1
|
Huang N, Chan BP. A 3D micro-printed single cell micro-niche with asymmetric niche signals - An in vitro model for asymmetric cell division study. Biomaterials 2024; 311:122684. [PMID: 38971120 DOI: 10.1016/j.biomaterials.2024.122684] [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: 03/16/2024] [Revised: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 07/08/2024]
Abstract
Intricate microenvironment signals orchestrate to affect cell behavior and fate during tissue morphogenesis. However, the underlying mechanisms on how specific local niche signals influence cell behavior and fate are not fully understood, owing to the lack of in vitro platform able to precisely, quantitatively, spatially, and independently manipulate individual niche signals. Here, microarrays of protein-based 3D single cell micro-niche (3D-SCμN), with precisely engineered biophysical and biochemical niche signals, are micro-printed by a multiphoton microfabrication and micropatterning technology. Mouse embryonic stem cell (mESC) is used as the model cell to study how local niche signals affect stem cell behavior and fate. By precisely engineering the internal microstructures of the 3D SCμNs, we demonstrate that the cell division direction can be controlled by the biophysical niche signals, in a cell shape-independent manner. After confining the cell division direction to a dominating axis, single mESCs are exposed to asymmetric biochemical niche signals, specifically, cell-cell adhesion molecule on one side and extracellular matrix on the other side. We demonstrate that, symmetry-breaking (asymmetric) niche signals successfully trigger cell polarity formation and bias the orientation of asymmetric cell division, the mitosis process resulting in two daughter cells with differential fates, in mESCs.
Collapse
Affiliation(s)
- Nan Huang
- Tissue Engineering Laboratory, Biomedical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Tissue Engineering Laboratory, School of Biomedical Sciences, Institute of Tissue Engineering and Regenerative Medicine, And Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong Special Administrative Region of China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory, Biomedical Engineering Program, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region of China; Tissue Engineering Laboratory, School of Biomedical Sciences, Institute of Tissue Engineering and Regenerative Medicine, And Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong Special Administrative Region of China.
| |
Collapse
|
2
|
Li B, Kwon C. Mesendodermal cells fail to contribute to heart formation following blastocyst injection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595392. [PMID: 38826381 PMCID: PMC11142170 DOI: 10.1101/2024.05.22.595392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Blastocyst complementation offers an opportunity for generating transplantable whole organs from donor sources. Pluripotent stem cells (PSCs) have traditionally served as the primary donor cells due to their ability to differentiate into any type of body cell. However, the use of PSCs raises ethical concerns, particularly regarding their uncontrollable differentiation potential to undesired cell lineages such as brain and germline cells. To address this issue, various strategies have been explored, including the use of genetically modified PSCs with restricted lineage potential or lineage-specified progenitor cells as donors. In this study, we tested whether nascent mesendodermal cells (MECs), which appear during early gastrulation, can be used as donor cells. To do this, we induced Bry-GFP+ MECs from mouse embryonic stem cells (ESCs) and introduced them into the blastocyst. While donor ESCs gave rise to various regions of embryos, including the heart, Bry-GFP+ MECs failed to contribute to the host embryos. This finding suggests that MECs, despite being specified from PSCs within a few days, lack the capacity to assimilate into the developing embryo.
Collapse
Affiliation(s)
- Biyi Li
- Division of Cardiology, Department of Medicine, Department of Biomedical Engineering, Department of Cell Biology, Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Department of Biomedical Engineering, Department of Cell Biology, Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Krup AL, Winchester SAB, Ranade SS, Agrawal A, Devine WP, Sinha T, Choudhary K, Dominguez MH, Thomas R, Black BL, Srivastava D, Bruneau BG. A Mesp1-dependent developmental breakpoint in transcriptional and epigenomic specification of early cardiac precursors. Development 2023; 150:dev201229. [PMID: 36994838 PMCID: PMC10259516 DOI: 10.1242/dev.201229] [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: 08/23/2022] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Transcriptional networks governing cardiac precursor cell (CPC) specification are incompletely understood owing, in part, to limitations in distinguishing CPCs from non-cardiac mesoderm in early gastrulation. We leveraged detection of early cardiac lineage transgenes within a granular single-cell transcriptomic time course of mouse embryos to identify emerging CPCs and describe their transcriptional profiles. Mesp1, a transiently expressed mesodermal transcription factor, is canonically described as an early regulator of cardiac specification. However, we observed perdurance of CPC transgene-expressing cells in Mesp1 mutants, albeit mislocalized, prompting us to investigate the scope of the role of Mesp1 in CPC emergence and differentiation. Mesp1 mutant CPCs failed to robustly activate markers of cardiomyocyte maturity and crucial cardiac transcription factors, yet they exhibited transcriptional profiles resembling cardiac mesoderm progressing towards cardiomyocyte fates. Single-cell chromatin accessibility analysis defined a Mesp1-dependent developmental breakpoint in cardiac lineage progression at a shift from mesendoderm transcriptional networks to those necessary for cardiac patterning and morphogenesis. These results reveal Mesp1-independent aspects of early CPC specification and underscore a Mesp1-dependent regulatory landscape required for progression through cardiogenesis.
Collapse
Affiliation(s)
- Alexis Leigh Krup
- Biomedical Sciences Program, University of California, San Francisco, CA 94158, USA
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sarah A. B. Winchester
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Sanjeev S. Ranade
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ayushi Agrawal
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - W. Patrick Devine
- Department of Pathology, University of California, San Francisco, CA 94158, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Krishna Choudhary
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Martin H. Dominguez
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
- Department of Medicine, Division of Cardiology, University of California, San Francisco, CA 94158, USA
- Cardiovascular Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Reuben Thomas
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
| | - Deepak Srivastava
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Benoit G. Bruneau
- Gladstone Institutes of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Pediatrics, University of California, San Francisco, CA 94158, USA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA 94158, USA
- Institute of Human Genetics, University of California, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
4
|
Armendariz DA, Goetsch SC, Sundarrajan A, Sivakumar S, Wang Y, Xie S, Munshi NV, Hon GC. CHD-associated enhancers shape human cardiomyocyte lineage commitment. eLife 2023; 12:e86206. [PMID: 37096669 PMCID: PMC10156167 DOI: 10.7554/elife.86206] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/10/2023] [Indexed: 04/26/2023] Open
Abstract
Enhancers orchestrate gene expression programs that drive multicellular development and lineage commitment. Thus, genetic variants at enhancers are thought to contribute to developmental diseases by altering cell fate commitment. However, while many variant-containing enhancers have been identified, studies to endogenously test the impact of these enhancers on lineage commitment have been lacking. We perform a single-cell CRISPRi screen to assess the endogenous roles of 25 enhancers and putative cardiac target genes implicated in genetic studies of congenital heart defects (CHDs). We identify 16 enhancers whose repression leads to deficient differentiation of human cardiomyocytes (CMs). A focused CRISPRi validation screen shows that repression of TBX5 enhancers delays the transcriptional switch from mid- to late-stage CM states. Endogenous genetic deletions of two TBX5 enhancers phenocopy epigenetic perturbations. Together, these results identify critical enhancers of cardiac development and suggest that misregulation of these enhancers could contribute to cardiac defects in human patients.
Collapse
Affiliation(s)
- Daniel A Armendariz
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
| | - Sean C Goetsch
- Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
| | - Anjana Sundarrajan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
| | - Sushama Sivakumar
- Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
| | - Yihan Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
| | - Shiqi Xie
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nikhil V Munshi
- Department of Internal Medicine, University of Texas Southwestern Medical CenterDallasUnited States
- Division of Cardiology, Department of Molecular Biology, McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical CenterDallasUnited States
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical CenterDallasUnited States
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical CenterDallasUnited States
- Lyda Hill Department of Bioinformatics, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
5
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
6
|
Zhang J, Gregorich ZR, Tao R, Kim GC, Lalit PA, Carvalho JL, Markandeya Y, Mosher DF, Palecek SP, Kamp TJ. Cardiac differentiation of human pluripotent stem cells using defined extracellular matrix proteins reveals essential role of fibronectin. eLife 2022; 11:e69028. [PMID: 35758861 PMCID: PMC9236614 DOI: 10.7554/elife.69028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/05/2022] [Indexed: 11/13/2022] Open
Abstract
Research and therapeutic applications using human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) require robust differentiation strategies. Efforts to improve hPSC-CM differentiation have largely overlooked the role of extracellular matrix (ECM). The present study investigates the ability of defined ECM proteins to promote hPSC cardiac differentiation. Fibronectin (FN), laminin-111, and laminin-521 enabled hPSCs to attach and expand. However, only addition of FN promoted cardiac differentiation in response to growth factors Activin A, BMP4, and bFGF in contrast to the inhibition produced by laminin-111 or laminin-521. hPSCs in culture produced endogenous FN which accumulated in the ECM to a critical level necessary for effective cardiac differentiation. Inducible shRNA knockdown of FN prevented Brachyury+ mesoderm formation and subsequent hPSC-CM generation. Antibodies blocking FN binding integrins α4β1 or αVβ1, but not α5β1, inhibited cardiac differentiation. Furthermore, inhibition of integrin-linked kinase led to a decrease in phosphorylated AKT, which was associated with increased apoptosis and inhibition of cardiac differentiation. These results provide new insights into defined matrices for culture of hPSCs that enable production of FN-enriched ECM which is essential for mesoderm formation and efficient cardiac differentiation.
