1
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2024:S1534-5807(24)00545-8. [PMID: 39413783 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
2
|
Miao L, Lu Y, Nusrat A, Zhao L, Castillo M, Xiao Y, Guo H, Liu Y, Gunaratne P, Schwartz RJ, Burns AR, Kumar A, DiPersio CM, Wu M. β1 integrins regulate cellular behaviour and cardiomyocyte organization during ventricular wall formation. Cardiovasc Res 2024; 120:1279-1294. [PMID: 38794925 PMCID: PMC11416060 DOI: 10.1093/cvr/cvae111] [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: 08/31/2023] [Revised: 01/30/2024] [Accepted: 03/17/2024] [Indexed: 05/26/2024] Open
Abstract
AIMS The mechanisms regulating the cellular behaviour and cardiomyocyte organization during ventricular wall morphogenesis are poorly understood. Cardiomyocytes are surrounded by extracellular matrix (ECM) and interact with ECM via integrins. This study aims to determine whether and how β1 integrins regulate cardiomyocyte behaviour and organization during ventricular wall morphogenesis in the mouse. METHODS AND RESULTS We applied mRNA deep sequencing and immunostaining to determine the expression repertoires of α/β integrins and their ligands in the embryonic heart. Integrin β1 subunit (β1) and some of its ECM ligands are asymmetrically distributed and enriched in the luminal side of cardiomyocytes, and fibronectin surrounds cardiomyocytes, creating a network for them. Itgb1, which encodes the β1, was deleted via Nkx2.5Cre/+ to generate myocardial-specific Itgb1 knockout (B1KO) mice. B1KO hearts display an absence of a trabecular zone but a thicker compact zone. The levels of hyaluronic acid and versican, essential for trabecular initiation, were not significantly different between control and B1KO. Instead, fibronectin, a ligand of β1, was absent in the myocardium of B1KO hearts. Furthermore, B1KO cardiomyocytes display a random cellular orientation and fail to undergo perpendicular cell division, be organized properly, and establish the proper tissue architecture to form trabeculae. Mosaic clonal lineage tracing showed that Itgb1 regulates cardiomyocyte transmural migration and proliferation autonomously. CONCLUSION β1 is asymmetrically localized in the cardiomyocytes, and some of its ECM ligands are enriched along the luminal side of the myocardium, and fibronectin surrounds cardiomyocytes. β1 integrins are required for cardiomyocytes to attach to the ECM network. This engagement provides structural support for cardiomyocytes to maintain shape, undergo perpendicular division, and establish cellular organization. Deletion of Itgb1 leads to loss of β1 and fibronectin and prevents cardiomyocytes from engaging the ECM network, resulting in failure to establish tissue architecture to form trabeculae.
Collapse
Affiliation(s)
- Lianjie Miao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Yangyang Lu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Anika Nusrat
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Luqi Zhao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Micah Castillo
- Department of Biology and Biochemistry, University of Houston Sequencing and Gene Editing Core, University of Houston, Houston, TX 77204-5001, USA
| | - Yongqi Xiao
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Hongyang Guo
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Yu Liu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston Sequencing and Gene Editing Core, University of Houston, Houston, TX 77204-5001, USA
| | - Robert J Schwartz
- Department of Biology and Biochemistry, University of Houston Sequencing and Gene Editing Core, University of Houston, Houston, TX 77204-5001, USA
| | - Alan R Burns
- College of Optometry, University of Houston, Houston, TX 77204-2020, USA
| | - Ashok Kumar
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| | | | - Mingfu Wu
- Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204-5039, USA
| |
Collapse
|
3
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Buffering Mechanism in Aortic Arch Artery Formation and Congenital Heart Disease. Circ Res 2024; 134:e112-e132. [PMID: 38618720 PMCID: PMC11081845 DOI: 10.1161/circresaha.123.322767] [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: 03/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.
Collapse
Affiliation(s)
- AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Christina A. Vyzas
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Kevin Eng
- Department of Statistics, Rutgers University, School of Arts and Sciences, Piscataway, NJ 08854
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
4
|
Leite DJ, Schönauer A, Blakeley G, Harper A, Garcia-Castro H, Baudouin-Gonzalez L, Wang R, Sarkis N, Nikola AG, Koka VSP, Kenny NJ, Turetzek N, Pechmann M, Solana J, McGregor AP. An atlas of spider development at single-cell resolution provides new insights into arthropod embryogenesis. EvoDevo 2024; 15:5. [PMID: 38730509 PMCID: PMC11083766 DOI: 10.1186/s13227-024-00224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 04/15/2024] [Indexed: 05/13/2024] Open
Abstract
Spiders are a diverse order of chelicerates that diverged from other arthropods over 500 million years ago. Research on spider embryogenesis, particularly studies using the common house spider Parasteatoda tepidariorum, has made important contributions to understanding the evolution of animal development, including axis formation, segmentation, and patterning. However, we lack knowledge about the cells that build spider embryos, their gene expression profiles and fate. Single-cell transcriptomic analyses have been revolutionary in describing these complex landscapes of cellular genetics in a range of animals. Therefore, we carried out single-cell RNA sequencing of P. tepidariorum embryos at stages 7, 8 and 9, which encompass the establishment and patterning of the body plan, and initial differentiation of many tissues and organs. We identified 20 cell clusters, from 18.5 k cells, which were marked by many developmental toolkit genes, as well as a plethora of genes not previously investigated. We found differences in the cell cycle transcriptional signatures, suggestive of different proliferation dynamics, which related to distinctions between endodermal and some mesodermal clusters, compared with ectodermal clusters. We identified many Hox genes as markers of cell clusters, and Hox gene ohnologs were often present in different clusters. This provided additional evidence of sub- and/or neo-functionalisation of these important developmental genes after the whole genome duplication in an arachnopulmonate ancestor (spiders, scorpions, and related orders). We also examined the spatial expression of marker genes for each cluster to generate a comprehensive cell atlas of these embryonic stages. This revealed new insights into the cellular basis and genetic regulation of head patterning, hematopoiesis, limb development, gut development, and posterior segmentation. This atlas will serve as a platform for future analysis of spider cell specification and fate, and studying the evolution of these processes among animals at cellular resolution.
Collapse
Affiliation(s)
- Daniel J Leite
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK.
| | - Anna Schönauer
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Grace Blakeley
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Amber Harper
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Helena Garcia-Castro
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | | | - Ruixun Wang
- Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674, Cologne, Germany
| | - Naïra Sarkis
- Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674, Cologne, Germany
| | - Alexander Günther Nikola
- Evolutionary Ecology, Faculty of Biology, Biocenter, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Venkata Sai Poojitha Koka
- Evolutionary Ecology, Faculty of Biology, Biocenter, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Nathan J Kenny
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
- Department of Biochemistry Te Tari Matū Koiora, University of Otago, Dunedin, New Zealand
| | - Natascha Turetzek
- Evolutionary Ecology, Faculty of Biology, Biocenter, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Matthias Pechmann
- Institute for Zoology, Biocenter, University of Cologne, Zuelpicher Str. 47B, 50674, Cologne, Germany
| | - Jordi Solana
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
| | - Alistair P McGregor
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK.
