1
|
Eintracht J, Owen N, Harding P, Moosajee M. Disruption of common ocular developmental pathways in patient-derived optic vesicle models of microphthalmia. Stem Cell Reports 2024; 19:839-858. [PMID: 38821055 PMCID: PMC11390689 DOI: 10.1016/j.stemcr.2024.05.001] [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: 09/13/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
Genetic perturbations influencing early eye development can result in microphthalmia, anophthalmia, and coloboma (MAC). Over 100 genes are associated with MAC, but little is known about common disease mechanisms. In this study, we generated induced pluripotent stem cell (iPSC)-derived optic vesicles (OVs) from two unrelated microphthalmia patients and healthy controls. At day 20, 35, and 50, microphthalmia patient OV diameters were significantly smaller, recapitulating the "small eye" phenotype. RNA sequencing (RNA-seq) analysis revealed upregulation of apoptosis-initiating and extracellular matrix (ECM) genes at day 20 and 35. Western blot and immunohistochemistry revealed increased expression of lumican, nidogen, and collagen type IV, suggesting ECM overproduction. Increased apoptosis was observed in microphthalmia OVs with reduced phospho-histone 3 (pH3+) cells confirming decreased cell proliferation at day 35. Pharmacological inhibition of caspase-8 activity with Z-IETD-FMK decreased apoptosis in one patient model, highlighting a potential therapeutic approach. These data reveal shared pathophysiological mechanisms contributing to a microphthalmia phenotype.
Collapse
Affiliation(s)
| | | | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; Moorfields Eye Hospital NHS Foundation Trust, London EC1V 9EL, UK; Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
2
|
Rasys AM, Wegerski A, Trainor PA, Hufnagel RB, Menke DB, Lauderdale JD. Dynamic changes in ocular shape during human development and its implications for retina fovea formation. Bioessays 2024; 46:e2300054. [PMID: 38037292 DOI: 10.1002/bies.202300054] [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: 03/27/2023] [Revised: 10/10/2023] [Accepted: 10/24/2023] [Indexed: 12/02/2023]
Abstract
The human fovea is known for its distinctive pit-like appearance, which results from the displacement of retinal layers superficial to the photoreceptors cells. The photoreceptors are found at high density within the foveal region but not the surrounding retina. Efforts to elucidate the mechanisms responsible for these unique features have ruled out cell death as an explanation for pit formation and changes in cell proliferation as the cause of increased photoreceptor density. These findings have led to speculation that mechanical forces acting within and on the retina during development underly the formation of foveal architecture. Here we review eye morphogenesis and retinal remodeling in human embryonic development. Our meta-analysis of the literature suggests that fovea formation is a protracted process involving dynamic changes in ocular shape that start early and continue throughout most of human embryonic development. From these observations, we propose a new model for fovea development.
Collapse
Affiliation(s)
- Ashley M Rasys
- Department of Cellular Biology, The University of Georgia, Athens, Georgia, USA
| | - Andrew Wegerski
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
- Department of Anatomy & Cell Biology, The University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Robert B Hufnagel
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Douglas B Menke
- Department of Genetics, The University of Georgia, Athens, Georgia, USA
| | - James D Lauderdale
- Department of Cellular Biology, The University of Georgia, Athens, Georgia, USA
- Neuroscience Division of the Biomedical and Health Sciences Institute, The University of Georgia, Athens, Georgia, USA
| |
Collapse
|
3
|
Sokolova N, Zilova L, Wittbrodt J. Unravelling the link between embryogenesis and adult stem cell potential in the ciliary marginal zone: A comparative study between mammals and teleost fish. Cells Dev 2023; 174:203848. [PMID: 37172718 DOI: 10.1016/j.cdev.2023.203848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
The discovery and study of adult stem cells have revolutionized regenerative medicine by offering new opportunities for treating various medical conditions. Anamniote stem cells, which retain their full proliferative capacity and full differentiation range throughout their lifetime, harbour a greater potential compared to mammalian adult stem cells, which only exhibit limited stem cell potential. Therefore, understanding the mechanisms underlying these differences is of significant interest. In this review, we examine the similarities and differences of adult retinal stem cells in anamniotes and mammals, from their embryonic stages in the optic vesicle to their residence in the postembryonic retinal stem cell niche, the ciliary marginal zone located in the retinal periphery. In anamniotes, developing precursors of retinal stem cells are exposed to various environmental cues during their migration in the complex morphogenetic remodelling of the optic vesicle to the optic cup. In contrast, their mammalian counterparts in the retinal periphery are primarily instructed by neighbouring tissues once they are in place. We explore the distinct modes of optic cup morphogenesis in mammals and teleost fish and highlight molecular mechanisms governing morphogenesis and stem cells instruction. The review concludes with the molecular mechanisms of ciliary marginal zone formation and offers a perspective on the impact of comparative single cell transcriptomic studies to reveal the evolutionary similarities and differences.
Collapse
Affiliation(s)
- Natalia Sokolova
- Centre for Organismal Studies Heidelberg, Germany; Heidelberg Biosciences International Graduate School, Germany
| | - Lucie Zilova
- Centre for Organismal Studies Heidelberg, Germany.
| | | |
Collapse
|
4
|
Caianiello S, Bertolaso M, Militello G. Thinking in 3 dimensions: philosophies of the microenvironment in organoids and organs-on-chip. HISTORY AND PHILOSOPHY OF THE LIFE SCIENCES 2023; 45:14. [PMID: 36949354 DOI: 10.1007/s40656-023-00560-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Organoids and organs-on-a-chip are currently the two major families of 3D advanced organotypic in vitro culture systems, aimed at reconstituting miniaturized models of physiological and pathological states of human organs. Both share the tenets of the so-called "three-dimensional thinking", a Systems Physiology approach focused on recapitulating the dynamic interactions between cells and their microenvironment. We first review the arguments underlying the "paradigm shift" toward three-dimensional thinking in the in vitro culture community. Then, through a historically informed account of the technical affordances and the epistemic commitments of these two approaches, we highlight how they embody two distinct experimental cultures. We finally argue that the current systematic effort for their integration requires not only innovative "synergistic" engineering solutions, but also conceptual integration between different perspectives on biological causality.