Collapse
Affiliation(s)
- Jianhua Zhang
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
| | - Zachery R Gregorich
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Ran Tao
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Gina C Kim
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Pratik A Lalit
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Juliana L Carvalho
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Department of Genomic Sciences and Biotechnology, University of BrasíliaBrasíliaBrazil
| | - Yogananda Markandeya
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| | - Deane F Mosher
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Morgridge Institute for ResearchMadisonUnited States
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-MadisonMadisonUnited States
| | - Sean P Palecek
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Chemical and Biological Engineering, College of Engineering, University of WisconsinMadisonUnited States
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
- Stem Cell and Regenerative Medicine Center, University of Wisconsin - MadisonMadisonUnited States
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - MadisonMadisonUnited States
| |
Collapse
|
7
|
Miyamoto M, Andersen P, Sulistio E, Liu X, Murphy S, Kannan S, Nam L, Miyamoto W, Tampakakis E, Hibino N, Uosaki H, Kwon C. Noncanonical Notch signals have opposing roles during cardiac development. Biochem Biophys Res Commun 2021; 577:12-16. [PMID: 34487959 PMCID: PMC8484041 DOI: 10.1016/j.bbrc.2021.08.094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The Notch pathway is an ancient intercellular signaling system with crucial roles in numerous cell-fate decision processes across species. While the canonical pathway is activated by ligand-induced cleavage and nuclear localization of membrane-bound Notch, Notch can also exert its activity in a ligand/transcription-independent fashion, which is conserved in Drosophila, Xenopus, and mammals. However, the noncanonical role remains poorly understood in in vivo processes. Here we show that increased levels of the Notch intracellular domain (NICD) in the early mesoderm inhibit heart development, potentially through impaired induction of the second heart field (SHF), independently of the transcriptional effector RBP-J. Similarly, inhibiting Notch cleavage, shown to increase noncanonical Notch activity, suppressed SHF induction in embryonic stem cell (ESC)-derived mesodermal cells. In contrast, NICD overexpression in late cardiac progenitor cells lacking RBP-J resulted in an increase in heart size. Our study suggests that noncanonical Notch signaling has stage-specific roles during cardiac development.
Collapse
Affiliation(s)
- Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Edrick Sulistio
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Xihe Liu
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Lucy Nam
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD,Present address: Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - William Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD,Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University Baltimore, MD
| | - Emmanouil Tampakakis
- Division of Cardiology, Department of Medicine, Johns Hopkins University Baltimore, MD,Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Narutoshi Hibino
- Department of Surgery, The University of Chicago Medical Center, Chicago, IL
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA; Heart and Vascular Institute, Cellular and Molecular Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
8
|
Kinases of the Focal Adhesion Complex Contribute to Cardiomyocyte Specification. Int J Mol Sci 2021; 22:ijms221910430. [PMID: 34638793 PMCID: PMC8508671 DOI: 10.3390/ijms221910430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/03/2021] [Accepted: 09/22/2021] [Indexed: 11/17/2022] Open
Abstract
Differentiation of pluripotent stem cells to cardiomyocytes is influenced by culture conditions including the extracellular matrices or similar synthetic scaffolds on which they are grown. However, the molecular mechanisms that link the scaffold with differentiation outcomes are not fully known. Here, we determined by immunofluorescence staining and mass spectrometry approaches that extracellular matrix (ECM) engagement by mouse pluripotent stem cells activates critical components of canonical wingless/integrated (Wnt) signaling pathways via kinases of the focal adhesion to drive cardiomyogenesis. These kinases were found to be differentially activated depending on type of ECM engaged. These outcomes begin to explain how varied ECM composition of in vivo tissues with development and in vitro model systems gives rise to different mature cell types, having broad practical applicability for the design of engineered tissues.
Collapse
|
9
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
10
|
Schussler O, Chachques JC, Alifano M, Lecarpentier Y. Key Roles of RGD-Recognizing Integrins During Cardiac Development, on Cardiac Cells, and After Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:179-203. [PMID: 34342855 DOI: 10.1007/s12265-021-10154-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Cardiac cells interact with the extracellular matrix (ECM) proteins through integrin mechanoreceptors that control many cellular events such as cell survival, apoptosis, differentiation, migration, and proliferation. Integrins play a crucial role in cardiac development as well as in cardiac fibrosis and hypertrophy. Integrins recognize oligopeptides present on ECM proteins and are involved in three main types of interaction, namely with collagen, laminin, and the oligopeptide RGD (Arg-Gly-Asp) present on vitronectin and fibronectin proteins. To date, the specific role of integrins recognizing the RGD has not been addressed. In this review, we examine their role during cardiac development, their role on cardiac cells, and their upregulation during pathological processes such as heart fibrosis and hypertrophy. We also examine their role in regenerative and angiogenic processes after myocardial infarction (MI) in the peri-infarct area. Specific targeting of these integrins may be a way of controlling some of these pathological events and thereby improving medical outcomes.
Collapse
Affiliation(s)
- Olivier Schussler
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.
| | - Juan C Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.,INSERM U1138 Team "Cancer, Immune Control, and Escape", Cordeliers Research Center, University of Paris, Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
11
|
Pakzad KK, Tan JJ, Anderson S, Board M, Clarke K, Carr CA. Metabolic maturation of differentiating cardiosphere-derived cells. Stem Cell Res 2021; 54:102422. [PMID: 34118565 PMCID: PMC8271094 DOI: 10.1016/j.scr.2021.102422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/13/2022] Open
Abstract
Collagen IV promotes proliferation of cardiosphere-derived cells. Fibronectin supports differentiation of cardiosphere-derived cells. Oxidative metabolism increases as cardiac progenitors mature. Stimulating fatty acid oxidation promotes cardiac progenitor cell maturation.
Cardiosphere-derived cells (CDCs) can be expanded in vitro and induced to differentiate along the cardiac lineage. To recapitulate the phenotype of an adult cardiomyocyte, differentiating progenitors need to upregulate mitochondrial glucose and fatty acid oxidation. Here we cultured and differentiated CDCs using protocols aimed to maintain stemness or to promote differentiation, including triggering fatty acid oxidation using an agonist of peroxisome proliferator-activated receptor alpha (PPARα). Metabolic changes were characterised in undifferentiated CDCs and during differentiation towards a cardiac phenotype. CDCs from rat atria were expanded on fibronectin or collagen IV via cardiosphere formation. Differentiation was assessed using flow cytometry and qPCR and substrate metabolism was quantified using radiolabelled substrates. Collagen IV promoted proliferation of CDCs whereas fibronectin primed cells for differentiation towards a cardiac phenotype. In both populations, treatment with 5-Azacytidine induced a switch towards oxidative metabolism, as shown by changes in gene expression, decreased glycolytic flux and increased oxidation of glucose and palmitate. Addition of a PPARα agonist during differentiation increased both glucose and fatty acid oxidation and expression of cardiac genes. We conclude that oxidative metabolism and cell differentiation act in partnership with increases in one driving an increase in the other.
Collapse
Affiliation(s)
| | - Jun Jie Tan
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK; Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | | | - Mary Board
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Kieran Clarke
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK.
| |
Collapse
|
12
|
Sun X, Malandraki-Miller S, Kennedy T, Bassat E, Klaourakis K, Zhao J, Gamen E, Vieira JM, Tzahor E, Riley PR. The extracellular matrix protein agrin is essential for epicardial epithelial-to-mesenchymal transition during heart development. Development 2021; 148:261801. [PMID: 33969874 PMCID: PMC8172119 DOI: 10.1242/dev.197525] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 04/03/2021] [Indexed: 12/15/2022]
Abstract
During heart development, epicardial cells residing within the outer layer undergo epithelial-mesenchymal transition (EMT) and migrate into the underlying myocardium to support organ growth and morphogenesis. Disruption of epicardial EMT results in embryonic lethality, yet its regulation is poorly understood. Here, we report epicardial EMT within the mesothelial layer of the mouse embryonic heart at ultra-high resolution using scanning electron microscopy combined with immunofluorescence analyses. We identified morphologically active EMT regions that associated with key components of the extracellular matrix, including the basement membrane-associated proteoglycan agrin. Deletion of agrin resulted in impaired EMT and compromised development of the epicardium, accompanied by downregulation of Wilms' tumor 1. Agrin enhanced EMT in human embryonic stem cell-derived epicardial-like cells by decreasing β-catenin and promoting pFAK localization at focal adhesions, and promoted the aggregation of dystroglycan within the Golgi apparatus in murine epicardial cells. Loss of agrin resulted in dispersal of dystroglycan in vivo, disrupting basement membrane integrity and impairing EMT. Our results provide new insights into the role of the extracellular matrix in heart development and implicate agrin as a crucial regulator of epicardial EMT.
Collapse
Affiliation(s)
- Xin Sun
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Sophia Malandraki-Miller
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Tahnee Kennedy
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Elad Bassat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Konstantinos Klaourakis
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Jia Zhao
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Elisabetta Gamen
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Joaquim Miguel Vieira
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Paul R Riley
- Burdon-Sanderson Cardiac Science Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK.,British Heart Foundation - Oxbridge Centre of Regenerative Medicine, CRM, University of Oxford, Oxford OX1 3PT, UK
| |
Collapse
|
13
|
Pezhouman A, Engel JL, Nguyen NB, Skelton RJP, Gilmore WB, Qiao R, Sahoo D, Zhao P, Elliott DA, Ardehali R. Isolation and characterization of hESC-derived heart field-specific cardiomyocytes unravels new insights into their transcriptional and electrophysiological profiles. Cardiovasc Res 2021; 118:828-843. [PMID: 33744937 DOI: 10.1093/cvr/cvab102] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/21/2020] [Accepted: 03/18/2021] [Indexed: 12/16/2022] Open
Abstract
AIMS We prospectively isolate and characterize first and second heart field- and nodal-like cardiomyocytes using a double reporter line from human embryonic stem cells. Our double reporter line utilizes two important transcription factors in cardiac development, TBX5 and NKX2-5. TBX5 expression marks first heart field progenitors and cardiomyocytes while NKX2-5 is expressed in nearly all myocytes of the developing heart (excluding nodal cells). We address the shortcomings of prior work in the generation of heart-field specific cardiomyocytes from induced pluripotent stem cells and provide a comprehensive early developmental transcriptomic as well as electrophysiological analyses of these three populations. METHODS AND RESULTS Transcriptional, immunocytochemical, and functional studies support the cellular identities of isolated populations based on the expression pattern of NKX2-5 and TBX5. Importantly, bulk and single-cell RNA sequencing analyses provide evidence of unique molecular signatures of isolated first and second heart-field cardiomyocytes, as well as nodal-like cells. Extensive electrophysiological analyses reveal dominant atrial action potential phenotypes in first and second heart fields in alignment with our findings in single-cell RNA sequencing. Lastly, we identify two novel surface markers, POPDC2 and CORIN, that enables purification of cardiomyocytes and first heart field cardiomyocytes, respectively. CONCLUSIONS We describe a high yield approach for isolation and characterization of human embryonic stem cell-derived heart field specific and nodal-like cardiomyocytes. Obtaining enriched populations of these different cardiomyocyte subtypes increases the resolution of gene expression profiling during early cardiogenesis, arrhythmia modeling, and drug screening. This paves the way for the development of effective stem cell therapy to treat diseases that affect specific regions of the heart or chamber-specific congenital heart defects. TRANSLATIONAL PERSPECTIVE Myocardial infarction leads to irreversible loss of cardiomyocytes and eventually heart failure. Human embryonic stem cells (hESCs) can be differentiated to cardiomyocytes and are considered a potential source of cell therapy for cardiac regeneration. However, current differentiation strategies yield a mixture of cardiomyocyte subtypes and safety concerns stemming from the use of a heterogenous population of cardiomyocytes have hindered its application. Here, we report generation of enriched heart field-specific cardiomyocytes using a hESC double reporter. Our study facilitates investigating early human cardiogenesis in vitro and generating chamber-specific cardiomyocytes to treat diseases that affect specific regions of the heart.