- Department of Biosciences, Durham University, Durham, DH1 3LE, UK.
| |
Collapse
|
5
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Identification of novel buffering mechanisms in aortic arch artery development and congenital heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.02.530833. [PMID: 38370627 PMCID: PMC10871175 DOI: 10.1101/2023.03.02.530833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Rationale The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease (CHD) arising from defective morphogenesis of pharyngeal arch arteries (PAA) and their derivatives. Objective To uncover mechanisms underlying the robustness of PAA morphogenesis. Methods and Results The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived ECs is regulated by VEGFR2 and Tbx1 . Remarkably, when SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated three-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of one VEGFR2 allele (VEGFR2 SHF-HET ) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2 SHF-KO ) abolishes it. The decrease in SHF-derived ECs in VEGFR2 SHF-HET and VEGFR2 SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2 SHF-KO mutants. Blocking the compensatory response in VEGFR2 SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1 +/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and CHD. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. Conclusions Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling. Nonstandard Abbreviations and Acronyms in Alphabetical Order CHD - congenital heart disease; ECs - endothelial cells; IAA-B - interrupted aortic arch type B; PAA - pharyngeal arch arteries; RERSA - retro-esophageal right subclavian artery; SHF - second heart field; VEGFR2 - Vascular endothelial growth factor receptor 2.
Collapse
|
6
|
Hajiali H, Rotherham M, El Haj AJ. Remote Activation of Mechanotransduction via Integrin Alpha-5 via Aptamer-Conjugated Magnetic Nanoparticles Promotes Osteogenesis. Pharmaceutics 2023; 16:21. [PMID: 38258032 PMCID: PMC10821094 DOI: 10.3390/pharmaceutics16010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Bone regeneration and repair are complex processes in the adult skeleton, and current research has focused on understanding and controlling these processes. Magnetic nanoparticle (MNP)-based platforms have shown potential in tissue engineering and regenerative medicine through the use of magnetic nanomaterials combined with remotely applied dynamic fields. Previous studies have demonstrated the ability of MNP-induced mechanoactivation to trigger downstream signaling and promote new bone formation. In this study, we aimed to compare the osteogenic induction achieved using the mechanoreceptor targets, Piezo1, Fzd1, Fzd2, and integrin alpha-5. We compared the binding efficacy of different types of agonists (antibodies vs. aptamers) to these receptors. Moreover, we optimized the aptamer concentration (2.5, 5, and 10 μg/mg) for the selected receptor to determine the optimum concentration for promoting bone formation. Our data demonstrated that the mechanoactivation of integrins (CD49e) significantly upregulated the RUNX2 and LEF1 genes compared to other selected receptors. Furthermore, comparing the mechanoactivation of cells using MNPs conjugated with CD49e antibodies and aptamers revealed that MNP-aptamers significantly enhanced the upregulation of LEF1 genes. This suggests that aptamer-mediated mechanoactivation is a promising alternative to antibody-mediated activation. Finally, our results showed that the concentration of the aptamer loaded onto the MNPs strongly influenced the mechanoactivation of the cells. These findings provide valuable insights into the use of MNP platforms for bone regeneration and highlight the potential of aptamers in promoting signaling pathways related to bone formation. The novelty of our study lies in elucidating the unique advantages of aptamers in mediating mechanoactivation, presenting a promising avenue for advancing bone regenerative strategies.
Collapse
Affiliation(s)
- Hadi Hajiali
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | | | - Alicia J. El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| |
Collapse
|
7
|
Miao L, Castillo M, Lu Y, Xiao Y, Liu Y, Burns AR, Kumar A, Gunaratne P, Michael DiPersio C, Wu M. β1 integrins regulate cellular behaviors and cardiomyocyte organization during ventricular wall formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.28.555112. [PMID: 37693495 PMCID: PMC10491119 DOI: 10.1101/2023.08.28.555112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Aims The mechanisms regulating the cellular behavior and cardiomyocyte organization during ventricular wall morphogenesis are poorly understood. Cardiomyocytes are surrounded by extracellular matrix (ECM) and interact with ECM via integrins. This study aims to determine whether and how β1 integrins regulate cardiomyocyte behavior and organization during ventricular wall morphogenesis in the mouse. Methods and Results We applied mRNA deep sequencing and immunostaining to determine the expression repertoires of α/β integrins and their ligands in the embryonic heart. Integrin β1 subunit (β1) and some of its ECM ligands are asymmetrically distributed and enriched in the luminal side of cardiomyocytes, while fibronectin surrounds cardiomyocytes, creating a network for them. Itgb1 , which encodes the β1 integrin subunit, was deleted via Nkx2.5 Cre/+ to generate myocardial-specific Itgb1 knockout (B1KO) mice. B1KO hearts display an absence of trabecular zone but a thicker compact zone. The abundances of hyaluronic acid and versican are not significantly different. Instead, fibronectin, a ligand of β1, was absent in B1KO. We examined cellular behaviors and organization via various tools. B1KO cardiomyocytes display a random cellular orientation and fail to undergo perpendicular cell division, be organized properly, and establish the proper tissue architecture to form trabeculae. The reduction of Notch1 activation was not the cause of the abnormal cellular organization in B1KO hearts. Mosaic clonal lineage tracing shows that Itgb1 regulates cardiomyocyte transmural migration and proliferation autonomously. Conclusions β1 is asymmetrically localized in the cardiomyocytes, and its ECM ligands are enriched in the luminal side of the myocardium and surrounding cardiomyocytes. β1 integrins are required for cardiomyocytes to attach to the ECM network. This engagement provides structural support for cardiomyocytes to maintain shape, undergo perpendicular division, and establish cellular organization. Deletion of Itgb1 , leading to ablation of β1 integrins, causes the dissociation of cardiomyocytes from the ECM network and failure to establish tissue architecture to form trabeculae.
