, University of Toyama, Toyama, Japan.,Department of Technology Development, Astellas Pharma Tech Co., Ltd., Toyama, Japan
| | - Masashi Ikutani
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan
| | - Yoshinori Nagai
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan.,JST, PRESTO, Kawaguchi, Saitama, Japan
| | - Hiroshi Sagara
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Bon-Nyeo Koo
- Department of Anesthesiology, Yonsei University College of Medicine, Seoul, Korea.,Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Maryland, USA
| | - Satomi Kita
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Erin O'Donnell
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Maryland, USA
| | - Tsuyoshi Osawa
- Laboratry for Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takahashi
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | - Ken-Ichi Takano
- Departments of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Mitsuko Dohmoto
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Ishikawa, Japan
| | - Michiya Sugimori
- Department of Integrative Neuroscience, University of Toyama, Toyama, Japan
| | - Isao Usui
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Yasuhide Watanabe
- Faculty of Medicine, School of Nursing, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Noboru Hatakeyama
- Department of Anesthesiology, Graduate School of Medicine, Aichi Medical University, Aichi, Japan
| | - Takahiro Iwamoto
- Department of Pharmacology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kiyoshi Takatsu
- Department of Immunobiology and Pharmacological Genetics, University of Toyama, Toyama, Japan.,Toyama Prefectural Institute for Pharmaceutical Research, Toyama, Japan
| | - Kazuyuki Tobe
- First Department of Internal Medicine, University of Toyama, Toyama, Japan
| | - Shumpei Niida
- Medical Genome Center, Center for Geriatrics and Gerontology, Aichi, Japan
| | - Naoyuki Matsuda
- Department of Emergency and Critical Care Medicine, Nagoya University, Nagoya, Japan
| | - Masabumi Shibuya
- Department of Research and Education, Jobu University, Gunma, Japan
| | | |
Collapse
|
40
|
Beggs MR, Alexander RT. Intestinal absorption and renal reabsorption of calcium throughout postnatal development. Exp Biol Med (Maywood) 2017; 242:840-849. [PMID: 28346014 DOI: 10.1177/1535370217699536] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Calcium is vital for many physiological functions including bone mineralization. Postnatal deposition of calcium into bone is greatest in infancy and continues through childhood and adolescence until peek mineral density is reached in early adulthood. Thereafter, bone mineral density remains static until it eventually declines in later life. A positive calcium balance, i.e. more calcium absorbed than excreted, is crucial to bone deposition during growth and thus to peek bone mineral density. Dietary calcium is absorbed from the intestine into the blood. It is then filtered by the renal glomerulus and either reabsorbed by the tubule or excreted in the urine. Calcium can be (re)absorbed across intestinal and renal epithelia via both transcellular and paracellular pathways. Current evidence suggests that significant intestinal and renal calcium transport changes occur throughout development. However, the molecular details of these alterations are incompletely delineated. Here we first briefly review the current model of calcium transport in the intestine and renal tubule in the adult. Then, we describe what is known with regard to calcium handling through postnatal development, and how alterations may aid in mediating a positive calcium balance. The role of transcellular and paracellular calcium transport pathways and the contribution of specific intestinal and tubular segments vary with age. However, the current literature highlights knowledge gaps in how specifically intestinal and renal calcium (re)absorption occurs early in postnatal development. Future research should clarify the specific changes in calcium transport throughout early postnatal development including mediators of these alterations enabling appropriate bone mineralization. Impact statement This mini review outlines the current state of knowledge pertaining to the molecules and mechanisms maintaining a positive calcium balance throughout postnatal development. This process is essential to achieving optimal bone mineral density in early adulthood, thereby lowering the lifetime risk of osteoporosis.
Collapse
Affiliation(s)
- Megan R Beggs
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - R Todd Alexander
- 1 Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2R7, Canada.,2 Department of Pediatrics, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
41
|
Reemst K, Noctor SC, Lucassen PJ, Hol EM. The Indispensable Roles of Microglia and Astrocytes during Brain Development. Front Hum Neurosci 2016; 10:566. [PMID: 27877121 PMCID: PMC5099170 DOI: 10.3389/fnhum.2016.00566] [Citation(s) in RCA: 351] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/25/2016] [Indexed: 01/17/2023] Open
Abstract
Glia are essential for brain functioning during development and in the adult brain. Here, we discuss the various roles of both microglia and astrocytes, and their interactions during brain development. Although both cells are fundamentally different in origin and function, they often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis and synaptic pruning. Due to their important instructive roles in these processes, dysfunction of microglia or astrocytes during brain development could contribute to neurodevelopmental disorders and potentially even late-onset neuropathology. A better understanding of the origin, differentiation process and developmental functions of microglia and astrocytes will help to fully appreciate their role both in the developing as well as in the adult brain, in health and disease.