Collapse
Affiliation(s)
- Arash Pezhouman
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA
| | - James L Engel
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA
| | - Ngoc B Nguyen
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, California 90095, USA
| | - Rhys J P Skelton
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA
| | - W Blake Gilmore
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA
| | - Rong Qiao
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA
| | - Debashis Sahoo
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Peng Zhao
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| | - David A Elliott
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, 3052, Australia.,Department of Paediatrics, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA.,Eli and Edy the Broad Stem Cell Research Center, University of California, Los Angeles, California 90095, USA.,Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, California 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Fibronectin in development and wound healing. Adv Drug Deliv Rev 2021; 170:353-368. [PMID: 32961203 DOI: 10.1016/j.addr.2020.09.005] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 09/15/2020] [Indexed: 01/15/2023]
Abstract
Fibronectin structure and composition regulate contextual cell signaling. Recent advances have been made in understanding fibronectin and its role in tissue organization and repair. This review outlines fibronectin splice variants and their functions, evaluates potential therapeutic strategies targeting or utilizing fibronectin, and concludes by discussing potential future directions to modulate fibronectin function in development and wound healing.
Collapse
|
15
|
Song R, Zhang L. Cardiac ECM: Its Epigenetic Regulation and Role in Heart Development and Repair. Int J Mol Sci 2020; 21:ijms21228610. [PMID: 33203135 PMCID: PMC7698074 DOI: 10.3390/ijms21228610] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/07/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) is the non-cellular component in the cardiac microenvironment, and serves essential structural and regulatory roles in establishing and maintaining tissue architecture and cellular function. The patterns of molecular and biochemical ECM alterations in developing and adult hearts depend on the underlying injury type. In addition to exploring how the ECM regulates heart structure and function in heart development and repair, this review conducts an inclusive discussion of recent developments in the role, function, and epigenetic guidelines of the ECM. Moreover, it contributes to the development of new therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Rui Song
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| | - Lubo Zhang
- Correspondence: (R.S.); (L.Z.); Tel.: +1-909-558-4325 (R.S. & L.Z.)
| |
Collapse
|
16
|
Pagliarosi O, Picchio V, Chimenti I, Messina E, Gaetani R. Building an Artificial Cardiac Microenvironment: A Focus on the Extracellular Matrix. Front Cell Dev Biol 2020; 8:559032. [PMID: 33015056 PMCID: PMC7500153 DOI: 10.3389/fcell.2020.559032] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
The increased knowledge in cell signals and stem cell differentiation, together with the development of new technologies, such as 3D bioprinting, has made the generation of artificial tissues more feasible for in vitro studies and in vivo applications. In the human body, cell fate, function, and survival are determined by the microenvironment, a rich and complex network composed of extracellular matrix (ECM), different cell types, and soluble factors. They all interconnect and communicate, receiving and sending signals, modulating and responding to cues. In the cardiovascular field, the culture of stem cells in vitro and their differentiation into cardiac phenotypes is well established, although differentiated cardiomyocytes often lack the functional maturation and structural organization typical of the adult myocardium. The recreation of an artificial microenvironment as similar as possible to the native tissue, though, has been shown to partly overcome these limitations, and can be obtained through the proper combination of ECM molecules, different cell types, bioavailability of growth factors (GFs), as well as appropriate mechanical and geometrical stimuli. This review will focus on the role of the ECM in the regulation of cardiac differentiation, will provide new insights on the role of supporting cells in the generation of 3D artificial tissues, and will also present a selection of the latest approaches to recreate a cardiac microenvironment in vitro through 3D bioprinting approaches.
Collapse
Affiliation(s)
- Olivia Pagliarosi
- Department of Molecular Medicine, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy
| | - Vittorio Picchio
- Department of Medical and Surgical Sciences and Biotechnology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy
| | - Isotta Chimenti
- Department of Medical and Surgical Sciences and Biotechnology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Elisa Messina
- Department of Maternal, Infantile, and Urological Sciences, “Umberto I” Hospital, Rome, Italy
| | - Roberto Gaetani
- Department of Molecular Medicine, Faculty of Pharmacy and Medicine, Sapienza University of Rome, Rome, Italy
- Department of Bioengineering, Sanford Consortium for Regenerative Medicine, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
17
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
18
|
Kupfer ME, Lin WH, Ravikumar V, Qiu K, Wang L, Gao L, Bhuiyan DB, Lenz M, Ai J, Mahutga RR, Townsend D, Zhang J, McAlpine MC, Tolkacheva EG, Ogle BM. In Situ Expansion, Differentiation, and Electromechanical Coupling of Human Cardiac Muscle in a 3D Bioprinted, Chambered Organoid. Circ Res 2020; 127:207-224. [PMID: 32228120 DOI: 10.1161/circresaha.119.316155] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
RATIONALE One goal of cardiac tissue engineering is the generation of a living, human pump in vitro that could replace animal models and eventually serve as an in vivo therapeutic. Models that replicate the geometrically complex structure of the heart, harboring chambers and large vessels with soft biomaterials, can be achieved using 3-dimensional bioprinting. Yet, inclusion of contiguous, living muscle to support pump function has not been achieved. This is largely due to the challenge of attaining high densities of cardiomyocytes-a notoriously nonproliferative cell type. An alternative strategy is to print with human induced pluripotent stem cells, which can proliferate to high densities and fill tissue spaces, and subsequently differentiate them into cardiomyocytes in situ. OBJECTIVE To develop a bioink capable of promoting human induced pluripotent stem cell proliferation and cardiomyocyte differentiation to 3-dimensionally print electromechanically functional, chambered organoids composed of contiguous cardiac muscle. METHODS AND RESULTS We optimized a photo-crosslinkable formulation of native ECM (extracellular matrix) proteins and used this bioink to 3-dimensionally print human induced pluripotent stem cell-laden structures with 2 chambers and a vessel inlet and outlet. After human induced pluripotent stem cells proliferated to a sufficient density, we differentiated the cells within the structure and demonstrated function of the resultant human chambered muscle pump. Human chambered muscle pumps demonstrated macroscale beating and continuous action potential propagation with responsiveness to drugs and pacing. The connected chambers allowed for perfusion and enabled replication of pressure/volume relationships fundamental to the study of heart function and remodeling with health and disease. CONCLUSIONS This advance represents a critical step toward generating macroscale tissues, akin to aggregate-based organoids, but with the critical advantage of harboring geometric structures essential to the pump function of cardiac muscle. Looking forward, human chambered organoids of this type might also serve as a test bed for cardiac medical devices and eventually lead to therapeutic tissue grafting.
Collapse
Affiliation(s)
- Molly E Kupfer
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Wei-Han Lin
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Vasanth Ravikumar
- Department of Electrical Engineering (V.R.), University of Minnesota-Twin Cities, Minneapolis
| | - Kaiyan Qiu
- Department of Mechanical Engineering (K.Q., M.C.M.), University of Minnesota-Twin Cities, Minneapolis
| | - Lu Wang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Ling Gao
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Didarul B Bhuiyan
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Megan Lenz
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Jeffrey Ai
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Ryan R Mahutga
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - DeWayne Townsend
- Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Department of Integrative Biology and Physiology (D.T.), University of Minnesota-Twin Cities, Minneapolis
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, University of Alabama at Birmingham (L.W., L.G., J.Z.)
| | - Michael C McAlpine
- Department of Mechanical Engineering (K.Q., M.C.M.), University of Minnesota-Twin Cities, Minneapolis
| | - Elena G Tolkacheva
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Institute for Engineering in Medicine (E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| | - Brenda M Ogle
- From the Department of Biomedical Engineering (M.E.K., W.-H.L., D.B.B., M.L., J.A., R.R.M., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Stem Cell Institute (M.E.K., W.-H.L., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Lillehei Heart Institute (D.T., E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Institute for Engineering in Medicine (E.G.T., B.M.O.), University of Minnesota-Twin Cities, Minneapolis.,Masonic Cancer Center (B.M.O.), University of Minnesota-Twin Cities, Minneapolis
| |
Collapse
|
19
|
Robert AW, Pereira IT, Dallagiovanna B, Stimamiglio MA. Secretome Analysis Performed During in vitro Cardiac Differentiation: Discovering the Cardiac Microenvironment. Front Cell Dev Biol 2020; 8:49. [PMID: 32117977 PMCID: PMC7025591 DOI: 10.3389/fcell.2020.00049] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Human pluripotent stem cells are an important tool for the study of developmental processes, such as cardiomyogenic differentiation. Despite the advances made in this field, the molecular and cellular signals involved in the commitment of embryonic stem cells to the cardiac phenotype are still under investigation. Therefore, this study focuses on identifying the extracellular signals involved in in vitro cardiac differentiation of human embryonic stem cells. Using a three-dimensional cardiomyogenic differentiation protocol, the conditioned medium and the extracellular matrix (ECM) of embryoid body cultures were collected and characterized at four specific time points. Mass spectrometry (MS) and antibody array analysis of the secretome identified a number of secreted proteins related to signaling pathways, such as Wnt and TGFβ, as well as many ECM proteins. When comparing the proteins identified at selected time points, our data pointed out protein interactions and biological process related to cardiac differentiation. Interestingly, the great changes in secretome profile occurred during the cardiac progenitor specification. The secretome results were also compared with our previous RNAseq data, indicating that the secreted proteins undergo some level of gene regulation. During cardiac commitment it was observed an increase in complexity of the ECM, and some proteins as IGFBP7, FN1, HSPG2, as well as other members of the basal lamina could be highlighted. Thus, these findings contribute valuable information about essential microenvironmental signals working on cardiomyogenic differentiation that may be used in future strategies for cardiac differentiation, cardiomyocyte maturation, and in advances for future acellular therapies.