Collapse
|
8
|
Amar K, Saha S, Debnath A, Weng CH, Roy A, Han KY, Chowdhury F. Reduced Cell-ECM Interactions in the EpiSC Colony Center Cause Heterogeneous Differentiation. Cells 2023; 12:cells12020326. [PMID: 36672261 PMCID: PMC9857087 DOI: 10.3390/cells12020326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Mechanoregulation of cell-extracellular matrix (ECM) interactions are crucial for dictating pluripotent stem cell differentiation. However, not all pluripotent cells respond homogeneously which results in heterogeneous cell populations. When cells, such as mouse epiblast stem cells (EpiSCs), are cultured in clusters, the heterogeneity effect during differentiation is even more pronounced. While past studies implicated variations in signaling pathways to be the root cause of heterogeneity, the biophysical aspects of differentiation have not been thoroughly considered. Here, we demonstrate that the heterogeneity of EpiSC differentiation arises from differences in the colony size and varying degrees of interactions between cells within the colonies and the ECM. Confocal imaging demonstrates that cells in the colony periphery established good contact with the surface while the cells in the colony center were separated by an average of 1-2 µm from the surface. Traction force measurements of the cells within the EpiSC colonies show that peripheral cells generate large tractions while the colony center cells do not. A finite element modeling of EpiSC colonies shows that tractions generated by the cells at the colony periphery lift off the colony center preventing the colony center from undergoing differentiation. Together, our results demonstrate a biophysical regulation of heterogeneous EpiSC colony differentiation.
Collapse
Affiliation(s)
- Kshitij Amar
- School of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Sanjoy Saha
- School of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Avishek Debnath
- School of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Chun Hung Weng
- CREOL, The College of Optics and Photonics, University of Central Florida, Orlando, FL 32816, USA
| | - Arpan Roy
- School of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Kyu Young Han
- CREOL, The College of Optics and Photonics, University of Central Florida, Orlando, FL 32816, USA
| | - Farhan Chowdhury
- School of Mechanical, Aerospace, and Materials Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
- Biomedical Engineering Program, School of Electrical, Computer, and Biomedical Engineering, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
- Materials Technology Center, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
- Correspondence:
| |
Collapse
|
9
|
Asrar H, Tucker AS. Endothelial cells during craniofacial development: Populating and patterning the head. Front Bioeng Biotechnol 2022; 10:962040. [PMID: 36105604 PMCID: PMC9465086 DOI: 10.3389/fbioe.2022.962040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022] Open
Abstract
Major organs and tissues require close association with the vasculature during development and for later function. Blood vessels are essential for efficient gas exchange and for providing metabolic sustenance to individual cells, with endothelial cells forming the basic unit of this complex vascular framework. Recent research has revealed novel roles for endothelial cells in mediating tissue morphogenesis and differentiation during development, providing an instructive role to shape the tissues as they form. This highlights the importance of providing a vasculature when constructing tissues and organs for tissue engineering. Studies in various organ systems have identified important signalling pathways crucial for regulating the cross talk between endothelial cells and their environment. This review will focus on the origin and migration of craniofacial endothelial cells and how these cells influence the development of craniofacial tissues. For this we will look at research on the interaction with the cranial neural crest, and individual organs such as the salivary glands, teeth, and jaw. Additionally, we will investigate the methods used to understand and manipulate endothelial networks during the development of craniofacial tissues, highlighting recent advances in this area.
Collapse
|
10
|
Wang X, Liang Y, Zhu Z, Li W, Shi B, Deng Y, Li C, Sha O. Fn1 Regulates the Third Pharyngeal Pouch Patterning and Morphogenesis. J Dent Res 2022; 101:1082-1091. [PMID: 35259939 DOI: 10.1177/00220345221078775] [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] [Indexed: 02/05/2023] Open
Abstract
The parathyroid and thymus are derived from the common primordia, the third pharyngeal pouch. During their development, endodermal cells actively interact with surrounding mesenchymal cells, mainly derived from neural crest cells (NCCs). However, the mechanism by which NCCs regulate the development of the third pharyngeal pouch remains largely unknown. In this study, we showed that fibronectin 1 (Fn1), which is synthesized by NCCs, modulates the functions of NCCs in the third pharyngeal pouch patterning and in the morphogenesis of the thymus/parathyroid. Loss of Fn1 in NCCs leads to decreased Foxn1 expression in the presumptive thymus domain at E11.5. In the mutant, we detected upregulation of the Hedgehog signaling activity in the presumptive parathyroid domain and downregulation of Bmp4 in the presumptive thymus domain. Tbx1, a Hedgehog signaling target gene in endoderm development, was ectopically expanded to the presumptive mutant thymus domain at E11.5. Fgf10, an important gene regulating the proliferation of endoderm development, was downregulated in the mutant NCCs. At later organogenesis stages, derivatives of the third pharyngeal pouch endoderm of mutant embryos were abnormal, showing conditions such as hypoparathyroidism, hypoplastic thymus, and ectopic thymus and parathyroid. These data support that Fn1 plays an important role in NCCs by regulating the patterning of the third pharyngeal pouch and morphogenesis of the thymus/parathyroid.
Collapse
Affiliation(s)
- X Wang
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Y Liang
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Z Zhu
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
| | - W Li
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - B Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Deng
- Department of Somatology, Shenzhen University General Hospital, Shenzhen, China
| | - C Li
- Department of Anatomy, Shantou University Medical College, Shantou, China
| | - O Sha
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
- School of Dentistry, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
11
|
Arrigo AB, Lin JHI. Endocytic Protein Defects in the Neural Crest Cell Lineage and Its Pathway Are Associated with Congenital Heart Defects. Int J Mol Sci 2021; 22:8816. [PMID: 34445520 PMCID: PMC8396181 DOI: 10.3390/ijms22168816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
Endocytic trafficking is an under-appreciated pathway in cardiac development. Several genes related to endocytic trafficking have been uncovered in a mutagenic ENU screen, in which mutations led to congenital heart defects (CHDs). In this article, we review the relationship between these genes (including LRP1 and LRP2) and cardiac neural crest cells (CNCCs) during cardiac development. Mice with an ENU-induced Lrp1 mutation exhibit a spectrum of CHDs. Conditional deletion using a floxed Lrp1 allele with different Cre drivers showed that targeting neural crest cells with Wnt1-Cre expression replicated the full cardiac phenotypes of the ENU-induced Lrp1 mutation. In addition, LRP1 function in CNCCs is required for normal OFT lengthening and survival/expansion of the cushion mesenchyme, with other cell lineages along the NCC migratory path playing an additional role. Mice with an ENU-induced and targeted Lrp2 mutation demonstrated the cardiac phenotype of common arterial trunk (CAT). Although there is no impact on CNCCs in Lrp2 mutants, the loss of LRP2 results in the depletion of sonic hedgehog (SHH)-dependent cells in the second heart field. SHH is known to be crucial for CNCC survival and proliferation, which suggests LRP2 has a non-autonomous role in CNCCs. In this article, other endocytic trafficking proteins that are associated with CHDs that may play roles in the NCC pathway during development, such as AP1B1, AP2B1, FUZ, MYH10, and HECTD1, are reviewed.