Collapse
Affiliation(s)
- Silvia Caianiello
- Institute for the History of Philosophy and Science in the Modern Age (ISPF), Consiglio Nazionale delle Ricerche, Naples, Italy.
- Stazione Zoologica "Anton Dohrn", Naples, Italy.
| | - Marta Bertolaso
- Faculty of Science and Technology for Sustainable Development and One Health, Universitá Campus Bio-Medico di Roma, Rome, Italy
| | - Guglielmo Militello
- Faculty of Science and Technology for Sustainable Development and One Health, Universitá Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
5
|
Cardozo MJ, Sánchez-Bustamante E, Bovolenta P. Optic cup morphogenesis across species and related inborn human eye defects. Development 2023; 150:dev200399. [PMID: 36714981 PMCID: PMC10110496 DOI: 10.1242/dev.200399] [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] [Indexed: 01/31/2023]
Abstract
The vertebrate eye is shaped as a cup, a conformation that optimizes vision and is acquired early in development through a process known as optic cup morphogenesis. Imaging living, transparent teleost embryos and mammalian stem cell-derived organoids has provided insights into the rearrangements that eye progenitors undergo to adopt such a shape. Molecular and pharmacological interference with these rearrangements has further identified the underlying molecular machineries and the physical forces involved in this morphogenetic process. In this Review, we summarize the resulting scenarios and proposed models that include common and species-specific events. We further discuss how these studies and those in environmentally adapted blind species may shed light on human inborn eye malformations that result from failures in optic cup morphogenesis, including microphthalmia, anophthalmia and coloboma.
Collapse
Affiliation(s)
- Marcos J. Cardozo
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Elena Sánchez-Bustamante
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| |
Collapse
|
6
|
Shen W, Shao A, Zhou W, Lou L, Grzybowski A, Jin K, Ye J. Retinogenesis in a Dish: Bibliometric Analysis and Visualization of Retinal Organoids From 2011 to 2022. Cell Transplant 2023; 32:9636897231214321. [PMID: 38044501 PMCID: PMC10695087 DOI: 10.1177/09636897231214321] [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: 08/20/2023] [Revised: 10/12/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Retinal organoid (RO) is the three-dimensional (3D) retinal culture derived from pluripotent or embryonic stem cells which recapitulates organ functions, which was a revolutionary milestone in stem cell technology. The purpose of this study is to explore the hotspots and future directions on ROs, as well as to better understand the fields of greatest research opportunities. Eligible publications related to RO from 2011 to 2022 were acquired from the Web of Science (WoS) Core Collection database. Bibliometric analysis was performed by using software including VOSviewer, CiteSpace, and ArcGIS. A total of 520 articles were included, and the number of annual publications showed a rapid increase with an average rate of 40.86%. The United States published the most articles (241/520, 46.35%) with highest total citation frequencies (5,344). University College London (UK) contributed the largest publication output (40/520, 7.69%) and received highest total citation frequencies. Investigative Ophthalmology & Visual Science was the most productive journal with 129 articles. Majlinda Lako contributed the most research with 32 articles, while Olivier Goureau has the strongest collaboration work. Research could be subdivided into four keyword clusters: "culture and differentiation," "morphogenesis and modeling," "gene therapy," and "transplantation and visual restoration," and evolution of keywords was identified. Last decade has witnessed the huge progress in the field of RO, which is a young and promising research area with extensive and in-depth studies. More attention should be paid to RO-related models and therapies based on specific retinal diseases, especially inherited retinopathies.
Collapse
Affiliation(s)
- Wenyue Shen
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - An Shao
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Wuyuan Zhou
- Zhejiang Academy of Science and Technology Information, Hangzhou, China
| | - Lixia Lou
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Poznan, Poland
| | - Kai Jin
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| |
Collapse
|
7
|
Diacou R, Nandigrami P, Fiser A, Liu W, Ashery-Padan R, Cvekl A. Cell fate decisions, transcription factors and signaling during early retinal development. Prog Retin Eye Res 2022; 91:101093. [PMID: 35817658 PMCID: PMC9669153 DOI: 10.1016/j.preteyeres.2022.101093] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Raven Diacou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Prithviraj Nandigrami
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruth Ashery-Padan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
8
|
Grigoryan EN. Self-Organization of the Retina during Eye Development, Retinal Regeneration In Vivo, and in Retinal 3D Organoids In Vitro. Biomedicines 2022; 10:1458. [PMID: 35740479 PMCID: PMC9221005 DOI: 10.3390/biomedicines10061458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022] Open
Abstract
Self-organization is a process that ensures histogenesis of the eye retina. This highly intricate phenomenon is not sufficiently studied due to its biological complexity and genetic heterogeneity. The review aims to summarize the existing central theories and ideas for a better understanding of retinal self-organization, as well as to address various practical problems of retinal biomedicine. The phenomenon of self-organization is discussed in the spatiotemporal context and illustrated by key findings during vertebrate retina development in vivo and retinal regeneration in amphibians in situ. Described also are histotypic 3D structures obtained from the disaggregated retinal progenitor cells of birds and retinal 3D organoids derived from the mouse and human pluripotent stem cells. The review highlights integral parts of retinal development in these conditions. On the cellular level, these include competence, differentiation, proliferation, apoptosis, cooperative movements, and migration. On the physical level, the focus is on the mechanical properties of cell- and cell layer-derived forces and on the molecular level on factors responsible for gene regulation, such as transcription factors, signaling molecules, and epigenetic changes. Finally, the self-organization phenomenon is discussed as a basis for the production of retinal organoids, a promising model for a wide range of basic scientific and medical applications.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
9
|
O’Hara-Wright M, Mobini S, Gonzalez-Cordero A. Bioelectric Potential in Next-Generation Organoids: Electrical Stimulation to Enhance 3D Structures of the Central Nervous System. Front Cell Dev Biol 2022; 10:901652. [PMID: 35656553 PMCID: PMC9152151 DOI: 10.3389/fcell.2022.901652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived organoid models of the central nervous system represent one of the most exciting areas in in vitro tissue engineering. Classically, organoids of the brain, retina and spinal cord have been generated via recapitulation of in vivo developmental cues, including biochemical and biomechanical. However, a lesser studied cue, bioelectricity, has been shown to regulate central nervous system development and function. In particular, electrical stimulation of neural cells has generated some important phenotypes relating to development and differentiation. Emerging techniques in bioengineering and biomaterials utilise electrical stimulation using conductive polymers. However, state-of-the-art pluripotent stem cell technology has not yet merged with this exciting area of bioelectricity. Here, we discuss recent findings in the field of bioelectricity relating to the central nervous system, possible mechanisms, and how electrical stimulation may be utilised as a novel technique to engineer “next-generation” organoids.