Collapse
Affiliation(s)
- Anny Waloski Robert
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas - Fiocruz-Paraná, Curitiba, Brazil
| | - Isabela Tiemy Pereira
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas - Fiocruz-Paraná, Curitiba, Brazil
| | - Bruno Dallagiovanna
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas - Fiocruz-Paraná, Curitiba, Brazil
| | - Marco Augusto Stimamiglio
- Laboratório de Biologia Básica de Células-Tronco, Instituto Carlos Chagas - Fiocruz-Paraná, Curitiba, Brazil
| |
Collapse
|
20
|
Sagy N, Slovin S, Allalouf M, Pour M, Savyon G, Boxman J, Nachman I. Prediction and control of symmetry breaking in embryoid bodies by environment and signal integration. Development 2019; 146:dev.181917. [PMID: 31575644 DOI: 10.1242/dev.181917] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/16/2019] [Indexed: 01/03/2023]
Abstract
During early embryogenesis, mechanical constraints and localized biochemical signals co-occur around anteroposterior axis determination and symmetry breaking. Their relative roles, however, are hard to tease apart in vivo Using brachyury (Bra), a primitive streak and mesendoderm marker in mouse embryoid bodies (EBs), we studied how contact, biochemical cues and neighboring cell cues affect the positioning of a primitive streak-like locus and thus determine the anteroposterior axis. We show that a Bra-competent layer must be formed in the EB before Bra expression initiates, and that Bra onset locus position is biased by contact points of the EB with its surrounding, probably through modulation of chemical cues rather than by mechanical signaling. We can push or pull Bra onset away from contact points by introducing a separate localized Wnt signal source, or maneuver Bra onset to a few loci or to an isotropic peripheral pattern. Furthermore, we show that Foxa2-positive cells are predictive of the future location of Bra onset, demonstrating an earlier symmetry-breaking event. Our analysis of factors affecting symmetry breaking and spatial fate choice during this developmental process could prove valuable for in vitro differentiation and organoid formation.
Collapse
Affiliation(s)
- Naor Sagy
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shaked Slovin
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Maya Allalouf
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Maayan Pour
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gaya Savyon
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Jonathan Boxman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Iftach Nachman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
21
|
ECM alterations in Fndc3a (Fibronectin Domain Containing Protein 3A) deficient zebrafish cause temporal fin development and regeneration defects. Sci Rep 2019; 9:13383. [PMID: 31527654 PMCID: PMC6746793 DOI: 10.1038/s41598-019-50055-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/05/2019] [Indexed: 11/08/2022] Open
Abstract
Fin development and regeneration are complex biological processes that are highly relevant in teleost fish. They share genetic factors, signaling pathways and cellular properties to coordinate formation of regularly shaped extremities. Especially correct tissue structure defined by extracellular matrix (ECM) formation is essential. Gene expression and protein localization studies demonstrated expression of fndc3a (fibronectin domain containing protein 3a) in both developing and regenerating caudal fins of zebrafish (Danio rerio). We established a hypomorphic fndc3a mutant line (fndc3awue1/wue1) via CRISPR/Cas9, exhibiting phenotypic malformations and changed gene expression patterns during early stages of median fin fold development. These developmental effects are mostly temporary, but result in a fraction of adults with permanent tail fin deformations. In addition, caudal fin regeneration in adult fndc3awue1/wue1 mutants is hampered by interference with actinotrichia formation and epidermal cell organization. Investigation of the ECM implies that loss of epidermal tissue structure is a common cause for both of the observed defects. Our results thereby provide a molecular link between these developmental processes and foreshadow Fndc3a as a novel temporal regulator of epidermal cell properties during extremity development and regeneration in zebrafish.
Collapse
|
22
|
Lee S, Kim JE, Seo HJ, Jang JH. Design of fibronectin type III domains fused to an elastin-like polypeptide for the osteogenic differentiation of human mesenchymal stem cells. Acta Biochim Biophys Sin (Shanghai) 2019; 51:856-863. [PMID: 31267123 DOI: 10.1093/abbs/gmz063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Indexed: 12/23/2022] Open
Abstract
Extracellular matrix (ECM) including fibronectin (FN) and elastin plays a pivotal role in providing a microenvironment to support tissue regeneration in stem cell therapy. To develop a novel biomimetic ECM for stem cell differentiation, we engineered FN type III 9 and 10 domains fused to elastin-like polypeptides (FN-ELPs). The recombinant FN-ELP fusion protein was expressed in Escherichia coli and purified by inverse transition cycling. Human mesenchymal stem cells (hMSCs) cultured on plates coated with FN-ELP had significantly greater adhesion activity and proliferation than cells grown on non-coated plates. FN-ELP induced the osteogenic differentiation by elevating alkaline phosphatase (ALP) and mineralization activity of hMSCs. Furthermore, the osteogenic marker gene expressions of ALP, collagen type I (Col I), osteopontin (OPN), and transcriptional coactivator with a PDZ-binding motif (TAZ) were increased in hMSCs cultured on plates coated with FN-ELP. We reported a novel biomimetic ECM with potential for bone regeneration that promotes the osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Sujin Lee
- Department of Biochemistry & IRIMS, Inha University School of Medicine, Incheon 22212, Korea
| | - Ji-Eun Kim
- Department of Biochemistry & IRIMS, Inha University School of Medicine, Incheon 22212, Korea
| | - Hye-Jin Seo
- Department of Biochemistry & IRIMS, Inha University School of Medicine, Incheon 22212, Korea
| | - Jun-Hyeog Jang
- Department of Biochemistry & IRIMS, Inha University School of Medicine, Incheon 22212, Korea
| |
Collapse
|
23
|
Fibronectin regulates the self-renewal of rabbit limbal epithelial stem cells by stimulating the Wnt11/Fzd7/ROCK non-canonical Wnt pathway. Exp Eye Res 2019; 185:107681. [DOI: 10.1016/j.exer.2019.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/23/2019] [Accepted: 05/26/2019] [Indexed: 12/13/2022]
|
24
|
Tampakakis E, Miyamoto M, Kwon C. In Vitro Generation of Heart Field-specific Cardiac Progenitor Cells. J Vis Exp 2019. [PMID: 31329174 DOI: 10.3791/59826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pluripotent stem cells offer great potential for understanding heart development and disease and for regenerative medicine. While recent advances in developmental cardiology have led to generating cardiac cells from pluripotent stem cells, it is unclear if the two cardiac fields - the first and second heart fields (FHF and SHF) - are induced in pluripotent stem cells systems. To address this, we generated a protocol for in vitro specification and isolation of heart field-specific cardiac progenitor cells. We used embryonic stem cells lines carrying Hcn4-GFP and Tbx1-Cre; Rosa-RFP reporters of the FHF and the SHF, respectively, and live cell immunostaining of the cell membrane protein Cxcr4, a SHF marker. With this approach, we generated progenitor cells which recapitulate the functional properties and transcriptome of their in vivo counterparts. Our protocol can be utilized to study early specification and segregation of the two heart fields and to generate chamber-specific cardiac cells for heart disease modelling. Since this is an in vitro organoid system, it may not provide precise anatomical information. However, this system overcomes the poor accessibility of gastrulation-stage embryos and can be upscaled for high-throughput screens.
Collapse
Affiliation(s)
| | - Matthew Miyamoto
- Division of Cardiology, Department Medicine, Johns Hopkins School of Medicine
| | - Chulan Kwon
- Division of Cardiology, Department Medicine, Johns Hopkins School of Medicine;
| |
Collapse
|
25
|
Hall ML, Ogle BM. Cardiac Extracellular Matrix Modification as a Therapeutic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1098:131-150. [PMID: 30238369 PMCID: PMC6584040 DOI: 10.1007/978-3-319-97421-7_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The cardiac extracellular matrix (cECM) is comprised of proteins and polysaccharides secreted by cardiac cell types, which provide structural and biochemical support to cardiovascular tissue. The roles of cECM proteins and the associated family of cell surface receptor, integrins, have been explored in vivo via the generation of knockout experimental animal models. However, the complexity of tissues makes it difficult to isolate the effects of individual cECM proteins on a particular cell process or disease state. The desire to further dissect the role of cECM has led to the development of a variety of in vitro model systems, which are now being used not only for basic studies but also for testing drug efficacy and toxicity and for generating therapeutic scaffolds. These systems began with 2D coatings of cECM derived from tissue and have developed to include recombinant ECM proteins, ECM fragments, and ECM mimics. Most recently 3D model systems have emerged, made possible by several developing technologies including, and most notably, 3D bioprinting. This chapter will attempt to track the evolution of our understanding of the relationship between cECM and cell behavior from in vivo model to in vitro control systems. We end the chapter with a summary of how basic studies such as these have informed the use of cECM as a direct therapy.