Collapse
Affiliation(s)
- Angelo B. Arrigo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
| | - Jiuann-Huey Ivy Lin
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
12
|
Karakaş N, Kiliç Ü. Integrin α5β1 Mediated Cellular Reorganization in Human Mesenchymal Stem Cells During Neuronal Differentiation. In Vivo 2021; 35:2127-2134. [PMID: 34182488 DOI: 10.21873/invivo.12482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Mesenchymal stem cells (MSCs) have been widely used for yielding neurons in culture to study nervous system pathologies and develop regenerative approaches. In this study, cellular rearrangements of human MSCs related to the expression of the fibronectin common receptor integrin α5β1 and its cell surface localization during neuronal differentiation, were examined. MATERIALS AND METHODS Proliferation kinetics of neuronal induced hMSCs (hMd-Neurons) were quantified by BrdU assay, and hMd-Neurons were immunostained for neuronal marker expression. Additionally, cDNA and protein samples were collected at different time points for integrin α5β1 expression analysis. RESULTS Endogenous integrin α5β1 expression was significantly upregulated by day 6 and maintained until day 12. Cell surface localization of α5β1 integrin was increased by day 6; the integrin was internalized into the cytosol by day 12. CONCLUSION Integrin dynamics around day 6 of differentiation might be involved in neuronal differentiation and maturation or specification of hMd-Neurons.
Collapse
Affiliation(s)
- Nihal Karakaş
- Department of Medical Biology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey; .,Regenerative and Restorative Medicine Research Center (REMER), Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Ülkan Kiliç
- Department of Medical Biology, Hamidiye School of Medicine, University of Health Sciences Turkey, Istanbul, Turkey
| |
Collapse
|
13
|
Warkala M, Chen D, Ramirez A, Jubran A, Schonning M, Wang X, Zhao H, Astrof S. Cell-Extracellular Matrix Interactions Play Multiple Essential Roles in Aortic Arch Development. Circ Res 2021; 128:e27-e44. [PMID: 33249995 PMCID: PMC7864893 DOI: 10.1161/circresaha.120.318200] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
RATIONALE Defects in the morphogenesis of the fourth pharyngeal arch arteries (PAAs) give rise to lethal birth defects. Understanding genes and mechanisms regulating PAA formation will provide important insights into the etiology and treatments for congenital heart disease. OBJECTIVE Cell-ECM (extracellular matrix) interactions play essential roles in the morphogenesis of PAAs and their derivatives, the aortic arch artery and its major branches; however, their specific functions are not well-understood. Previously, we demonstrated that integrin α5β1 and Fn1 (fibronectin) expressed in the Isl1 lineages regulate PAA formation. The objective of the current studies was to investigate cellular mechanisms by which integrin α5β1 and Fn1 regulate aortic arch artery morphogenesis. METHODS AND RESULTS Using temporal lineage tracing, whole-mount confocal imaging, and quantitative analysis of the second heart field (SHF) and endothelial cell (EC) dynamics, we show that the majority of PAA EC progenitors arise by E7.5 in the SHF and contribute to pharyngeal arch endothelium between E7.5 and E9.5. Consequently, SHF-derived ECs in the pharyngeal arches form a plexus of small blood vessels, which remodels into the PAAs by 35 somites. The remodeling of the vascular plexus is orchestrated by signals dependent on the pharyngeal ECM microenvironment, extrinsic to the endothelium. Conditional ablation of integrin α5β1 or Fn1 in the Isl1 lineages showed that signaling by the ECM regulates aortic arch artery morphogenesis at multiple steps: (1) accumulation of SHF-derived ECs in the pharyngeal arches, (2) remodeling of the EC plexus in the fourth arches into the PAAs, and (3) differentiation of neural crest-derived cells adjacent to the PAA endothelium into vascular smooth muscle cells. CONCLUSIONS PAA formation is a multistep process entailing dynamic contribution of SHF-derived ECs to pharyngeal arches, the remodeling of endothelial plexus into the PAAs, and the remodeling of the PAAs into the aortic arch artery and its major branches. Cell-ECM interactions regulated by integrin α5β1 and Fn1 play essential roles at each of these developmental stages.
Collapse
Affiliation(s)
- Michael Warkala
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Molecular Biology, Genetics, and Cancer Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Dongying Chen
- Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Ali Jubran
- Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | | | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Molecular Biology, Genetics, and Cancer Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| |
Collapse
|
14
|
Petrescu NB, Jurj A, Sorițău O, Lucaciu OP, Dirzu N, Raduly L, Berindan-Neagoe I, Cenariu M, Boșca BA, Campian RS, Ilea A. Cannabidiol and Vitamin D3 Impact on Osteogenic Differentiation of Human Dental Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2020; 56:medicina56110607. [PMID: 33198232 PMCID: PMC7697067 DOI: 10.3390/medicina56110607] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Background and objective: The aim of the present study was to establish a new differentiation protocol using cannabidiol (CBD) and vitamin D3 (Vit. D3) for a better and faster osteogenic differentiation of dental tissue derived mesenchymal stem cells (MSCs). Materials and methods: MSCs were harvested from dental follicle (DFSCs), dental pulp (DPSCs), and apical papilla (APSCs) of an impacted third molar of a 17-year old patient. The stem cells were isolated and characterized using flow cytometry; reverse transcription polymerase chain reaction (RT-PCR); and osteogenic, chondrogenic, and adipogenic differentiation. The effects of CBD and Vit. D3 on osteogenic differentiation of dental-derived stem cell were evaluated in terms of viability/metabolic activity by alamar test, expression of collagen1A, osteopontin (OP), osteocalcin (OC), and osteonectin genes and by quantification of calcium deposits by alizarin red assay. Results: Stem cell characterization revealed more typical stemness characteristics for DFSCs and DPSCs and atypical morphology and markers expression for APSCs, a phenotype that was confirmed by differences in multipotential ability. The RT-PCR quantification of bone matrix proteins expression revealed a different behavior for each cell type, APSCs having the best response for CBD. DPSCs showed the best osteogenic potential when treated with Vit. D3. Cultivation of DFSC in standard stem cell conditions induced the highest expression of osteogenic genes, suggesting the spontaneous differentiation capacity of these cells. Regarding mineralization, alizarin red assay indicated that DFSCs and APSCs were the most responsive to low doses of CBD and Vit. D3. DPSCs had the lowest mineralization levels, with a slightly better response to Vit. D3. Conclusions: This study provides evidence that DFSCs, DPSCs, and APSCs respond differently to osteoinduction stimuli and that CBD and Vit. D3 can enhance osteogenic differentiation of these types of cells under certain conditions and doses.
Collapse
Affiliation(s)
- Nausica B. Petrescu
- Department of Oral Health, University of Medicine and Pharmacy “Iuliu Hatieganu”, Victor Babes Street, No. 15, 400012 Cluj-Napoca, Romania; (N.B.P.); (R.S.C.)