Collapse
Affiliation(s)
- Kitty Reemst
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Stephen C. Noctor
- Department of Psychiatry and Behavioral Sciences, UC Davis MIND InstituteSacramento, CA, USA
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
| | - Elly M. Hol
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdam, Netherlands
- Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
- Netherlands Institute for NeuroscienceAmsterdam, Netherlands
| |
Collapse
|
42
|
Tyser RC, Miranda AM, Chen CM, Davidson SM, Srinivas S, Riley PR. Calcium handling precedes cardiac differentiation to initiate the first heartbeat. eLife 2016; 5. [PMID: 27725084 PMCID: PMC5059139 DOI: 10.7554/elife.17113] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/13/2016] [Indexed: 11/30/2022] Open
Abstract
The mammalian heartbeat is thought to begin just prior to the linear heart tube stage of development. How the initial contractions are established and the downstream consequences of the earliest contractile function on cardiac differentiation and morphogenesis have not been described. Using high-resolution live imaging of mouse embryos, we observed randomly distributed spontaneous asynchronous Ca2+-oscillations (SACOs) in the forming cardiac crescent (stage E7.75) prior to overt beating. Nascent contraction initiated at around E8.0 and was associated with sarcomeric assembly and rapid Ca2+ transients, underpinned by sequential expression of the Na+-Ca2+ exchanger (NCX1) and L-type Ca2+ channel (LTCC). Pharmacological inhibition of NCX1 and LTCC revealed rapid development of Ca2+ handling in the early heart and an essential early role for NCX1 in establishing SACOs through to the initiation of beating. NCX1 blockade impacted on CaMKII signalling to down-regulate cardiac gene expression, leading to impaired differentiation and failed crescent maturation. DOI:http://dx.doi.org/10.7554/eLife.17113.001 The heart is the first organ to form and to begin working in an embryo during pregnancy. It must begin pumping early to supply oxygen and nutrients to the developing embryo. Coordinated contractions of specialised muscle cells in the heart, called cardiomyocytes, generate the force needed to pump blood. The flow of calcium ions into and out of the cardiomyocytes triggers these heartbeats. In addition to triggering heart contractions, calcium ions also act as a messenger that drives changes in which genes are active in the cardiomyocytes and how these cells behave. Scientists commonly think of the first heartbeat as occurring after a tube-like structure forms in the embryo that will eventually develop into the heart. However, it is not yet clear how the first heartbeat starts or how the initial heartbeats affect further heart development. Tyser, Miranda et al. now show that the first heartbeat actually occurs much earlier in embryonic development than widely appreciated. In the experiments, videos of live mouse embryos showed that prior to the first heartbeat the flow of calcium ions between different cardiomyocytes is not synchronised. However, as the heart grows these calcium flows become coordinated leading to the first heartbeat. The heartbeats also become faster as the heart grows. Using drugs to block the movement of calcium ions, Tyser, Miranda et al. also show that a protein called NCX1 is required to trigger the calcium flows prior to the first heartbeat. Moreover, the experiments revealed that these early heartbeats help drive the growth of cardiomyocytes and shape the developing heart. Together, the experiments show that the first heartbeats are essential for normal heart development. Future studies are needed to determine what controls the speed of the first heartbeats, and what organises the calcium flows that trigger the first heartbeat. Such studies may help scientists better understand birth defects of the heart, and may suggest strategies to rebuild hearts that have been damaged by a heart attack or other injury. DOI:http://dx.doi.org/10.7554/eLife.17113.002
Collapse
Affiliation(s)
- Richard Cv Tyser
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.,The Hatter Cardiovascular Institute, University College London and Medical School, London, United Kingdom
| | - Antonio Ma Miranda
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London and Medical School, London, United Kingdom
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
43
|
Ectopic expression of Cripto-1 in transgenic mouse embryos causes hemorrhages, fatal cardiac defects and embryonic lethality. Sci Rep 2016; 6:34501. [PMID: 27687577 PMCID: PMC5043281 DOI: 10.1038/srep34501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 09/15/2016] [Indexed: 12/23/2022] Open
Abstract
Targeted disruption of Cripto-1 in mice caused embryonic lethality at E7.5, whereas we unexpectedly found that ectopic Cripto-1 expression in mouse embryos also led to embryonic lethality, which prompted us to characterize the causes and mechanisms underlying embryonic death due to ectopic Cripto-1 expression. RCLG/EIIa-Cre embryos displayed complex phenotypes between embryonic day 14.5 (E14.5) and E17.5, including fatal hemorrhages (E14.5-E15.5), embryo resorption (E14.5-E17.5), pale body surface (E14.5-E16.5) and no abnormal appearance (E14.5-E16.5). Macroscopic and histological examination revealed that ectopic expression of Cripto-1 transgene in RCLG/EIIa-Cre embryos resulted in lethal cardiac defects, as evidenced by cardiac malformations, myocardial thinning, failed assembly of striated myofibrils and lack of heartbeat. In addition, Cripto-1 transgene activation beginning after E8.5 also caused the aforementioned lethal cardiac defects in mouse embryos. Furthermore, ectopic Cripto-1 expression in embryonic hearts reduced the expression of cardiac transcription factors, which is at least partially responsible for the aforementioned lethal cardiac defects. Our results suggest that hemorrhages and cardiac abnormalities are two important lethal factors in Cripto-1 transgenic mice. Taken together, these findings are the first to demonstrate that sustained Cripto-1 transgene expression after E11.5 causes fatal hemorrhages and lethal cardiac defects, leading to embryonic death at E14.5-17.5.