Collapse
Affiliation(s)
- Mikayla L Hall
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Stem Cell Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Lillehei Heart Institute, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
- Institute for Engineering in Medicine, University of Minnesota - Twin Cities, Minneapolis, MN, USA.
| |
Collapse
|
26
|
Ji W, Álvarez Z, Edelbrock AN, Sato K, Stupp SI. Bioactive Nanofibers Induce Neural Transdifferentiation of Human Bone Marrow Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2018; 10:41046-41055. [PMID: 30475573 DOI: 10.1021/acsami.8b13653] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The combination of biomaterials with stem cells is a promising therapeutic strategy to repair traumatic injuries in the central nervous system, and human bone marrow mesenchymal stem cells (BMSCs) offer a clinically translatable option among other possible sources of stem cells. We report here on the use of a supramolecular bioactive material based on a peptide amphiphile (PA), displaying a laminin-mimetic IKVAV sequence to drive neural transdifferentiation of human BMSCs. The IKVAV-PA self-assembles into supramolecular nanofibers that induce neuroectodermal lineage commitment after 1 week, as evidenced by the upregulation of the neural progenitor gene nestin ( NES) and glial fibrillary acidic protein ( GFAP). After 2 weeks, the bioactive IKVAV-PA nanofibers induce significantly higher expression of neuronal markers β-III tubulin (TUJ-1), microtubule-associated protein-2 (MAP-2), and neuronal nuclei (NEUN), as well as the extracellular matrix laminin (LMN). Furthermore, the human BMSCs exposed to the biomaterial reveal a polarized cytoskeletal architecture and a decrease in cellular size, resembling neuron-like cells. We conclude that the investigated supramolecular biomaterial opens the opportunity to transdifferentiate adult human BMSCs into neuronal lineage.
Collapse
Affiliation(s)
- Wei Ji
- Skeletal Biology and Engineering Research Center, Department of Development and Regeneration , KU Leuven , Leuven 3000 , Belgium
| | | | | | | | | |
Collapse
|
27
|
Precardiac organoids form two heart fields via Bmp/Wnt signaling. Nat Commun 2018; 9:3140. [PMID: 30087351 PMCID: PMC6081372 DOI: 10.1038/s41467-018-05604-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 07/17/2018] [Indexed: 12/24/2022] Open
Abstract
The discovery of the first heart field (FHF) and the second heart field (SHF) led us to understand how cardiac lineages and structures arise during development. However, it remains unknown how they are specified. Here, we generate precardiac spheroids with pluripotent stem cells (PSCs) harboring GFP/RFP reporters under the control of FHF/SHF markers, respectively. GFP+ cells and RFP+ cells appear from two distinct areas and develop in a complementary fashion. Transcriptome analysis shows a high degree of similarities with embryonic FHF/SHF cells. Bmp and Wnt are among the most differentially regulated pathways, and gain- and loss-of-function studies reveal that Bmp specifies GFP+ cells and RFP+ cells via the Bmp/Smad pathway and Wnt signaling, respectively. FHF/SHF cells can be isolated without reporters by the surface protein Cxcr4. This study provides novel insights into understanding the specification of two cardiac origins, which can be leveraged for PSC-based modeling of heart field/chamber-specific disease. The heart arises from distinct progenitor cells of both the first and second heart fields (FHF and SHF). Here, the authors generated precardiac organoids from mouse and human pluripotent cells and show that FHF and SHF cells form similarly to their in vivo counterparts in response to BMP and Wnt signalling, respectively.
Collapse
|
28
|
Gibbs BC, Shenje L, Andersen P, Miyamoto M, Kwon C. β1-integrin is a cell-autonomous factor mediating the Numb pathway for cardiac progenitor maintenance. Biochem Biophys Res Commun 2018; 500:256-260. [PMID: 29653101 DOI: 10.1016/j.bbrc.2018.04.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 11/18/2022]
Abstract
Proper control of multipotent/stem cell number and fate is essential for ensuing organ formation during development. β1-integrin, a subfamily of cell surface receptors, has a conserved role in maintenance of multipotent/stem cells, including renal progenitor cells, follicle stem cells, epidermal stem cells and neural stem cells. However, it remains unclear whether β1-integrin has a role in cardiac progenitor cell (CPC) development. Here we show that a mesodermal deletion of β1-integrin decreases Isl1+ cell number in the second pharyngeal arch (PA2), where CPCs undergo renewal and expansion. Mesp1 lineage-specific mosaicism revealed that β1-integrin-deleted Isl1+ cells do not proliferate in the PA2. Consistently, β1-integrin-deleted Isl1+ CPCs failed to expand in vitro, independent of PA2 cells. β1-integrin co-localized and physically associated with Numb, a crucial regulator of CPC renewal and expansion. Importantly, Numb/Numbl-deleted CPCs showed dramatic reduction in β1-integrin levels. These findings suggest that β1-integrin is a key mediator of the Numb pathway in CPC maintenance.
Collapse
Affiliation(s)
- Brian C Gibbs
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lincoln Shenje
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
29
|
Cho GS, Lee DI, Tampakakis E, Murphy S, Andersen P, Uosaki H, Chelko S, Chakir K, Hong I, Seo K, Chen HSV, Chen X, Basso C, Houser SR, Tomaselli GF, O'Rourke B, Judge DP, Kass DA, Kwon C. Neonatal Transplantation Confers Maturation of PSC-Derived Cardiomyocytes Conducive to Modeling Cardiomyopathy. Cell Rep 2017; 18:571-582. [PMID: 28076798 DOI: 10.1016/j.celrep.2016.12.040] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 12/05/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022] Open
Abstract
Pluripotent stem cells (PSCs) offer unprecedented opportunities for disease modeling and personalized medicine. However, PSC-derived cells exhibit fetal-like characteristics and remain immature in a dish. This has emerged as a major obstacle for their application for late-onset diseases. We previously showed that there is a neonatal arrest of long-term cultured PSC-derived cardiomyocytes (PSC-CMs). Here, we demonstrate that PSC-CMs mature into adult CMs when transplanted into neonatal hearts. PSC-CMs became similar to adult CMs in morphology, structure, and function within a month of transplantation into rats. The similarity was further supported by single-cell RNA-sequencing analysis. Moreover, this in vivo maturation allowed patient-derived PSC-CMs to reveal the disease phenotype of arrhythmogenic right ventricular cardiomyopathy, which manifests predominantly in adults. This study lays a foundation for understanding human CM maturation and pathogenesis and can be instrumental in PSC-based modeling of adult heart diseases.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emmanouil Tampakakis
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hideki Uosaki
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephen Chelko
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Khalid Chakir
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kinya Seo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Huei-Sheng Vincent Chen
- Del E. Webb Neuroscience, Aging & Stem Cell Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Xiongwen Chen
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Cristina Basso
- Department of Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140 USA; Department of Cardiac, Thoracic, and Vascular Sciences, University of Padua, Padova, Italy
| | - Steven R Houser
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Gordon F Tomaselli
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daniel P Judge
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
30
|
Jeffery N, Richardson S, Beall C, Harries LW. The species origin of the cellular microenvironment influences markers of beta cell fate and function in EndoC-βH1 cells. Exp Cell Res 2017; 361:284-291. [PMID: 29107069 DOI: 10.1016/j.yexcr.2017.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/30/2022]
Abstract
Interaction between islet cell subtypes and the extracellular matrix influences beta-cell function in mammals. The tissue architecture of rodent islets is very different to that of human islets; cell-to-cell communication and interaction with the extracellular matrix may vary between species. In this work, we have compared the responses of the human EndoC-βH1 cell line to non-human and human-derived growth matrices in terms of growth morphology, gene expression and glucose-stimulated insulin secretion (GSIS). EndoC-βH1 cells demonstrated a greater tendency to form cell clusters when cultured in a human microenvironment and exhibited reduced alpha cell markers at the mRNA level; mean expression difference - 0.23 and - 0.51; p = 0.009 and 0.002 for the Aristaless-related homeobox (ARX) and Glucagon (GCG) genes respectively. No differences were noted in the protein expression of mature beta cell markers such as Pdx1 and NeuroD1 were noted in EndoC-βH1 cells grown in a human microenvironment but cells were however more sensitive to glucose (4.3-fold increase in insulin secretion following glucose challenge compared with a 1.9-fold increase in cells grown in a non-human microenvironment; p = 0.0003). Our data suggests that the tissue origin of the cellular microenvironment has effects on the function of EndoC-βH1 cells in vitro, and the use of a more human-like culture microenvironment may bring benefits in terms of increased physiological relevance.
Collapse
Affiliation(s)
- N Jeffery
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon EX2 5DW, UK
| | - S Richardson
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon EX2 5DW, UK
| | - C Beall
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon EX2 5DW, UK
| | - L W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon EX2 5DW, UK.
| |
Collapse
|
31
|
Trapani V, Bonaldo P, Corallo D. Role of the ECM in notochord formation, function and disease. J Cell Sci 2017; 130:3203-3211. [PMID: 28883093 DOI: 10.1242/jcs.175950] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The notochord is a midline structure common to all chordate animals; it provides mechanical and signaling cues for the developing embryo. In vertebrates, the notochord plays key functions during embryogenesis, being a source of developmental signals that pattern the surrounding tissues. It is composed of a core of vacuolated cells surrounded by an epithelial-like sheath of cells that secrete a thick peri-notochordal basement membrane made of different extracellular matrix (ECM) proteins. The correct deposition and organization of the ECM is essential for proper notochord morphogenesis and function. Work carried out in the past two decades has allowed researchers to dissect the contribution of different ECM components to this embryonic tissue. Here, we will provide an overview of these genetic and mechanistic studies. In particular, we highlight the specific functions of distinct matrix molecules in regulating notochord development and notochord-derived signals. Moreover, we also discuss the involvement of ECM synthesis and its remodeling in the pathogenesis of chordoma, a malignant bone cancer that originates from remnants of notochord remaining after embryogenesis.