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Gheorghe Marinescu Street, No. 23, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (I.B.-N.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii Street, No. 34-36, 400015 Cluj-Napoca, Romania
| | - Olga Sorițău
- Radiotherapy, Radio-biology and Tumor Biology Laboratory, The Oncology Institute “Prof. dr. Ion Chiricuta”, Republicii Street, No. 34-36, 400015 Cluj-Napoca, Romania;
| | - Ondine P. Lucaciu
- Department of Oral Health, University of Medicine and Pharmacy “Iuliu Hatieganu”, Victor Babes Street, No. 15, 400012 Cluj-Napoca, Romania; (N.B.P.); (R.S.C.)
- Correspondence: ; Tel.: +40-743-140-777
| | - Noemi Dirzu
- Research Center for Advanced Medicine, MedFuture, University of Medicine and Pharmacy “Iuliu Hatieganu”, Louis Pasteur Street, No, 4, 400000 Cluj-Napoca, Romania;
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Gheorghe Marinescu Street, No. 23, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (I.B.-N.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii Street, No. 34-36, 400015 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, Gheorghe Marinescu Street, No. 23, 400337 Cluj-Napoca, Romania; (A.J.); (L.R.); (I.B.-N.)
- Department of Functional Genomics and Experimental Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, Republicii Street, No. 34-36, 400015 Cluj-Napoca, Romania
| | - Mihai Cenariu
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Calea Manastur, No. 3-5, 400372 Cluj-Napoca, Romania;
| | - Bianca A. Boșca
- Department of Histology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Louis Pasteur Street, No. 6, 400349 Cluj-Napoca, Romania;
| | - Radu S. Campian
- Department of Oral Health, University of Medicine and Pharmacy “Iuliu Hatieganu”, Victor Babes Street, No. 15, 400012 Cluj-Napoca, Romania; (N.B.P.); (R.S.C.)
| | - Aranka Ilea
- Department of Oral Rehabilitation, University of Medicine and Pharmacy “Iuliu Hatieganu”, Victor Babes street, No. 15, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
15
|
Barqué A, Jan K, De La Fuente E, Nicholas CL, Hynes RO, Naba A. Knockout of the gene encoding the extracellular matrix protein SNED1 results in early neonatal lethality and craniofacial malformations. Dev Dyn 2020; 250:274-294. [PMID: 33012048 DOI: 10.1002/dvdy.258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/10/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The extracellular matrix (ECM) is a fundamental component of multicellular organisms that orchestrates developmental processes and controls cell and tissue organization. We previously identified the novel ECM protein SNED1 as a promoter of breast cancer metastasis and showed that its level of expression negatively correlated with breast cancer patient survival. Here, we sought to identify the roles of SNED1 during murine development. RESULTS We generated two novel Sned1 knockout mouse strains and showed that Sned1 is essential since homozygous ablation of the gene led to early neonatal lethality. Phenotypic analysis of the surviving knockout mice revealed a role for SNED1 in the development of craniofacial and skeletal structures since Sned1 knockout resulted in growth defects, nasal cavity occlusion, and craniofacial malformations. Sned1 is widely expressed in embryos, notably by cell populations undergoing epithelial-to-mesenchymal transition, such as the neural crest cells. We further show that mice with a neural-crest-cell-specific deletion of Sned1 survive, but display facial anomalies partly phenocopying the global knockout mice. CONCLUSIONS Our results demonstrate requisite roles for SNED1 during development and neonatal survival. Importantly, the deletion of 2q37.3 in humans, a region that includes the SNED1 locus, has been associated with facial dysmorphism and short stature.
Collapse
Affiliation(s)
- Anna Barqué
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Kyleen Jan
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Emanuel De La Fuente
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Christina L Nicholas
- Department of Orthodontics, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anthropology, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Alexandra Naba
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
16
|
Reciprocal integrin/integrin antagonism through kindlin-2 and Rho GTPases regulates cell cohesion and collective migration. Matrix Biol 2020; 93:60-78. [PMID: 32450218 DOI: 10.1016/j.matbio.2020.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
Collective cell behaviour during embryogenesis and tissue repair requires the coordination of intercellular junctions, cytoskeleton-dependent shape changes controlled by Rho GTPases, and integrin-dependent cell-matrix adhesion. Many different integrins are simultaneously expressed during wound healing, embryonic development, and sprouting angiogenesis, suggesting that there is extensive integrin/integrin cross-talk to regulate cell behaviour. Here, we show that fibronectin-binding β1 and β3 integrins do not act synergistically, but rather antagonize each other during collective cell processes in neuro-epithelial cells, placental trophoblasts, and endothelial cells. Reciprocal β1/β3 antagonism controls RhoA activity in a kindlin-2-dependent manner, balancing cell spreading, contractility, and intercellular adhesion. In this way, reciprocal β1/β3 antagonism controls cell cohesion and cellular plasticity to switch between extreme and opposing states, including epithelial versus mesenchymal-like phenotypes and collective versus individual cell migration. We propose that integrin/integrin antagonism is a universal mechanism to effectuate social cellular interactions, important for tissue morphogenesis, endothelial barrier function, trophoblast invasion, and sprouting angiogenesis.
Collapse
|
17
|
Cardiac Neural Crest Cells: Their Rhombomeric Specification, Migration, and Association with Heart and Great Vessel Anomalies. Cell Mol Neurobiol 2020; 41:403-429. [PMID: 32405705 DOI: 10.1007/s10571-020-00863-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Outflow tract abnormalities are the most frequent congenital heart defects. These are due to the absence or dysfunction of the two main cell types, i.e., neural crest cells and secondary heart field cells that migrate in opposite directions at the same stage of development. These cells directly govern aortic arch patterning and development, ascending aorta dilatation, semi-valvular and coronary artery development, aortopulmonary septation abnormalities, persistence of the ductus arteriosus, trunk and proximal pulmonary arteries, sub-valvular conal ventricular septal/rotational defects, and non-compaction of the left ventricle. In some cases, depending on the functional defects of these cells, additional malformations are found in the expected spatial migratory area of the cells, namely in the pharyngeal arch derivatives and cervico-facial structures. Associated non-cardiovascular anomalies are often underestimated, since the multipotency and functional alteration of these cells can result in the modification of multiple neural, epidermal, and cervical structures at different levels. In most cases, patients do not display the full phenotype of abnormalities, but congenital cardiac defects involving the ventricular outflow tract, ascending aorta, aortic arch and supra-aortic trunks should be considered as markers for possible impaired function of these cells. Neural crest cells should not be considered as a unique cell population but on the basis of their cervical rhombomere origins R3-R5 or R6-R7-R8 and specific migration patterns: R3-R4 towards arch II, R5-R6 arch III and R7-R8 arch IV and VI. A better understanding of their development may lead to the discovery of unknown associated abnormalities, thereby enabling potential improvements to be made to the therapeutic approach.