Collapse
|
44
|
Höfer T, Busch K, Klapproth K, Rodewald HR. Fate Mapping and Quantitation of Hematopoiesis In Vivo. Annu Rev Immunol 2016; 34:449-78. [DOI: 10.1146/annurev-immunol-032414-112019] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Kay Klapproth
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| |
Collapse
|
45
|
Watanabe M, Rollins AM, Polo-Parada L, Ma P, Gu S, Jenkins MW. Probing the Electrophysiology of the Developing Heart. J Cardiovasc Dev Dis 2016; 3:jcdd3010010. [PMID: 29367561 PMCID: PMC5715694 DOI: 10.3390/jcdd3010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/08/2016] [Accepted: 03/10/2016] [Indexed: 12/14/2022] Open
Abstract
Many diseases that result in dysfunction and dysmorphology of the heart originate in the embryo. However, the embryonic heart presents a challenging subject for study: especially challenging is its electrophysiology. Electrophysiological maturation of the embryonic heart without disturbing its physiological function requires the creation and deployment of novel technologies along with the use of classical techniques on a range of animal models. Each tool has its strengths and limitations and has contributed to making key discoveries to expand our understanding of cardiac development. Further progress in understanding the mechanisms that regulate the normal and abnormal development of the electrophysiology of the heart requires integration of this functional information with the more extensively elucidated structural and molecular changes.
Collapse
Affiliation(s)
- Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Andrew M Rollins
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Luis Polo-Parada
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65201, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65201, USA.
| | - Pei Ma
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Shi Gu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Michael W Jenkins
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
- Rainbow Babies and Children's Hospital, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
46
|
Frame JM, Fegan KH, Conway SJ, McGrath KE, Palis J. Definitive Hematopoiesis in the Yolk Sac Emerges from Wnt-Responsive Hemogenic Endothelium Independently of Circulation and Arterial Identity. Stem Cells 2016; 34:431-44. [PMID: 26418893 PMCID: PMC4755868 DOI: 10.1002/stem.2213] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/21/2015] [Accepted: 09/04/2015] [Indexed: 12/20/2022]
Abstract
Adult-repopulating hematopoietic stem cells (HSCs) emerge in low numbers in the midgestation mouse embryo from a subset of arterial endothelium, through an endothelial-to-hematopoietic transition. HSC-producing arterial hemogenic endothelium relies on the establishment of embryonic blood flow and arterial identity, and requires β-catenin signaling. Specified prior to and during the formation of these initial HSCs are thousands of yolk sac-derived erythro-myeloid progenitors (EMPs). EMPs ensure embryonic survival prior to the establishment of a permanent hematopoietic system, and provide subsets of long-lived tissue macrophages. While an endothelial origin for these HSC-independent definitive progenitors is also accepted, the spatial location and temporal output of yolk sac hemogenic endothelium over developmental time remain undefined. We performed a spatiotemporal analysis of EMP emergence, and document the morphological steps of the endothelial-to-hematopoietic transition. Emergence of rounded EMPs from polygonal clusters of Kit(+) cells initiates prior to the establishment of arborized arterial and venous vasculature in the yolk sac. Interestingly, Kit(+) polygonal clusters are detected in both arterial and venous vessels after remodeling. To determine whether there are similar mechanisms regulating the specification of EMPs with other angiogenic signals regulating adult-repopulating HSCs, we investigated the role of embryonic blood flow and Wnt/β-catenin signaling during EMP emergence. In embryos lacking a functional circulation, rounded Kit(+) EMPs still fully emerge from unremodeled yolk sac vasculature. In contrast, canonical Wnt signaling appears to be a common mechanism regulating hematopoietic emergence from hemogenic endothelium. These data illustrate the heterogeneity in hematopoietic output and spatiotemporal regulation of primary embryonic hemogenic endothelium.