Collapse
Affiliation(s)
- Valeria Trapani
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,CRIBI Biotechnology Center, University of Padova, Padova, 35131, Italy
| | - Diana Corallo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy .,Pediatric Research Institute, Città della Speranza, 35127 Padova, Italy
| |
Collapse
|
32
|
Sart S, Bejoy J, Li Y. Characterization of 3D pluripotent stem cell aggregates and the impact of their properties on bioprocessing. Process Biochem 2017. [DOI: 10.1016/j.procbio.2016.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
33
|
Aguado T, Gutiérrez FJ, Aix E, Schneider RP, Giovinazzo G, Blasco MA, Flores I. Telomere Length Defines the Cardiomyocyte Differentiation Potency of Mouse Induced Pluripotent Stem Cells. Stem Cells 2016; 35:362-373. [PMID: 27612935 DOI: 10.1002/stem.2497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 12/21/2022]
Abstract
Induced pluripotent stem cells (iPSCs) can be differentiated in vitro and in vivo to all cardiovascular lineages and are therefore a promising cell source for cardiac regenerative therapy. However, iPSC lines do not all differentiate into cardiomyocytes (CMs) with the same efficiency. Here, we show that telomerase-competent iPSCs with relatively long telomeres and high expression of the shelterin-complex protein TRF1 (iPSChighT ) differentiate sooner and more efficiently into CMs than those with relatively short telomeres and low TRF1 expression (iPSClowT ). Ascorbic acid, an enhancer of cardiomyocyte differentiation, further increases the cardiomyocyte yield from iPSChighT but does not rescue the cardiomyogenic potential of iPSClowT . Interestingly, although iPSCslowT differentiate very poorly to the mesoderm and endoderm lineages, they differentiate very efficiently to the ectoderm lineage, indicating that cell fate can be determined by in vitro selection of iPSCs with different telomere content. Our findings highlight the importance of selecting iPSCs with ample telomere reserves in order to generate high numbers of CMs in a fast, reliable, and efficient way. Stem Cells 2017;35:362-373.
Collapse
Affiliation(s)
- Tania Aguado
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Francisco J Gutiérrez
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Esther Aix
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Ralph P Schneider
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - María A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ignacio Flores
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| |
Collapse
|
34
|
Segade F, Cota C, Famiglietti A, Cha A, Davidson B. Fibronectin contributes to notochord intercalation in the invertebrate chordate, Ciona intestinalis. EvoDevo 2016; 7:21. [PMID: 27583126 PMCID: PMC5006582 DOI: 10.1186/s13227-016-0056-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/13/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genomic analysis has upended chordate phylogeny, placing the tunicates as the sister group to the vertebrates. This taxonomic rearrangement raises questions about the emergence of a tunicate/vertebrate ancestor. RESULTS Characterization of developmental genes uniquely shared by tunicates and vertebrates is one promising approach for deciphering developmental shifts underlying acquisition of novel, ancestral traits. The matrix glycoprotein Fibronectin (FN) has long been considered a vertebrate-specific gene, playing a major instructive role in vertebrate embryonic development. However, the recent computational prediction of an orthologous "vertebrate-like" Fn gene in the genome of a tunicate, Ciona savignyi, challenges this viewpoint suggesting that Fn may have arisen in the shared tunicate/vertebrate ancestor. Here we verify the presence of a tunicate Fn ortholog. Transgenic reporter analysis was used to characterize a Ciona Fn enhancer driving expression in the notochord. Targeted knockdown in the notochord lineage indicates that FN is required for proper convergent extension. CONCLUSIONS These findings suggest that acquisition of Fn was associated with altered notochord morphogenesis in the vertebrate/tunicate ancestor.
Collapse
Affiliation(s)
- Fernando Segade
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104 USA
| | - Christina Cota
- Department of Biology, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| | - Amber Famiglietti
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Anna Cha
- Department of Systems Biology, Harvard Medical School, Boston, MA USA
| | - Brad Davidson
- Department of Biology, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| |
Collapse
|
35
|
Boxman J, Sagy N, Achanta S, Vadigepalli R, Nachman I. Integrated live imaging and molecular profiling of embryoid bodies reveals a synchronized progression of early differentiation. Sci Rep 2016; 6:31623. [PMID: 27530599 PMCID: PMC4987683 DOI: 10.1038/srep31623] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/22/2016] [Indexed: 01/23/2023] Open
Abstract
Embryonic stem cells can spontaneously differentiate into cell types of all germ layers within embryoid bodies (EBs) in a highly variable manner. Whether there exists an intrinsic differentiation program common to all EBs is unknown. Here, we present a novel combination of high-throughput live two-photon imaging and gene expression profiling to study early differentiation dynamics spontaneously occurring within developing EBs. Onset timing of Brachyury-GFP was highly variable across EBs, while the spatial patterns as well as the dynamics of mesendodermal progression following onset were remarkably similar. We therefore defined a 'developmental clock' using the Brachyury-GFP signal onset timing. Mapping snapshot gene expression measurements to this clock revealed their temporal trends, indicating that loss of pluripotency, formation of primitive streak and mesodermal lineage progression are synchronized in EBs. Exogenous activation of Wnt or BMP signaling accelerated the intrinsic clock. CHIR down-regulated Wnt3, allowing insights into dependency mechanisms between canonical Wnt signaling and multiple genes. Our findings reveal a developmental clock characteristic of an early differentiation program common to all EBs, further establishing them as an in vitro developmental model.
Collapse
Affiliation(s)
- Jonathan Boxman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| | - Naor Sagy
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| | - Sirisha Achanta
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rajanikanth Vadigepalli
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Iftach Nachman
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Israel
| |
Collapse
|
36
|
Abstract
Stem cells hold great promise in treating many diseases either through promoting endogenous cell repair or through direct cell transplants. In order to maximize their potential, understanding the fundamental signals and mechanisms that regulate their behavior is essential. The extracellular matrix (ECM) is one such component involved in mediating stem cell fate. Recent studies have made significant progress in understanding stem cell-ECM interactions. Technological developments have provided greater clarity in how cells may sense and respond to the ECM, in particular the physical properties of the matrix. This review summarizes recent developments, providing illustrative examples of the different modes with which the ECM controls both embryonic and adult stem cell behavior.
Collapse
|
37
|
Huang G, Zhang F, Ye Q, Wang H. The circadian clock regulates autophagy directly through the nuclear hormone receptor Nr1d1/Rev-erbα and indirectly via Cebpb/(C/ebpβ) in zebrafish. Autophagy 2016; 12:1292-309. [PMID: 27171500 PMCID: PMC4968235 DOI: 10.1080/15548627.2016.1183843] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Autophagy is a highly conserved intracellular degradation system, and recently was shown to display circadian rhythms in mice. The mechanisms underlying circadian regulation of autophagy, however, are still unclear. Here, we observed that numbers of autophagosomes and autolysosomes exhibit daily rhythms in the zebrafish liver, and cebpb/(c/ebpβ) and various autophagy genes are rhythmically expressed in zebrafish larvae but significantly upregulated in per1b and TALEN-generated nr1d1/rev-erbα mutant fish, indicating that both Per1b and Nr1d1 play critical roles in autophagy rhythms. Luciferase reporter and ChIP assays show that the circadian clock directly regulates autophagy genes through Nr1d1, and also regulates transcription of cebpb through Per1b. We also found that fasting leads to altered expression of both circadian clock genes and autophagy genes in zebrafish adult peripheral organs. Further, transcriptome analysis reveals multiple functions of Nr1d1 in zebrafish. Taken together, these findings provide evidence for how the circadian clock regulates autophagy, imply that nutritional signaling affects both circadian regulation and autophagy activities in peripheral organs, and shed light on how circadian gene mutations act through autophagy to contribute to common metabolic diseases such as obesity.
Collapse
Affiliation(s)
- Guodong Huang
- a Center for Circadian Clocks , Soochow University ; Suzhou , Jiangsu , China.,b School of Biology & Basic Medical Sciences , Medical College, Soochow University , Suzhou , Jiangsu , China
| | - Fanmiao Zhang
- a Center for Circadian Clocks , Soochow University ; Suzhou , Jiangsu , China.,b School of Biology & Basic Medical Sciences , Medical College, Soochow University , Suzhou , Jiangsu , China
| | - Qiang Ye
- a Center for Circadian Clocks , Soochow University ; Suzhou , Jiangsu , China.,b School of Biology & Basic Medical Sciences , Medical College, Soochow University , Suzhou , Jiangsu , China
| | - Han Wang
- a Center for Circadian Clocks , Soochow University ; Suzhou , Jiangsu , China.,b School of Biology & Basic Medical Sciences , Medical College, Soochow University , Suzhou , Jiangsu , China
| |
Collapse
|
38
|
Jenkins MH, Alrowaished SS, Goody MF, Crawford BD, Henry CA. Laminin and Matrix metalloproteinase 11 regulate Fibronectin levels in the zebrafish myotendinous junction. Skelet Muscle 2016; 6:18. [PMID: 27141287 PMCID: PMC4852425 DOI: 10.1186/s13395-016-0089-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 03/31/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Remodeling of the extracellular matrix (ECM) regulates cell adhesion as well as signaling between cells and their microenvironment. Despite the importance of tightly regulated ECM remodeling for normal muscle development and function, mechanisms underlying ECM remodeling in vivo remain elusive. One excellent paradigm in which to study ECM remodeling in vivo is morphogenesis of the myotendinous junction (MTJ) during zebrafish skeletal muscle development. During MTJ development, there are dramatic shifts in the primary components comprising the MTJ matrix. One such shift involves the replacement of Fibronectin (Fn)-rich matrix, which is essential for both somite and early muscle development, with laminin-rich matrix essential for normal function of the myotome. Here, we investigate the mechanism underlying this transition. RESULTS We show that laminin polymerization indirectly promotes Fn downregulation at the MTJ, via a matrix metalloproteinase 11 (Mmp11)-dependent mechanism. Laminin deposition and organization is required for localization of Mmp11 to the MTJ, where Mmp11 is both necessary and sufficient for Fn downregulation in vivo. Furthermore, reduction of residual Mmp11 in laminin mutants promotes a Fn-rich MTJ that partially rescues skeletal muscle architecture. CONCLUSIONS These results identify a mechanism for Fn downregulation at the MTJ, highlight crosstalk between laminin and Fn, and identify a new in vivo function for Mmp11. Taken together, our data demonstrate a novel signaling pathway mediating Fn downregulation. Our data revealing new regulatory mechanisms that guide ECM remodeling during morphogenesis in vivo may inform pathological conditions in which Fn is dysregulated.