Collapse
Affiliation(s)
- Michelle O’Hara-Wright
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM + CSIC), Madrid, Spain
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
- *Correspondence: Anai Gonzalez-Cordero,
| |
Collapse
|
10
|
Phan TKT, Do TL, Tachibana K, Kihara T. Alpha-mangostin dephosphorylates ERM to induce adhesion and decrease surface stiffness in KG-1 cells. Hum Cell 2021; 35:189-198. [PMID: 34817798 DOI: 10.1007/s13577-021-00651-8] [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: 08/18/2021] [Accepted: 11/18/2021] [Indexed: 11/30/2022]
Abstract
Surface stiffness is a unique indicator of various cellular states and events and needs to be tightly controlled. α-Mangostin, a natural compound with numerous bioactivities, reduces the mechanical stiffness of various cells; however, the mechanism by which it affects the actin cytoskeleton remains unclear. We aimed to elucidate the mechanism underlying α-mangostin activity on the surface stiffness of leukocytes. We treated spherical non-adherent myelomonocytic KG-1 cells with α-mangostin; it clearly reduced their surface stiffness and disrupted their microvilli. The α-mangostin-induced reduction in surface stiffness was inhibited by calyculin A, a protein phosphatase inhibitor. α-Mangostin also induced KG-1 cell adhesion to a fibronectin-coated surface. In KG-1 cells, a decrease in surface stiffness and the induction of cell adhesion are largely attributed to the dephosphorylation of ezrin/radixin/moesin proteins (ERMs); α-mangostin reduced the levels of phosphorylated ERMs. It further increased protein kinase C (PKC) activity. α-Mangostin-induced KG-1 cell adhesion and cell surface softness were inhibited by the PKC inhibitor GF109203X. The results of the present study suggest that α-mangostin decreases stiffness and induces adhesion of KG-1 cells via PKC activation and ERM dephosphorylation.
Collapse
Affiliation(s)
- Thi Kieu Trang Phan
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Health Care System, 458 Minh Khai, Hai Ba Trung, Hanoi, Vietnam
| | - Thi Ly Do
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan
| | - Kouichi Tachibana
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
- Department of Hematology and Oncology, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Takanori Kihara
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, 1-1 Hibikino, Wakamatsu, Kitakyushu, Fukuoka, 808-0135, Japan.
| |
Collapse
|
11
|
Rasys AM, Pau SH, Irwin KE, Luo S, Kim HQ, Wahle MA, Trainor PA, Menke DB, Lauderdale JD. Ocular elongation and retraction in foveated reptiles. Dev Dyn 2021; 250:1584-1599. [PMID: 33866663 PMCID: PMC10731578 DOI: 10.1002/dvdy.348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Pronounced asymmetric changes in ocular globe size during eye development have been observed in a number of species ranging from humans to lizards. In contrast, largely symmetric changes in globe size have been described for other species like rodents. We propose that asymmetric changes in the three-dimensional structure of the developing eye correlate with the types of retinal remodeling needed to produce areas of high photoreceptor density. To test this idea, we systematically examined three-dimensional aspects of globe size as a function of eye development in the bifoveated brown anole, Anolis sagrei. RESULTS During embryonic development, the anole eye undergoes dynamic changes in ocular shape. Initially spherical, the eye elongates in the presumptive foveal regions of the retina and then proceeds through a period of retraction that returns the eye to its spherical shape. During this period of retraction, pit formation and photoreceptor cell packing are observed. We found a similar pattern of elongation and retraction associated with the single fovea of the veiled chameleon, Chamaeleo calyptratus. CONCLUSIONS These results, together with those reported for other foveated species, support the idea that areas of high photoreceptor packing occur in regions where the ocular globe asymmetrically elongates and retracts during development.