Collapse
Affiliation(s)
- Jenna M. Frame
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Katherine H. Fegan
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Simon J. Conway
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathleen E. McGrath
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY, USA
| | - James Palis
- Department of Pediatrics, Center for Pediatric Biomedical Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
47
|
Chabab S, Lescroart F, Rulands S, Mathiah N, Simons BD, Blanpain C. Uncovering the Number and Clonal Dynamics of Mesp1 Progenitors during Heart Morphogenesis. Cell Rep 2016; 14:1-10. [PMID: 26725109 PMCID: PMC4709258 DOI: 10.1016/j.celrep.2015.12.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/07/2015] [Accepted: 11/23/2015] [Indexed: 12/22/2022] Open
Abstract
The heart arises from distinct sources of cardiac progenitors that independently express Mesp1 during gastrulation. The precise number of Mesp1 progenitors that are specified during the early stage of gastrulation, and their clonal behavior during heart morphogenesis, is currently unknown. Here, we used clonal and mosaic tracing of Mesp1-expressing cells combined with quantitative biophysical analysis of the clonal data to define the number of cardiac progenitors and their mode of growth during heart development. Our data indicate that the myocardial layer of the heart derive from ∼250 Mesp1-expressing cardiac progenitors born during gastrulation. Despite arising at different time points and contributing to different heart regions, the temporally distinct cardiac progenitors present very similar clonal dynamics. These results provide insights into the number of cardiac progenitors and their mode of growth and open up avenues to decipher the clonal dynamics of progenitors in other organs and tissues.
Collapse
Affiliation(s)
- Samira Chabab
- Université Libre de Bruxelles, IRIBHM, Brussels 1070, Belgium
| | | | - Steffen Rulands
- Cavendish Laboratory, Department of Physics, University of Cambridge, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Navrita Mathiah
- Université Libre de Bruxelles, IRIBHM, Brussels 1070, Belgium
| | - Benjamin D Simons
- Cavendish Laboratory, Department of Physics, University of Cambridge, J. J. Thomson Avenue, Cambridge CB3 0HE, UK; The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK.
| | - Cédric Blanpain
- Université Libre de Bruxelles, IRIBHM, Brussels 1070, Belgium; WELBIO, Université Libre de Bruxelles, Brussels 1070, Belgium.
| |
Collapse
|
48
|
Karppinen S, Rapila R, Naumenko N, Tuomainen T, Koivumäki JT, Hänninen SL, Korhonen T, Tavi P. Ca(2+) -activated K(+) current is essential for maintaining excitability and gene transcription in early embryonic cardiomyocytes. Acta Physiol (Oxf) 2016; 216:101-11. [PMID: 26095188 DOI: 10.1111/apha.12540] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 11/11/2014] [Accepted: 06/03/2015] [Indexed: 01/06/2023]
Abstract
AIM Activity of early embryonic cardiomyocytes relies on spontaneous Ca(2+) oscillations that are induced by interplay between sarcoplasmic reticulum (SR) - Ca(2+) release and ion currents of the plasma membrane. In a variety of cell types, Ca(2+) -activated K(+) current (IK(Ca) ) serves as a link between Ca(2+) signals and membrane voltage. This study aimed to determine the role of IK (Ca) in developing cardiomyocytes. METHODS Ion currents and membrane voltage of embryonic (E9-11) mouse cardiomyocytes were measured by patch clamp; [Ca(2+) ]i signals by confocal microscopy. Transcription of specific genes was measured with RT-qPCR and Ca(2+) -dependent transcriptional activity using NFAT-luciferase assay. Myocyte structure was assessed with antibody labelling and confocal microscopy. RESULTS E9-11 cardiomyocytes express small conductance (SK) channel subunits SK2 and SK3 and have a functional apamin-sensitive K(+) current, which is also sensitive to changes in cytosolic [Ca(2+) ]i . In spontaneously active cardiomyocytes, inhibition of IK (Ca) changed action and resting potentials, reduced SR Ca(2+) load and suppressed the amplitude and the frequency of spontaneously evoked Ca(2+) oscillations. Apamin caused dose-dependent suppression of NFAT-luciferase reporter activity, induced downregulation of a pattern of genes vital for cardiomyocyte development and triggered changes in the myocyte morphology. CONCLUSION The results show that apamin-sensitive IK (Ca) is required for maintaining excitability and activity of the developing cardiomyocytes as well as having a fundamental role in promoting Ca(2+) - dependent gene expression.