Collapse
Affiliation(s)
- Molly H Jenkins
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469 USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469 USA.,Present Address: Minerva Biotechnologies, Waltham, MA 02451 USA
| | - Sarah S Alrowaished
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469 USA
| | - Michelle F Goody
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469 USA
| | - Bryan D Crawford
- Department of Biology, University of New Brunswick, Fredericton, NB Canada
| | - Clarissa A Henry
- School of Biology and Ecology, University of Maine, 217 Hitchner Hall, Orono, ME 04469 USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469 USA
| |
Collapse
|
39
|
Malta DFB, Reticker-Flynn NE, da Silva CL, Cabral JMS, Fleming HE, Zaret KS, Bhatia SN, Underhill GH. Extracellular matrix microarrays to study inductive signaling for endoderm specification. Acta Biomater 2016; 34:30-40. [PMID: 26883775 DOI: 10.1016/j.actbio.2016.02.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/29/2016] [Accepted: 02/10/2016] [Indexed: 12/31/2022]
Abstract
During tissue development, stem and progenitor cells are faced with fate decisions coordinated by microenvironmental cues. Although insights have been gained from in vitro and in vivo studies, the role of the microenvironment remains poorly understood due to the inability to systematically explore combinations of stimuli at a large scale. To overcome such restrictions, we implemented an extracellular matrix (ECM) array platform that facilitates the study of 741 distinct combinations of 38 different ECM components in a systematic, unbiased and high-throughput manner. Using embryonic stem cells as a model system, we derived definitive endoderm progenitors and applied them to the array platform to study the influence of ECM, including the interactions of ECM with growth factor signaling, on the specification of definitive endoderm cells towards the liver and pancreas fates. We identified ECM combinations that influence endoderm fate decisions towards these lineages, and demonstrated the utility of this platform for studying ECM-mediated modifications to signal activation during liver specification. In particular, defined combinations of fibronectin and laminin isoforms, as well as combinations of distinct collagen subtypes, were shown to influence SMAD pathway activation and the degree of hepatic differentiation. Overall, our systematic high-throughput approach suggests that ECM components of the microenvironment have modulatory effects on endoderm differentiation, including effects on lineage fate choice and cell adhesion and survival during the differentiation process. This platform represents a robust tool for analyzing effects of ECM composition towards the continued improvement of stem cell differentiation protocols and further elucidation of tissue development processes. STATEMENT OF SIGNIFICANCE Cellular microarrays can provide the capability to perform high-throughput investigations into the role of microenvironmental signals in a variety of cell functions. This study demonstrates the utility of a high-throughput cellular microarray approach for analyzing the effects of extracellular matrix (ECM) in liver and pancreas differentiation of endoderm progenitor cells. Despite an appreciation that ECM is likely involved in these processes, the influence of ECM, particularly combinations of matrix proteins, had not been systematically explored. In addition to the identification of relevant ECM compositions, this study illustrates the capability of the cellular microarray platform to be integrated with a diverse range of cell fate measurements, which could be broadly applied towards the investigation of cell fate regulation in other tissue development and disease contexts.
Collapse
Affiliation(s)
- D F Braga Malta
- Massachusetts Institute of Technology, Cambridge, MA, United States; Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | | | - C L da Silva
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - J M S Cabral
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - H E Fleming
- Massachusetts Institute of Technology, Cambridge, MA, United States
| | - K S Zaret
- University of Pennsylvania, Philadelphia, PA, United States
| | - S N Bhatia
- Massachusetts Institute of Technology, Cambridge, MA, United States; The Howard Hughes Medical Institute, Cambridge, MA, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 021392, United States; Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - G H Underhill
- University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
40
|
Toya SP, Wary KK, Mittal M, Li F, Toth PT, Park C, Rehman J, Malik AB. Integrin α6β1 Expressed in ESCs Instructs the Differentiation to Endothelial Cells. Stem Cells 2016; 33:1719-29. [PMID: 25693840 DOI: 10.1002/stem.1974] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/25/2014] [Accepted: 01/14/2015] [Indexed: 12/19/2022]
Abstract
Adhesion of embryonic stem cells (ESCs) to the extracellular matrix may influence differentiation potential and cell fate decisions. Here, we investigated the inductive role of binding of integrin α6β1 expressed in mouse (m)ESCs to laminin-1 (LN1) in mediating the differentiation of ESCs to endothelial cells (ECs). We observed that α6β1 binding to LN1 was required for differentiation to ECs. α6β1 functioned by recruiting the adaptor tetraspanin protein CD151, which activated FAK and Akt signaling and mediated the EC lineage-specifying transcription factor Er71. In contrast, association of the ESC-expressed α3β1, another highly expressed LN1 binding integrin, with CD151, prevented α6β1-mediated differentiation. CD151 thus functioned as a bifurcation router to direct ESCs toward ECs when α6β1 associated with CD151, or prevented transition to ECs when α3β1 associated with CD151. These observations were recapitulated in mice in which α6 integrin or CD151 knockdown reduced the expression of Er71-regulated angiogenesis genes and development of blood vessels. Thus, interaction of α6β1 in ESCs with LN1 activates α6β1/CD151 signaling which programs ESCs toward the EC lineage fate.
Collapse
Affiliation(s)
- Sophie P Toya
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Manish Mittal
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Fei Li
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Peter T Toth
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Changwon Park
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, USA.,The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
41
|
den Hartogh SC, Wolstencroft K, Mummery CL, Passier R. A comprehensive gene expression analysis at sequential stages of in vitro cardiac differentiation from isolated MESP1-expressing-mesoderm progenitors. Sci Rep 2016; 6:19386. [PMID: 26783251 PMCID: PMC4726039 DOI: 10.1038/srep19386] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/02/2015] [Indexed: 01/03/2023] Open
Abstract
In vitro cardiac differentiation of human pluripotent stem cells (hPSCs) closely recapitulates in vivo embryonic heart development, and therefore, provides an excellent model to study human cardiac development. We recently generated the dual cardiac fluorescent reporter MESP1mCherry/wNKX2-5eGFP/w line in human embryonic stem cells (hESCs), allowing the visualization of pre-cardiac MESP1+ mesoderm and their further commitment towards the cardiac lineage, marked by activation of the cardiac transcription factor NKX2-5. Here, we performed a comprehensive whole genome based transcriptome analysis of MESP1-mCherry derived cardiac-committed cells. In addition to previously described cardiac-inducing signalling pathways, we identified novel transcriptional and signalling networks indicated by transient activation and interactive network analysis. Furthermore, we found a highly dynamic regulation of extracellular matrix components, suggesting the importance to create a versatile niche, adjusting to various stages of cardiac differentiation. Finally, we identified cell surface markers for cardiac progenitors, such as the Leucine-rich repeat-containing G-protein coupled receptor 4 (LGR4), belonging to the same subfamily of LGR5, and LGR6, established tissue/cancer stem cells markers. We provide a comprehensive gene expression analysis of cardiac derivatives from pre-cardiac MESP1-progenitors that will contribute to a better understanding of the key regulators, pathways and markers involved in human cardiac differentiation and development.
Collapse
Affiliation(s)
- Sabine C den Hartogh
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Katherine Wolstencroft
- Leiden Institute of Advanced Computer Science Leiden Institute of Advanced Computer Science, Leiden University, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands.,Department of Applied Stem cell Technologies. MIRA Institute for Biomedical Technology and Technical Medicine. University of Twente, P.O.Box 217, Enschede, The Netherlands
| |
Collapse
|
42
|
Andersen P, Kwon C. Ex Vivo Culture of Pharyngeal Arches to Study Heart and Muscle Progenitors and Their Niche. J Vis Exp 2015:e52876. [PMID: 26274057 DOI: 10.3791/52876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The pharyngeal mesoderm of developing embryos contributes to broad regions of head and heart musculature. We have developed a novel method to study head and heart progenitor cell development with pharyngeal arches (also known as branchial arches) ex vivo. Using this method, we have recently described that the second pharyngeal arch contains self-renewing heart progenitors and serves as a microenvironment for expansion of the progenitors during mouse heart development. The progenitor cells remain undifferentiated and expansive inside the arch, but quickly become functional cardiomyocytes as they migrate out of the arch. We also reported that first pharyngeal arch contains muscle progenitors giving rise to myotubes after leaving the arch. Here, we demonstrate the procedure for the dissection and ex vivo culture of first and second pharyngeal arches from developing mouse embryos. The method enables one to study head and heart progenitor/muscle development, including cardiomyocyte and myotube formation in detail ex vivo.
Collapse
Affiliation(s)
- Peter Andersen
- Division of Cardiology, Institute for Cell Engineering, Johns Hopkins University School of Medicine
| | - Chulan Kwon
- Division of Cardiology, Institute for Cell Engineering, Johns Hopkins University School of Medicine;
| |
Collapse
|
43
|
Viotti M, Foley AC, Hadjantonakis AK. Gutsy moves in mice: cellular and molecular dynamics of endoderm morphogenesis. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0547. [PMID: 25349455 DOI: 10.1098/rstb.2013.0547] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite the importance of the gut and its accessory organs, our understanding of early endoderm development is still incomplete. Traditionally, endoderm has been difficult to study because of its small size and relative fragility. However, recent advances in live cell imaging technologies have dramatically expanded our understanding of this tissue, adding a new appreciation for the complex molecular and morphogenetic processes that mediate gut formation. Several spatially and molecularly distinct subpopulations have been shown to exist within the endoderm before the onset of gastrulation. Here, we review findings that have uncovered complex cell movements within the endodermal layer, before and during gastrulation, leading to the conclusion that cells from primitive endoderm contribute descendants directly to gut.