Collapse
|
18
|
Leonard CE, Taneyhill LA. The road best traveled: Neural crest migration upon the extracellular matrix. Semin Cell Dev Biol 2020; 100:177-185. [PMID: 31727473 PMCID: PMC7071992 DOI: 10.1016/j.semcdb.2019.10.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/29/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022]
Abstract
Neural crest cells have the extraordinary task of building much of the vertebrate body plan, including the craniofacial cartilage and skeleton, melanocytes, portions of the heart, and the peripheral nervous system. To execute these developmental programs, stationary premigratory neural crest cells first acquire the capacity to migrate through an extensive process known as the epithelial-to-mesenchymal transition. Once motile, neural crest cells must traverse a complex environment consisting of other cells and the protein-rich extracellular matrix in order to get to their final destinations. Herein, we will highlight some of the main molecular machinery that allow neural crest cells to first exit the neuroepithelium and then later successfully navigate this intricate in vivo milieu. Collectively, these extracellular and intracellular factors mediate the appropriate migration of neural crest cells and allow for the proper development of the vertebrate embryo.
Collapse
Affiliation(s)
- Carrie E Leonard
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA.
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742 USA.
| |
Collapse
|
19
|
Abstract
The morphogenesis of the mammalian secondary plate is a series of highly dynamic developmental process, including the palate shelves vertical outgrowth, elevation to the horizontal plane and complete fusion in the midline. Extracellular matrix (ECM) proteins not only form the basic infrastructure for palatal mesenchymal cells to adhere via integrins but also interact with cells to regulate their functions such as proliferation and differentiation. ECM remodeling is essential for palatal outgrowth, expansion, elevation, and fusion. Multiple signaling pathways important for palatogenesis such as FGF, TGF β, BMP, and SHH remodels ECM dynamics. Dysregulation of ECM such as HA synthesis or ECM breakdown enzymes MMPs or ADAMTS causes cleft palate in mouse models. A better understanding of ECM remodeling will contribute to revealing the pathogenesis of cleft palate.
Collapse
Affiliation(s)
- Xia Wang
- Health Science Center, Shenzhen University , Shenzhen, China
| | - Chunman Li
- Health Science Center, Shenzhen University , Shenzhen, China
| | - Zeyao Zhu
- Health Science Center, Shenzhen University , Shenzhen, China
| | - Li Yuan
- Department of Stomatology, Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University , Shenzhen, China
| | - Wood Yee Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong , Hong Kong, China
| | - Ou Sha
- Health Science Center, Shenzhen University , Shenzhen, China
| |
Collapse
|
20
|
Alfano D, Altomonte A, Cortes C, Bilio M, Kelly RG, Baldini A. Tbx1 regulates extracellular matrix-cell interactions in the second heart field. Hum Mol Genet 2019; 28:2295-2308. [DOI: 10.1093/hmg/ddz058] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 12/31/2022] Open
Abstract
Abstract
Tbx1, the major candidate gene for DiGeorge or 22q11.2 deletion syndrome, is required for efficient incorporation of cardiac progenitors of the second heart field (SHF) into the heart. However, the mechanisms by which TBX1 regulates this process are still unclear. Here, we have used two independent models, mouse embryos and cultured cells, to define the role of TBX1 in establishing morphological and dynamic characteristics of SHF in the mouse. We found that loss of TBX1 impairs extracellular matrix (ECM)-integrin-focal adhesion (FA) signaling in both models. Mosaic analysis in embryos suggested that this function is non-cell autonomous, and, in cultured cells, loss of TBX1 impairs cell migration and FAs. Additionally, we found that ECM-mediated integrin signaling is disrupted upon loss of TBX1. Finally, we show that interfering with the ECM-integrin-FA axis between E8.5 and E9.5 in mouse embryos, corresponding to the time window within which TBX1 is required in the SHF, causes outflow tract dysmorphogenesis. Our results demonstrate that TBX1 is required to maintain the integrity of ECM-cell interactions in the SHF and that this interaction is critical for cardiac outflow tract development. More broadly, our data identifies a novel TBX1 downstream pathway as an important player in SHF tissue architecture and cardiac morphogenesis.
Collapse
Affiliation(s)
- Daniela Alfano
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Alessandra Altomonte
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Claudio Cortes
- Aix-Marseille Université, CNRS UMR, IBDM, Marseille, France
| | - Marchesa Bilio
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR, IBDM, Marseille, France
| | - Antonio Baldini
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
21
|
Gunawan F, Gentile A, Fukuda R, Tsedeke AT, Jiménez-Amilburu V, Ramadass R, Iida A, Sehara-Fujisawa A, Stainier DYR. Focal adhesions are essential to drive zebrafish heart valve morphogenesis. J Cell Biol 2019; 218:1039-1054. [PMID: 30635353 PMCID: PMC6400548 DOI: 10.1083/jcb.201807175] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/07/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022] Open
Abstract
Gunawan et al. analyze at single-cell resolution collective endocardial cell migration into the extracellular matrix and the cellular rearrangements forming leaflets during zebrafish heart valve formation. They show that focal adhesion activity driven by Integrin α5β1 and Talin1 are essential to drive cardiac valve morphogenesis in zebrafish. Elucidating the morphogenetic events that shape vertebrate heart valves, complex structures that prevent retrograde blood flow, is critical to understanding valvular development and aberrations. Here, we used the zebrafish atrioventricular (AV) valve to investigate these events in real time and at single-cell resolution. We report the initial events of collective migration of AV endocardial cells (ECs) into the extracellular matrix (ECM), and their subsequent rearrangements to form the leaflets. We functionally characterize integrin-based focal adhesions (FAs), critical mediators of cell–ECM interactions, during valve morphogenesis. Using transgenes to block FA signaling specifically in AV ECs as well as loss-of-function approaches, we show that FA signaling mediated by Integrin α5β1 and Talin1 promotes AV EC migration and overall shaping of the valve leaflets. Altogether, our investigation reveals the critical processes driving cardiac valve morphogenesis in vivo and establishes the zebrafish AV valve as a vertebrate model to study FA-regulated tissue morphogenesis.
Collapse
Affiliation(s)
- Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Alessandra Gentile
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ryuichi Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ayele Taddese Tsedeke
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Vanesa Jiménez-Amilburu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Radhan Ramadass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Atsuo Iida
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
22
|
Lu S, Liu S, Wietelmann A, Kojonazarov B, Atzberger A, Tang C, Schermuly RT, Gröne HJ, Offermanns S. Developmental vascular remodeling defects and postnatal kidney failure in mice lacking Gpr116 (Adgrf5) and Eltd1 (Adgrl4). PLoS One 2017; 12:e0183166. [PMID: 28806758 PMCID: PMC5555693 DOI: 10.1371/journal.pone.0183166] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 07/31/2017] [Indexed: 11/18/2022] Open
Abstract
GPR116 (ADGRF5) and ELTD1 (ADGRL4) belong to different subfamilies of the adhesion G-protein-coupled receptor group but are both expressed in endothelial cells. We therefore analyzed their functions in mice lacking these receptors. While loss of GPR116 or ELTD1 alone had no obvious effect on cardiovascular or kidney function, mice lacking both, GPR116 and ELTD1, showed malformations of the aortic arch arteries and the cardiac outflow tract leading to perinatal lethality in about 50% of the mutants. In addition to cardiovascular malformations, surviving mice developed renal thrombotic microangiopathy as well as hemolysis and splenomegaly, and their lifespan was significantly reduced. Loss of GPR116 and ELTD1 specifically in endothelial cells or neural crest-derived cells did not recapitulate any of the phenotypes observed in GPR116-ELTD1 double deficient mice, indicating that loss of GPR116 and ELTD1 expressed by other cells accounts for the observed cardiovascular and renal defects.