Collapse
Affiliation(s)
- Ashley M. Rasys
- Department of Cellular Biology, The University of Georgia, Athens, Georgia
| | - Shana H. Pau
- Department of Genetics, The University of Georgia, Athens, Georgia
| | - Katherine E. Irwin
- Department of Cellular Biology, The University of Georgia, Athens, Georgia
| | - Sherry Luo
- Department of Genetics, The University of Georgia, Athens, Georgia
| | - Hannah Q. Kim
- Department of Cellular Biology, The University of Georgia, Athens, Georgia
| | | | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri
- Department of Anatomy & Cell Biology, The University of Kansas School of Medicine, Kansas City, Kansas
| | - Douglas B. Menke
- Department of Genetics, The University of Georgia, Athens, Georgia
| | - James D. Lauderdale
- Department of Cellular Biology, The University of Georgia, Athens, Georgia
- Neuroscience Division of the Biomedical and Translational Sciences Institute, The University of Georgia, Athens, Georgia
| |
Collapse
|
12
|
Moreno-Mármol T, Ledesma-Terrón M, Tabanera N, Martin-Bermejo MJ, Cardozo MJ, Cavodeassi F, Bovolenta P. Stretching of the retinal pigment epithelium contributes to zebrafish optic cup morphogenesis. eLife 2021; 10:63396. [PMID: 34545806 PMCID: PMC8530511 DOI: 10.7554/elife.63396] [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: 09/23/2020] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
The vertebrate eye primordium consists of a pseudostratified neuroepithelium, the optic vesicle (OV), in which cells acquire neural retina or retinal pigment epithelium (RPE) fates. As these fates arise, the OV assumes a cup shape, influenced by mechanical forces generated within the neural retina. Whether the RPE passively adapts to retinal changes or actively contributes to OV morphogenesis remains unexplored. We generated a zebrafish Tg(E1-bhlhe40:GFP) line to track RPE morphogenesis and interrogate its participation in OV folding. We show that, in virtual absence of proliferation, RPE cells stretch and flatten, thereby matching the retinal curvature and promoting OV folding. Localized interference with the RPE cytoskeleton disrupts tissue stretching and OV folding. Thus, extreme RPE flattening and accelerated differentiation are efficient solutions adopted by fast-developing species to enable timely optic cup formation. This mechanism differs in amniotes, in which proliferation drives RPE expansion with a much-reduced need of cell flattening. Rounded eyeballs help to optimize vision – but how do they acquire their distinctive shape? In animals with backbones, including humans, the eye begins to form early in development. A single layer of embryonic tissue called the optic vesicle reorganizes itself into a two-layered structure: a thin outer layer of cells, known as the retinal pigmented epithelium (RPE for short), and a thicker inner layer called the neural retina. If this process fails, the animal may be born blind or visually impaired. How this flat two-layered structure becomes round is still being investigated. In fish, studies have shown that the inner cell layer – the neural retina – generates mechanical forces that cause the developing tissue to curve inwards to form a cup-like shape. But it was unclear whether the outer layer of cells (the RPE) also contributed to this process. Moreno-Marmol et al. were able to investigate this question by genetically modifying zebrafish to make all new RPE cells fluoresce. Following the early development of the zebrafish eye under a microscope revealed that RPE cells flattened themselves into long thin structures that stretched to cover the entire neural retina. This change was made possible by the cell’s internal skeleton reorganizing. In fact, preventing this reorganization stopped the RPE cells from flattening, and precluded the optic cup from acquiring its curved shape. The results thus confirmed a direct role for the RPE in generating curvature. The entire process did not require the RPE to produce new cells, allowing the curved shape to emerge in just a few hours. This is a major advantage for fast-developing species such as zebrafish. In species whose embryos develop more slowly, such as mice and humans, the RPE instead grows by producing additional cells – a process that takes many days. The development of the eye thus shows how various species use different evolutionary approaches to achieve a common goal.
Collapse
Affiliation(s)
- Tania Moreno-Mármol
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Mario Ledesma-Terrón
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain
| | - Noemi Tabanera
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Maria Jesús Martin-Bermejo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marcos J Cardozo
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, c/ Nicolás Cabrera, 1, Campus de la Universidad Autónoma de Madrid, Madrid, Spain.,CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| |
Collapse
|
13
|
Döpper H, Menges J, Bozet M, Brenzel A, Lohmann D, Steenpass L, Kanber D. Differentiation Protocol for 3D Retinal Organoids, Immunostaining and Signal Quantitation. ACTA ACUST UNITED AC 2021; 55:e120. [PMID: 32956559 DOI: 10.1002/cpsc.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Structures resembling whole organs, called organoids, are generated using pluripotent stem cells and 3D culturing methods. This relies on the ability of cells to self-reorganize after dissociation. In combination with certain supplemented factors, differentiation can be directed toward the formation of several organ-like structures. Here, a protocol for the generation of retinal organoids containing all seven retinal cell types is described. This protocol does not depend on Matrigel, and by keeping the organoids single and independent at all times, fusion is prevented and monitoring of differentiation is improved. Comprehensive phenotypic characterization of the in vitro-generated retinal organoids is achieved by the protocol for immunostaining outlined here. By comparing different stages of retinal organoids, the decrease and increase of certain cell populations can be determined. In order to be able to detect even small differences, it is necessary to quantify the immunofluorescent signals, for which we have provided a detailed protocol describing signal quantitation using the image-processing program Fiji. © 2020 The Authors. Basic Protocol 1: Differentiation protocol for 3D retinal organoids Basic Protocol 2: Immunostaining protocol for cryosections of retinal organoids Support Protocol: Embedding and sectioning protocol for 3D retinal organoids Basic Protocol 3: Quantitation protocol using Fiji.
Collapse
Affiliation(s)
- Hannah Döpper
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julia Menges
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Morgane Bozet
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Brenzel
- Institute for Experimental Immunology and Imaging, Imaging Center Essen (LMU), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dietmar Lohmann
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Laura Steenpass
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Present address: Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures GmbH, Braunschweig, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
14
|
O'Hara-Wright M, Gonzalez-Cordero A. Retinal organoids: a window into human retinal development. Development 2020; 147:147/24/dev189746. [PMID: 33361444 PMCID: PMC7774906 DOI: 10.1242/dev.189746] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Retinal development and maturation are orchestrated by a series of interacting signalling networks that drive the morphogenetic transformation of the anterior developing brain. Studies in model organisms continue to elucidate these complex series of events. However, the human retina shows many differences from that of other organisms and the investigation of human eye development now benefits from stem cell-derived organoids. Retinal differentiation methods have progressed from simple 2D adherent cultures to self-organising micro-physiological systems. As models of development, these have collectively offered new insights into the previously unexplored early development of the human retina and informed our knowledge of the key cell fate decisions that govern the specification of light-sensitive photoreceptors. Although the developmental trajectories of other retinal cell types remain more elusive, the collation of omics datasets, combined with advanced culture methodology, will enable modelling of the intricate process of human retinogenesis and retinal disease in vitro. Summary: Retinal organoid systems derived from human pluripotent stem cells are micro-physiological systems that offer new insights into previously unexplored human retina development.