Collapse
Affiliation(s)
- Manuel Viotti
- Genentech Incorporated, South San Francisco, CA 94080, USA
| | - Ann C Foley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA
| | | |
Collapse
|
44
|
Maartens AP, Brown NH. Anchors and signals: the diverse roles of integrins in development. Curr Top Dev Biol 2015; 112:233-72. [PMID: 25733142 DOI: 10.1016/bs.ctdb.2014.11.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Integrins mediate cell adhesion by providing a link between the actin cytoskeleton and the extracellular matrix. As well as acting to anchor cells, integrin adhesions provide sensory input via mechanotransduction and synergism with signaling pathways, and provide the cell with the conditions necessary for differentiation in a permissive manner. In this review, we explore how integrins contribute to development, and what this tells us about how they work. From a signaling perspective, the influence of integrins on cell viability and fate is muted in a developmental context as compared to cell culture. Integrin phenotypes tend to arise from a failure of normally specified cells to create tissues properly, due to defective adhesion. The diversity of integrin functions in development shows how cell adhesion is continuously adjusted, both within and between animals, to fit developmental purpose.
Collapse
Affiliation(s)
- Aidan P Maartens
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas H Brown
- Department of Physiology, Development and Neuroscience, The Gurdon Institute, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
45
|
Den Hartogh SC, Schreurs C, Monshouwer-Kloots JJ, Davis RP, Elliott DA, Mummery CL, Passier R. Dual ReporterMESP1mCherry/w-NKX2-5eGFP/whESCs Enable Studying Early Human Cardiac Differentiation. Stem Cells 2014; 33:56-67. [DOI: 10.1002/stem.1842] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 08/15/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Sabine C. Den Hartogh
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | - Chantal Schreurs
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | | | - Richard P. Davis
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | - David A. Elliott
- Murdoch Childrens Research Institute, Royal Children's Hospital; Parkville Melbourne Victoria Australia
| | - Christine L. Mummery
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology; Leiden University Medical Centre; Leiden The Netherlands
| |
Collapse
|
46
|
Cho GS, Fernandez L, Kwon C. Regenerative medicine for the heart: perspectives on stem-cell therapy. Antioxid Redox Signal 2014; 21:2018-31. [PMID: 25133793 PMCID: PMC4208610 DOI: 10.1089/ars.2014.6063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Despite decades of progress in cardiovascular biology and medicine, heart disease remains the leading cause of death, and there is no cure for the failing heart. Since heart failure is mostly caused by loss or dysfunction of cardiomyocytes (CMs), replacing dead or damaged CMs with new CMs might be an ideal way to reverse the disease. However, the adult heart is composed mainly of terminally differentiated CMs that have no significant self-regeneration capacity. RECENT ADVANCES Stem cells have tremendous regenerative potential and, thus, current cardiac regenerative research has focused on developing stem cell sources to repair damaged myocardium. CRITICAL ISSUES In this review, we examine the potential sources of cells that could be used for heart therapies, including embryonic stem cells and induced pluripotent stem cells, as well as alternative methods for activating the endogenous regenerative mechanisms of the heart via transdifferentiation and cell reprogramming. We also discuss the current state of knowledge of cell purification, delivery, and retention. FUTURE DIRECTIONS Efforts are underway to improve the current stem cell strategies and methodologies, which will accelerate the development of innovative stem-cell therapies for heart regeneration.
Collapse
Affiliation(s)
- Gun-Sik Cho
- Division of Cardiology, Department of Medicine, Institute for Cell Engineering, Johns Hopkins University , Baltimore, Maryland
| | | | | |
Collapse
|
47
|
Kim SG, Lee DS, Lee S, Jang JH. Osteocalcin/fibronectin-functionalized collagen matrices for bone tissue engineering. J Biomed Mater Res A 2014; 103:2133-40. [PMID: 25346429 DOI: 10.1002/jbm.a.35351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 09/29/2014] [Accepted: 10/09/2014] [Indexed: 02/04/2023]
Abstract
Collagen is the most abundant protein found in the extracellular matrix and is widely used to build scaffolds for biomedical applications which are the result of its biocompatibility and biodegradability. In the present study, we constructed a rhOCN/FNIII9-10 fusion protein and rhOCN/FNIII9-10-functionalized collagen matrices and investigated the potential value for bone tissue engineering. In vitro studies carried out with preosteoblastic MC3T3-E1 cells showed that rhOCN/FNIII9-10 fusion protein promoted cell adhesion and the mRNA levels of osteogenic markers including osteocalcin, runt-related transcription factor 2, alkaline phosphatase (ALP), and collagen type I. In addition, rhOCN/FNIII9-10-functionalized collagen matrices showed significant induction of the ALP activity more than rhFNIII9-10-functionalized collagen matrices or collagen matrices alone. These results suggested that rhOCN/FNIII9-10-functionalized collagen matrices have potential for bone tissue engineering.
Collapse
Affiliation(s)
- S G Kim
- Department of Biochemistry, Inha University School of Medicine, Incheon, 400-712, Republic of Korea
| | | | | | | |
Collapse
|
48
|
Kelly RG, Buckingham ME, Moorman AF. Heart fields and cardiac morphogenesis. Cold Spring Harb Perspect Med 2014; 4:4/10/a015750. [PMID: 25274757 DOI: 10.1101/cshperspect.a015750] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this review, we focus on two important steps in the formation of the embryonic heart: (i) the progressive addition of late differentiating progenitor cells from the second heart field that drives heart tube extension during looping morphogenesis, and (ii) the emergence of patterned proliferation within the embryonic myocardium that generates distinct cardiac chambers. During the transition between these steps, the major site of proliferation switches from progenitor cells outside the early heart to proliferation within the embryonic myocardium. The second heart field and ballooning morphogenesis concepts have major repercussions on our understanding of human heart development and disease. In particular, they provide a framework to dissect the origin of congenital heart defects and the regulation of myocardial proliferation and differentiation of relevance for cardiac repair.
Collapse
Affiliation(s)
- Robert G Kelly
- Aix Marseille University, CNRS, IBDM UMR 7288, 13288 Marseilles, France
| | - Margaret E Buckingham
- Department of Developmental and Stem Cell Biology, URA CNRS 2578, Pasteur Institute, 75015 Paris, France
| | - Antoon F Moorman
- Department of Anatomy, Embryology & Physiology, Academic Medical Centre, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
49
|
Brachyury cooperates with Wnt/β-catenin signalling to elicit primitive-streak-like behaviour in differentiating mouse embryonic stem cells. BMC Biol 2014; 12:63. [PMID: 25115237 PMCID: PMC4171571 DOI: 10.1186/s12915-014-0063-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/25/2014] [Indexed: 12/13/2022] Open
Abstract
Background The formation of the primitive streak is the first visible sign of gastrulation, the process by which the three germ layers are formed from a single epithelium during early development. Embryonic stem cells (ESCs) provide a good system for understanding the molecular and cellular events associated with these processes. Previous work, both in embryos and in culture, has shown how converging signals from both nodal/TGFβR and Wnt/β-catenin signalling pathways specify cells to adopt a primitive-streak-like fate and direct them to undertake an epithelial-to-mesenchymal transition (EMT). However, many of these approaches have relied on genetic analyses without taking into account the temporal progression of events within single cells. In addition, it is still unclear to what extent events in the embryo are able to be reproduced in culture. Results Here, we combine flow cytometry and a quantitative live single-cell imaging approach to demonstrate how the controlled differentiation of mouse ESCs towards a primitive streak fate in culture results in cells displaying many of the characteristics observed during early mouse development including transient brachyury expression, EMT and increased motility. We also find that the EMT initiates the process, and this is both fuelled and terminated by the action of brachyury, whose expression is dependent on the EMT and β-catenin activity. Conclusions As a consequence of our analysis, we propose that a major output of brachyury expression is in controlling the velocity of the cells that are transiting out of the primitive streak. Electronic supplementary material The online version of this article (doi:10.1186/s12915-014-0063-7) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
Nakayama KH, Hou L, Huang NF. Role of extracellular matrix signaling cues in modulating cell fate commitment for cardiovascular tissue engineering. Adv Healthc Mater 2014; 3:628-41. [PMID: 24443420 PMCID: PMC4031033 DOI: 10.1002/adhm.201300620] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/09/2013] [Indexed: 01/01/2023]
Abstract
It is generally agreed that engineered cardiovascular tissues require cellular interactions with the local milieu. Within the microenvironment, the extracellular matrix (ECM) is an important support structure that provides dynamic signaling cues in part through its chemical, physical, and mechanical properties. In response to ECM factors, cells activate biochemical and mechanotransduction pathways that modulate their survival, growth, migration, differentiation, and function. This Review describes the role of ECM chemical composition, spatial patterning, and mechanical stimulation in the specification of cardiovascular lineages, with a focus on stem cell differentiation, direct transdifferentiation, and endothelial-to-mesenchymal transition. The translational application of ECMs is discussed in the context of cardiovascular tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Karina H Nakayama
- Department of Cardiothoracic Surgery, Stanford University, 300 Pasteur Dr, Stanford, CA, 94305, USA; Cardiovascular Institute, Stanford University, 265 Campus Drive, G1120, MC-5454, Stanford, CA, 94305, USA; Veterans Affairs Palo Alto Health Care System, 3801 Miranda Avenue, Mail Code 153, Palo Alto, CA, 94304 60031l, 650-493-5000, USA
| | | | | |
Collapse
|