Collapse
Affiliation(s)
- S. Karppinen
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - R. Rapila
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - N. Naumenko
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - T. Tuomainen
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - J. T. Koivumäki
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - S. L. Hänninen
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
- Institute of Biomedicine; Department of Physiology and Biocenter Oulu; University of Oulu; Oulu Finland
| | - T. Korhonen
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| | - P. Tavi
- Department of Biotechnology and Molecular Medicine; A.I. Virtanen Institute for Molecular Sciences; University of Eastern Finland; Kuopio Finland
| |
Collapse
|
49
|
Weisbrod D, Khun SH, Bueno H, Peretz A, Attali B. Mechanisms underlying the cardiac pacemaker: the role of SK4 calcium-activated potassium channels. Acta Pharmacol Sin 2016; 37:82-97. [PMID: 26725737 PMCID: PMC4722971 DOI: 10.1038/aps.2015.135] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/25/2015] [Indexed: 12/25/2022] Open
Abstract
The proper expression and function of the cardiac pacemaker is a critical feature of heart physiology. The sinoatrial node (SAN) in human right atrium generates an electrical stimulation approximately 70 times per minute, which propagates from a conductive network to the myocardium leading to chamber contractions during the systoles. Although the SAN and other nodal conductive structures were identified more than a century ago, the mechanisms involved in the generation of cardiac automaticity remain highly debated. In this short review, we survey the current data related to the development of the human cardiac conduction system and the various mechanisms that have been proposed to underlie the pacemaker activity. We also present the human embryonic stem cell-derived cardiomyocyte system, which is used as a model for studying the pacemaker. Finally, we describe our latest characterization of the previously unrecognized role of the SK4 Ca(2+)-activated K(+) channel conductance in pacemaker cells. By exquisitely balancing the inward currents during the diastolic depolarization, the SK4 channels appear to play a crucial role in human cardiac automaticity.
Collapse
Affiliation(s)
- David Weisbrod
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shiraz Haron Khun
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hanna Bueno
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Asher Peretz
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Bernard Attali
- Department of Physiology & Pharmacology, Sackler Faculty of Medicine, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
50
|
Lewis AE, Hwa J, Wang R, Soriano P, Bush JO. Neural crest defects in ephrin-B2 mutant mice are non-autonomous and originate from defects in the vasculature. Dev Biol 2015; 406:186-95. [PMID: 26385750 DOI: 10.1016/j.ydbio.2015.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/28/2015] [Accepted: 08/29/2015] [Indexed: 10/23/2022]
Abstract
Ephrin-B2, a member of the Eph/ephrin family of cell signaling molecules, has been implicated in the guidance of cranial and trunk neural crest cells (NCC) and development of the branchial arches(BA), but detailed examination in mice has been hindered by embryonic lethality of Efnb2 null loss of function due to a requirement in angiogenic remodeling. To elucidate the developmental roles for Efnb2, we generated a conditional rescue knock-in allele that allows rescue of ephrin-B2 specifically in the vascular endothelium (VE), but is otherwise ephrin-B2 deficient. Restoration of ephrin-B2 expression specifically to the VE completely circumvents angiogenic phenotypes, indicating that the requirement of ephrin-B2 in angiogenesis is limited to the VE. Surprisingly, we find that expression of ephrin-B2 specifically in the VE is also sufficient for normal NCC migration and that conversely, embryos in which ephrin-B2 is absent specifically from the VE exhibit NCC migration and survival defects. Disruption of vascular development independent of loss of ephrin-B2 function also leads to defects in NCC and BA development. Together, these data indicate that direct ephrin-B2 signaling to NCCs is not required for NCC guidance, which instead depends on proper organization of the embryonic vasculature.
Collapse
Affiliation(s)
- Ace E Lewis
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, CA 94143, United States
| | - Jennifer Hwa
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, CA 94143, United States
| | - Rong Wang
- Laboratory for Accelerated Vascular Research, Division of Vascular Surgery, Department of Surgery, University of California, San Francisco, CA 94143, United States
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Mt. Sinai School of Medicine, New York, NY 10029, United States
| | - Jeffrey O Bush
- Department of Cell and Tissue Biology, Program in Craniofacial Biology and Institute for Human Genetics, University of California, San Francisco, CA 94143, United States.
| |
Collapse
|