Collapse
Affiliation(s)
- Michelle O'Hara-Wright
- Stem Cell Medicine Group, Children's Medical Research Institute, University of Sydney, Westmead, 2145, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145, NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, University of Sydney, Westmead, 2145, NSW, Australia .,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145, NSW, Australia
| |
Collapse
|
15
|
Grocott T, Lozano-Velasco E, Mok GF, Münsterberg AE. The Pax6 master control gene initiates spontaneous retinal development via a self-organising Turing network. Development 2020; 147:dev185827. [PMID: 33214222 PMCID: PMC7774904 DOI: 10.1242/dev.185827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 11/05/2020] [Indexed: 12/14/2022]
Abstract
Understanding how complex organ systems are assembled from simple embryonic tissues is a major challenge. Across the animal kingdom a great diversity of visual organs are initiated by a 'master control gene' called Pax6, which is both necessary and sufficient for eye development. Yet precisely how Pax6 achieves this deeply homologous function is poorly understood. Using the chick as a model organism, we show that vertebrate Pax6 interacts with a pair of morphogen-coding genes, Tgfb2 and Fst, to form a putative Turing network, which we have computationally modelled. Computer simulations suggest that this gene network is sufficient to spontaneously polarise the developing retina, establishing the first organisational axis of the eye and prefiguring its further development. Our findings reveal how retinal self-organisation may be initiated independently of the highly ordered tissue interactions that help to assemble the eye in vivo These results help to explain how stem cell aggregates spontaneously self-organise into functional eye-cups in vitro We anticipate these findings will help to underpin retinal organoid technology, which holds much promise as a platform for disease modelling, drug development and regenerative therapies.
Collapse
Affiliation(s)
- Timothy Grocott
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
| | | | | | | |
Collapse
|
16
|
West EL, Ribeiro J, Ali RR. Development of Stem Cell Therapies for Retinal Degeneration. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035683. [PMID: 31818854 DOI: 10.1101/cshperspect.a035683] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Degenerative retinal disease is the major cause of sight loss in the developed world and currently there is a lack of effective treatments. As the loss of vision is directly the result of the loss of retinal cells, effective cell replacement through stem-cell-based therapies may have the potential to treat a great number of retinal diseases whatever their underlying etiology. The eye is an ideal organ to develop cell therapies as it is immune privileged, and modern surgical techniques enable precise delivery of cells to the retina. Furthermore, a range of noninvasive diagnostic tests and high-resolution imaging techniques facilitate the evaluation of any therapeutic intervention. In this review, we evaluate the progress to date of current cell therapy strategies for retinal repair, focusing on transplantation of pluripotent stem-cell-derived retinal pigment epithelium (RPE) and photoreceptor cells.
Collapse
Affiliation(s)
- Emma L West
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Joana Ribeiro
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Robin R Ali
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom.,Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan 48105, USA
| |
Collapse
|
17
|
Gong Y, He X, Li Q, He J, Bian B, Li Y, Ge L, Zeng Y, Xu H, Yin ZQ. SCF/SCFR signaling plays an important role in the early morphogenesis and neurogenesis of human embryonic neural retina. Development 2019; 146:dev.174409. [PMID: 31548215 DOI: 10.1242/dev.174409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/16/2019] [Indexed: 12/20/2022]
Abstract
The stem cell factor receptor (SCFR) has been demonstrated to be expressed in the neural retina of mice, rat and human for decades. Previous reports indicated that the SCFR correlates with glia differentiation of late retinal progenitor cells (RPCs), retinal vasculogenesis and homeostasis of the blood-retinal barrier. However, the role of SCF/SCFR signaling in the growth and development of the neural retina (NR), especially in the early embryonic stage, remains poorly understood. Here, we show that SCF/SCFR signaling orchestrates invagination of the human embryonic stem cell (hESC)-derived NR via regulation of cell cycle progression, cytoskeleton dynamic and apical constriction of RPCs in the ciliary marginal zone (CMZ). Furthermore, activation of SCF/SCFR signaling promotes neurogenesis in the central-most NR via acceleration of the migration of immature ganglion cells and repressing apoptosis. Our study reveals an unreported role for SCF/SCFR signaling in controlling ciliary marginal cellular behaviors during early morphogenesis and neurogenesis of the human embryonic NR, providing a new potential therapeutic target for human congenital eye diseases such as anophthalmia, microphthalmia and congenital high myopia.
Collapse
Affiliation(s)
- Yu Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Xiangyu He
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Qiyou Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Juncai He
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Baishijiao Bian
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Yijian Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Linlin Ge
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Yuxiao Zeng
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Haiwei Xu
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Zheng Qin Yin
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| |
Collapse
|
18
|
Bayir E, Sendemir A, Missirlis YF. Mechanobiology of cells and cell systems, such as organoids. Biophys Rev 2019; 11:721-728. [PMID: 31502190 DOI: 10.1007/s12551-019-00590-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/30/2019] [Indexed: 01/04/2023] Open
Abstract
Organoids are in vitro 3D self-organizing tissues that mimic embryogenesis. Organoid research is advancing at a tremendous pace, since it offers great opportunities for disease modeling, drug development and screening, personalized medicine, as well as understanding organogenesis. Mechanobiology of organoids is an unexplored area, which can shed light to several unexplained aspects of self-organization behavior in organogenesis. It is becoming evident that collective cell behavior is distinctly different from individual cells' conduct against certain stimulants. Inherently consisting of higher number of degrees of freedom for cell motility and more complex cell-to-cell and cell-to-extracellular matrix behavior, understanding mechanotransduction in organoids is even more challenging compared with cell communities in 2D culture conditions. Yet, deciphering mechanobiology of organoids can help us understand effects of mechanical cues in health and disease, and translate findings of basic research toward clinical diagnosis and therapy.