Collapse
Affiliation(s)
- Shun Lu
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (SL); (SO)
| | - Shuya Liu
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Scientific Service Group Nuclear Magnetic Resonance Imaging, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Baktybek Kojonazarov
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ann Atzberger
- Flow Cytometry Service Facility, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Cong Tang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany
- * E-mail: (SL); (SO)
| |
Collapse
|
23
|
Liu Y. Earlier and broader roles of Mesp1 in cardiovascular development. Cell Mol Life Sci 2017; 74:1969-1983. [PMID: 28050627 PMCID: PMC11107530 DOI: 10.1007/s00018-016-2448-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 12/12/2022]
Abstract
Mesoderm posterior 1 is one of earliest markers of the nascent mesoderm. Its best-known function is driving the onset of the cardiovascular system. In the past decade, new evidence supports that Mesp1 acts earlier with greater breadth in cell fate decisions, and through cell-autonomous and cell non-autonomous mechanisms. This review summarizes these new aspects, with an emphasis on the upstream and downstream regulation around Mesp1 and how they may guide cell fate reprogramming.
Collapse
Affiliation(s)
- Yu Liu
- Department of Biology and Biochemistry, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
24
|
Extracellular matrix and α 5β 1 integrin signaling control the maintenance of bone formation capacity by human adipose-derived stromal cells. Sci Rep 2017; 7:44398. [PMID: 28290502 PMCID: PMC5349595 DOI: 10.1038/srep44398] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/07/2017] [Indexed: 02/07/2023] Open
Abstract
Stromal vascular fraction (SVF) cells of human adipose tissue have the capacity to generate osteogenic grafts with intrinsic vasculogenic properties. However, adipose-derived stromal/stem cells (ASC), even after minimal monolayer expansion, display poor osteogenic capacity in vivo. We investigated whether ASC bone-forming capacity may be maintained by culture within a self-produced extracellular matrix (ECM) that recapitulates the native environment. SVF cells expanded without passaging up to 28 days (Unpass-ASC) deposited a fibronectin-rich extracellular matrix and displayed greater clonogenicity and differentiation potential in vitro compared to ASC expanded only for 6 days (P0-ASC) or for 28 days with regular passaging (Pass-ASC). When implanted subcutaneously, Unpass-ASC produced bone tissue similarly to SVF cells, in contrast to P0- and Pass-ASC, which mainly formed fibrous tissue. Interestingly, clonogenic progenitors from native SVF and Unpass-ASC expressed low levels of the fibronectin receptor α5 integrin (CD49e), which was instead upregulated in P0- and Pass-ASC. Mechanistically, induced activation of α5β1 integrin in Unpass-ASC led to a significant loss of bone formation in vivo. This study shows that ECM and regulation of α5β1-integrin signaling preserve ASC progenitor properties, including bone tissue-forming capacity, during in vitro expansion.
Collapse
|
25
|
Wang X, Chen D, Chen K, Jubran A, Ramirez A, Astrof S. Endothelium in the pharyngeal arches 3, 4 and 6 is derived from the second heart field. Dev Biol 2016; 421:108-117. [PMID: 27955943 DOI: 10.1016/j.ydbio.2016.12.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/18/2016] [Accepted: 12/03/2016] [Indexed: 12/22/2022]
Abstract
Oxygenated blood from the heart is directed into the systemic circulation through the aortic arch arteries (AAAs). The AAAs arise by remodeling of three symmetrical pairs of pharyngeal arch arteries (PAAs), which connect the heart with the paired dorsal aortae at mid-gestation. Aberrant PAA formation results in defects frequently observed in patients with lethal congenital heart disease. How the PAAs form in mammals is not understood. The work presented in this manuscript shows that the second heart field (SHF) is the major source of progenitors giving rise to the endothelium of the pharyngeal arches 3 - 6, while the endothelium in the pharyngeal arches 1 and 2 is derived from a different source. During the formation of the PAAs 3 - 6, endothelial progenitors in the SHF extend cellular processes toward the pharyngeal endoderm, migrate from the SHF and assemble into a uniform vascular plexus. This plexus then undergoes remodeling, whereby plexus endothelial cells coalesce into a large PAA in each pharyngeal arch. Taken together, our studies establish a platform for investigating cellular and molecular mechanisms regulating PAA formation and alterations that lead to disease.
Collapse
Affiliation(s)
- Xia Wang
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | - Dongying Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kelley Chen
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Jefferson Medical College of Thomas Jefferson University, Clinical & Translational Research Track, USA
| | - Ali Jubran
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; The Master's of Science Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - AnnJosette Ramirez
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sophie Astrof
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA, 19107, USA; Graduate Program in Cell & Developmental Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
26
|
George L, Dunkel H, Hunnicutt BJ, Filla M, Little C, Lansford R, Lefcort F. In vivo time-lapse imaging reveals extensive neural crest and endothelial cell interactions during neural crest migration and formation of the dorsal root and sympathetic ganglia. Dev Biol 2016; 413:70-85. [PMID: 26988118 PMCID: PMC4834247 DOI: 10.1016/j.ydbio.2016.02.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/11/2016] [Accepted: 02/27/2016] [Indexed: 11/21/2022]
Abstract
During amniote embryogenesis the nervous and vascular systems interact in a process that significantly affects the respective morphogenesis of each network by forming a "neurovascular" link. The importance of neurovascular cross-talk in the central nervous system has recently come into focus with the growing awareness that these two systems interact extensively both during development, in the stem-cell niche, and in neurodegenerative conditions such as Alzheimer's Disease and Amyotrophic Lateral Sclerosis. With respect to the peripheral nervous system, however, there have been no live, real-time investigations of the potential relationship between these two developing systems. To address this deficit, we used multispectral 4D time-lapse imaging in a transgenic quail model in which endothelial cells (ECs) express a yellow fluorescent marker, while neural crest cells (NCCs) express an electroporated red fluorescent marker. We monitored EC and NCC migration in real-time during formation of the peripheral nervous system. Our time-lapse recordings indicate that NCCs and ECs are physically juxtaposed and dynamically interact at multiple locations along their trajectories. These interactions are stereotypical and occur at precise anatomical locations along the NCC migratory pathway. NCCs migrate alongside the posterior surface of developing intersomitic vessels, but fail to cross these continuous streams of motile ECs. NCCs change their morphology and migration trajectory when they encounter gaps in the developing vasculature. Within the nascent dorsal root ganglion, proximity to ECs causes filopodial retraction which curtails forward persistence of NCC motility. Overall, our time-lapse recordings support the conclusion that primary vascular networks substantially influence the distribution and migratory behavior of NCCs and the patterned formation of dorsal root and sympathetic ganglia.