Collapse
Affiliation(s)
- Ece Bayir
- Central Research Test and Analysis Laboratory Application and Research Center (EGE-MATAL), Ege University, Izmir, Turkey
| | - Aylin Sendemir
- Department of Bioengineering, Ege University, Izmir, Turkey
| | - Yannis F Missirlis
- Department of Mechanical Engineering & Aeronautics, University of Patras, Patras, Greece.
| |
Collapse
|
19
|
Takeda H, Kameo Y, Inoue Y, Adachi T. An energy landscape approach to understanding variety and robustness in tissue morphogenesis. Biomech Model Mechanobiol 2019; 19:471-479. [PMID: 31494791 PMCID: PMC7105444 DOI: 10.1007/s10237-019-01222-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/27/2019] [Indexed: 12/17/2022]
Abstract
During morphogenesis in development, multicellular tissues deform by mechanical forces induced by spatiotemporally regulated cellular activities, such as cell proliferation and constriction. Various morphologies are formed because of various spatiotemporal combinations and sequences of multicellular activities. Despite its potential to variations, morphogenesis is a surprisingly robust process, in which qualitatively similar morphologies are reproducibly formed even under spatiotemporal fluctuation of multicellular activities. To understand these essential characteristics of tissue morphogenesis, which involves the coexistence of various morphologies and robustness of the morphogenetic process, in this study, we propose a novel approach to capture the overall view of morphogenesis from mechanical viewpoints. This approach will enable visualization of the energy landscape, which includes morphogenetic processes induced by admissible histories of cellular activities. This approach was applied to investigate the morphogenesis of a sheet-like tissue with curvature, where it deformed to a concave or convex morphology depending on the history of growth and constriction. Qualitatively different morphologies were produced by bifurcation of the valley in the energy landscape. The depth and steepness of the valley near the stable states represented the degree of robustness to fluctuations of multicellular activities. Furthermore, as a realistic example, we showed an application of this approach to luminal folding observed in the initial stage of intestinal villus formation. This approach will be helpful to understand the mechanism of how various morphologies are formed and how tissues reproducibly achieve specific morphologies.
Collapse
Affiliation(s)
- Hironori Takeda
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
| | - Yoshitaka Kameo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
- Division of Systemic Life Science, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
| | - Yasuhiro Inoue
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
- Division of Systemic Life Science, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan.
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan.
- Division of Systemic Life Science, Graduate School of Biostudies, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo, Kyoto, 606-8507, Japan.
| |
Collapse
|
20
|
Cavodeassi F, Wilson SW. Looking to the future of zebrafish as a model to understand the genetic basis of eye disease. Hum Genet 2019; 138:993-1000. [PMID: 31422478 PMCID: PMC6710215 DOI: 10.1007/s00439-019-02055-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 02/07/2023]
Abstract
In this brief commentary, we provide some of our thoughts and opinions on the current and future use of zebrafish to model human eye disease, dissect pathological progression and advance in our understanding of the genetic bases of microphthalmia, andophthalmia and coloboma (MAC) in humans. We provide some background on eye formation in fish and conservation and divergence across vertebrates in this process, discuss different approaches for manipulating gene function and speculate on future research areas where we think research using fish may prove to be particularly effective.
Collapse
Affiliation(s)
- Florencia Cavodeassi
- Institute of Medical and Biomedical Education, St. George's, University of London, Cranmer Terrace, London, SW17 0RE, UK.
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, Biosciences, UCL, Gower St, London, WC1E 6BT, UK
| |
Collapse
|
21
|
Hunt NC, Hallam D, Chichagova V, Steel DH, Lako M. The Application of Biomaterials to Tissue Engineering Neural Retina and Retinal Pigment Epithelium. Adv Healthc Mater 2018; 7:e1800226. [PMID: 30175520 DOI: 10.1002/adhm.201800226] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/16/2018] [Indexed: 12/21/2022]
Abstract
The prevalence of degenerative retinal disease is ever increasing as life expectancy rises globally. The human retina fails to regenerate and the use of human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) to engineer retinal tissue is of particular interest due to the limited availability of suitable allogeneic or autologous tissue. Retinal tissue and its development are well characterized, which have resulted in robust assays to assess the development of tissue-engineered retina. Retinal tissue can be generated in vitro from hESCs and hiPSCs without biomaterial scaffolds, but despite advancements, protocols remain slow, expensive, and fail to result in mature functional tissue. Several recent studies have demonstrated the potential of biomaterial scaffolds to enhance generation of hESC/hiPSC-derived retinal tissue, including synthetic polymers, silk, alginate, hyaluronic acid, and extracellular matrix molecules. This review outlines the advances that have been made toward tissue-engineered neural retina and retinal pigment epithelium (RPE) for clinical application in recent years, including the success of clinical trials involving transplantation of cells and tissue to promote retinal repair; and the evidence from in vitro and animal studies that biomaterials can enhance development and integration of retinal tissue.
Collapse
Affiliation(s)
- Nicola C. Hunt
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Dean Hallam
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Valeria Chichagova
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
- Biomedicine WestInternational Centre for LifeTimes SquareNewcastle upon Tyne NE1 4EP UK
| | - David H. Steel
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Majlinda Lako
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| |
Collapse
|
22
|
Okuda S, Takata N, Hasegawa Y, Kawada M, Inoue Y, Adachi T, Sasai Y, Eiraku M. Strain-triggered mechanical feedback in self-organizing optic-cup morphogenesis. SCIENCE ADVANCES 2018; 4:eaau1354. [PMID: 30474058 PMCID: PMC6248953 DOI: 10.1126/sciadv.aau1354] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 10/19/2018] [Indexed: 05/27/2023]
Abstract
Organogenesis is a self-organizing process of multiple cells in three-dimensional (3D) space, where macroscopic tissue deformations are robustly regulated by multicellular autonomy. It is clear that this robust regulation requires cells to sense and modulate 3D tissue formation across different scales, but its underlying mechanisms are still unclear. To address this question, we developed a versatile computational model of 3D multicellular dynamics at single-cell resolution and combined it with the 3D culture system of pluripotent stem cell-derived optic-cup organoid. The complementary approach enabled quantitative prediction of morphogenesis and its corresponding verification and elucidated that the macroscopic 3D tissue deformation is fed back to individual cellular force generations via mechanosensing. We hereby conclude that mechanical force plays a key role as a feedback regulator to establish the robustness of organogenesis.
Collapse
Affiliation(s)
- S. Okuda
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
- PRESTO, Japan Science and Technology Agency, Kawaguchi 332-0012, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - N. Takata
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Y. Hasegawa
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - M. Kawada
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Y. Inoue
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - T. Adachi
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Y. Sasai
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - M. Eiraku
- RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
23
|
Moreno-Marmol T, Cavodeassi F, Bovolenta P. Setting Eyes on the Retinal Pigment Epithelium. Front Cell Dev Biol 2018; 6:145. [PMID: 30406103 PMCID: PMC6207792 DOI: 10.3389/fcell.2018.00145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/08/2018] [Indexed: 01/08/2023] Open
Abstract
The neural component of the zebrafish eye derives from a small group of cells known as the eye/retinal field. These cells, positioned in the anterior neural plate, rearrange extensively and generate the optic vesicles (OVs). Each vesicle subsequently folds over itself to form the double-layered optic cup, from which the mature eye derives. During this transition, cells of the OV are progressively specified toward three different fates: the retinal pigment epithelium (RPE), the neural retina, and the optic stalk. Recent studies have shown that folding of the zebrafish OV into a cup is in part driven by basal constriction of the cells of the future neural retina. During folding, however, RPE cells undergo an even more dramatic shape conversion that seems to entail the acquisition of unique properties. How these changes occur and whether they contribute to optic cup formation is still poorly understood. Here we will review present knowledge on RPE morphogenesis and discuss potential mechanisms that may explain such transformation using examples taken from embryonic Drosophila tissues that undergo similar shape changes. We will also put forward a hypothesis for optic cup folding that considers an active contribution from the RPE.
Collapse
Affiliation(s)
- Tania Moreno-Marmol
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| | - Florencia Cavodeassi
- Institute of Medical and Biomedical Education, University of London, London, United Kingdom
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
24
|
Luo Z, Zhong X, Li K, Xie B, Liu Y, Ye M, Li K, Xu C, Ge J. An Optimized System for Effective Derivation of Three-Dimensional Retinal Tissue via Wnt Signaling Regulation. Stem Cells 2018; 36:1709-1722. [DOI: 10.1002/stem.2890] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Ziming Luo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Xiufeng Zhong
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Bingbing Xie
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Yuchun Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Meifang Ye
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Kang Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Chaochao Xu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| |
Collapse
|
25
|
Tanner K. Perspective: The role of mechanobiology in the etiology of brain metastasis. APL Bioeng 2018; 2:031801. [PMID: 31069312 PMCID: PMC6324204 DOI: 10.1063/1.5024394] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Tumor latency and dormancy are obstacles to effective cancer treatment. In brain
metastases, emergence of a lesion can occur at varying intervals from diagnosis
and in some cases following successful treatment of the primary tumor. Genetic
factors that drive brain metastases have been identified, such as those involved
in cell adhesion, signaling, extravasation, and metabolism. From this wealth of
knowledge, vexing questions still remain; why is there a difference in strategy
to facilitate outgrowth and why is there a difference in latency? One missing
link may be the role of tissue biophysics of the brain microenvironment in
infiltrating cells. Here, I discuss the mechanical cues that may influence
disseminated tumor cells in the brain, as a function of age and disease. I
further discuss in vitro and in vivo
preclinical models such as 3D culture systems and zebrafish to study the role of
the mechanical environment in brain metastasis in an effort of providing novel
targeted therapeutics.
Collapse
Affiliation(s)
- Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
26
|
Dynamic Tissue Rearrangements during Vertebrate Eye Morphogenesis: Insights from Fish Models. J Dev Biol 2018; 6:jdb6010004. [PMID: 29615553 PMCID: PMC5875564 DOI: 10.3390/jdb6010004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/22/2018] [Accepted: 02/27/2018] [Indexed: 12/16/2022] Open
Abstract
Over the last thirty years, fish models, such as the zebrafish and medaka, have become essential to pursue developmental studies and model human disease. Community efforts have led to the generation of wide collections of mutants, a complete sequence of their genomes, and the development of sophisticated genetic tools, enabling the manipulation of gene activity and labelling and tracking of specific groups of cells during embryonic development. When combined with the accessibility and optical clarity of fish embryos, these approaches have made of them an unbeatable model to monitor developmental processes in vivo and in real time. Over the last few years, live-imaging studies in fish have provided fascinating insights into tissue morphogenesis and organogenesis. This review will illustrate the advantages of fish models to pursue morphogenetic studies by highlighting the findings that, in the last decade, have transformed our understanding of eye morphogenesis.