Collapse
Affiliation(s)
- Lynn George
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States; Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT 59101, United States.
| | - Haley Dunkel
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| | - Barbara J Hunnicutt
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| | - Michael Filla
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Charles Little
- University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Rusty Lansford
- Department of Radiology and Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA 90027, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Frances Lefcort
- Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59717, United States
| |
Collapse
|
27
|
Vega ME, Schwarzbauer JE. Collaboration of fibronectin matrix with other extracellular signals in morphogenesis and differentiation. Curr Opin Cell Biol 2016; 42:1-6. [PMID: 27062478 DOI: 10.1016/j.ceb.2016.03.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Tissue formation and cell differentiation depend on a properly assembled extracellular matrix (ECM). Fibronectin is a key constituent of the pericellular ECM, forming essential connections between cell surface integrin receptors and structural components of the ECM. Recent studies using vertebrate models, conditional gene knockouts, tissue explants, and cell culture systems have identified developmental processes that depend on fibronectin and its receptor α5β1 integrin. We describe requirements for fibronectin matrix in the cardiovascular system, somite and precartilage development, and epithelial-mesenchymal transition. Information about molecular mechanisms shows the importance of fibronectin and integrins during tissue morphogenesis and cell differentiation, as well as their cooperation with growth factors to mediate changes in cell behaviors.
Collapse
Affiliation(s)
- Maria E Vega
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, United States
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, United States.
| |
Collapse
|
28
|
Wang X, Astrof S. Neural crest cell-autonomous roles of fibronectin in cardiovascular development. Development 2015; 143:88-100. [PMID: 26552887 DOI: 10.1242/dev.125286] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022]
Abstract
The chemical and mechanical properties of extracellular matrices (ECMs) modulate diverse aspects of cellular fates; however, how regional heterogeneity in ECM composition regulates developmental programs is not well understood. We discovered that fibronectin 1 (Fn1) is expressed in strikingly non-uniform patterns during mouse development, suggesting that regionalized synthesis of the ECM plays cell-specific regulatory roles during embryogenesis. To test this hypothesis, we ablated Fn1 in the neural crest (NC), a population of multi-potent progenitors expressing high levels of Fn1. We found that Fn1 synthesized by the NC mediated morphogenesis of the aortic arch artery and differentiation of NC cells into vascular smooth muscle cells (VSMCs) by regulating Notch signaling. We show that NC Fn1 signals in an NC cell-autonomous manner through integrin α5β1 expressed by the NC, leading to activation of Notch and differentiation of VSMCs. Our data demonstrate an essential role of the localized synthesis of Fn1 in cardiovascular development and spatial regulation of Notch signaling.
Collapse
Affiliation(s)
- Xia Wang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Sophie Astrof
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| |
Collapse
|
29
|
Chen D, Wang X, Liang D, Gordon J, Mittal A, Manley N, Degenhardt K, Astrof S. Fibronectin signals through integrin α5β1 to regulate cardiovascular development in a cell type-specific manner. Dev Biol 2015; 407:195-210. [PMID: 26434918 DOI: 10.1016/j.ydbio.2015.09.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/09/2015] [Accepted: 09/12/2015] [Indexed: 01/23/2023]
Abstract
Fibronectin (Fn1) is an evolutionarily conserved extracellular matrix glycoprotein essential for embryonic development. Global deletion of Fn1 leads to mid-gestation lethality from cardiovascular defects. However, severe morphogenetic defects that occur early in embryogenesis in these embryos precluded assigning a direct role for Fn1 in cardiovascular development. We noticed that Fn1 is expressed in strikingly non-uniform patterns during mouse embryogenesis, and that its expression is particularly enriched in the pharyngeal region corresponding with the pharyngeal arches 3, 4, and 6. This region bears a special importance for the developing cardiovascular system, and we hypothesized that the localized enrichment of Fn1 in the pharyngeal region may be essential for cardiovascular morphogenesis. To test this hypothesis, we ablated Fn1 using the Isl1(Cre) knock-in strain of mice. Deletion of Fn1 using the Isl1(Cre) strain resulted in defective formation of the 4th pharyngeal arch arteries (PAAs), aberrant development of the cardiac outflow tract (OFT), and ventricular septum defects. To determine the cell types responding to Fn1 signaling during cardiovascular development, we deleted a major Fn1 receptor, integrin α5 using the Isl1(Cre) strain, and observed the same spectrum of abnormalities seen in the Fn1 conditional mutants. Additional conditional mutagenesis studies designed to ablate integrin α5 in distinct cell types within the Isl1(+) tissues and their derivatives, suggested that the expression of integrin α5 in the pharyngeal arch mesoderm, endothelium, surface ectoderm and the neural crest were not required for PAA formation. Our studies suggest that an (as yet unknown) integrin α5-dependent signal extrinsic to the pharyngeal endothelium mediates the formation of the 4th PAAs.
Collapse
Affiliation(s)
- Dongying Chen
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA; Cell and Developmental Biology graduate program, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xia Wang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Dong Liang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Julie Gordon
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Ashok Mittal
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Nancy Manley
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107, USA
| | - Sophie Astrof
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA; Cell and Developmental Biology graduate program, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
30
|
Funato N, Nakamura M, Yanagisawa H. Molecular basis of cleft palates in mice. World J Biol Chem 2015; 6:121-138. [PMID: 26322171 PMCID: PMC4549757 DOI: 10.4331/wjbc.v6.i3.121] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 05/26/2015] [Accepted: 07/14/2015] [Indexed: 02/05/2023] Open
Abstract
Cleft palate, including complete or incomplete cleft palates, soft palate clefts, and submucosal cleft palates, is the most frequent congenital craniofacial anomaly in humans. Multifactorial conditions, including genetic and environmental factors, induce the formation of cleft palates. The process of palatogenesis is temporospatially regulated by transcription factors, growth factors, extracellular matrix proteins, and membranous molecules; a single ablation of these molecules can result in a cleft palate in vivo. Studies on knockout mice were reviewed in order to identify genetic errors that lead to cleft palates. In this review, we systematically describe these mutant mice and discuss the molecular mechanisms of palatogenesis.
Collapse
|