Collapse
|
27
|
How mechanical forces shape the developing eye. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 137:25-36. [PMID: 29432780 DOI: 10.1016/j.pbiomolbio.2018.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/08/2018] [Accepted: 01/15/2018] [Indexed: 12/29/2022]
Abstract
In the vertebrate embryo, the eyes develop from optic vesicles that grow laterally outward from the brain tube and contact the overlying surface ectoderm. Within the region of contact, each optic vesicle and the surface ectoderm thicken to form placodes, which then invaginate to create the optic cup and lens pit, respectively. Eventually, the optic cup becomes the retina, while the lens pit closes to form the lens vesicle. Here, we review current hypotheses for the physical mechanisms that create these structures and present novel three-dimensional computer (finite-element) models to illustrate the plausibility and limitations of these hypotheses. Taken together, experimental and numerical results suggest that the driving forces for early eye morphogenesis are generated mainly by differential growth, actomyosin contraction, and regional apoptosis, with morphology mediated by physical constraints provided by adjacent tissues and extracellular matrix. While these studies offer new insight into the mechanics of eye development, future work is needed to better understand how these mechanisms are regulated to precisely control the shape of the eye.
Collapse
|
28
|
Kim MH, Kino-oka M. Bioprocessing Strategies for Pluripotent Stem Cells Based on Waddington’s Epigenetic Landscape. Trends Biotechnol 2018; 36:89-104. [DOI: 10.1016/j.tibtech.2017.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
|
29
|
Martinez-Morales JR, Cavodeassi F, Bovolenta P. Coordinated Morphogenetic Mechanisms Shape the Vertebrate Eye. Front Neurosci 2017; 11:721. [PMID: 29326547 PMCID: PMC5742352 DOI: 10.3389/fnins.2017.00721] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022] Open
Abstract
The molecular bases of vertebrate eye formation have been extensively investigated during the past 20 years. This has resulted in the definition of the backbone of the gene regulatory networks controlling the different steps of eye development and has further highlighted a substantial conservation of these networks among vertebrates. Yet, the precise morphogenetic events allowing the formation of the optic cup from a small group of cells within the anterior neural plate are still poorly understood. It is also unclear if the morphogenetic events leading to eyes of very similar shape are indeed comparable among all vertebrates or if there are any species-specific peculiarities. Improved imaging techniques have enabled to follow how the eye forms in living embryos of a few vertebrate models, whereas the development of organoid cultures has provided fascinating tools to recapitulate tissue morphogenesis of other less accessible species. Here, we will discuss what these advances have taught us about eye morphogenesis, underscoring possible similarities and differences among vertebrates. We will also discuss the contribution of cell shape changes to this process and how morphogenetic and patterning mechanisms integrate to assemble the final architecture of the eye.
Collapse
Affiliation(s)
| | - Florencia Cavodeassi
- Centro de Biología Molecular Severo Ochoa, (CSIC/UAM), Madrid, Spain.,CIBERER, ISCIII, Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, (CSIC/UAM), Madrid, Spain.,CIBERER, ISCIII, Madrid, Spain
| |
Collapse
|
30
|
Stem cells and genome editing: approaches to tissue regeneration and regenerative medicine. J Hum Genet 2017; 63:165-178. [PMID: 29192237 DOI: 10.1038/s10038-017-0348-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Abstract
Understanding the basis of regeneration of each tissue and organ, and incorporating this knowledge into clinical treatments for degenerative tissues and organs in patients, are major goals for researchers in regenerative biology. Here we provide an overview of current work, from high-regeneration animal models, to stem cell-based culture models, transplantation technologies, large-animal chimeric models, and programmable nuclease-based genome-editing technologies. Three-dimensional culture generating organoids, which represents intact tissue/organ identity including cell fate and morphology are getting more general approaches in the fields by taking advantage of embryonic stem cells, induced pluripotent stem cells and adult stem cells. The organoid culture system potentially has profound impact on the field of regenerative medicine. We also emphasize that the large animal model, in particular pig model would be a hope to manufacture humanized organs in in vivo empty (vacant) niche, which now potentially allows not only appropriate cell fate identity but nearly the same property as human organs in size. Therefore, integrative and collaborative researches across different fields might be critical to the aims needed in clinical trial.
Collapse
|
31
|
Postnatal evolution of the ciliary processes in the gerbil (Meriones unguiculatus): a structural, ultrastructural and morphometric study. ZOOMORPHOLOGY 2017. [DOI: 10.1007/s00435-017-0378-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Dahl-Jensen S, Grapin-Botton A. The physics of organoids: a biophysical approach to understanding organogenesis. Development 2017; 144:946-951. [PMID: 28292839 DOI: 10.1242/dev.143693] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Organoids representing a diversity of tissues have recently been created, bridging the gap between cell culture and experiments performed in vivo Being small and amenable to continuous monitoring, they offer the opportunity to scrutinize the dynamics of organ development, including the exciting prospect of observing aspects of human embryo development live. From a physicist's perspective, their ability to self-organize - to differentiate and organize cells in space - calls for the identification of the simple rules that underlie this capacity. Organoids provide tractable conditions to investigate the effects of the growth environment, including its molecular composition and mechanical properties, along with the initial conditions such as cell number and type(s). From a theoretical standpoint, different types of in silico modeling can complement the measurements performed in organoids to understand the role of chemical diffusion, contact signaling, differential cell adhesion and mechanical controls. Here, we discuss what it means to take a biophysical approach to understanding organogenesis in vitro and how we might expect such approaches to develop in the future.
Collapse
Affiliation(s)
- Svend Dahl-Jensen
- DanStem and StemPhys, University of Copenhagen, 3B Blegdamsvej, Copenhagen N DK-2200, Denmark .,StemPhys, Niels Bohr Institute, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Anne Grapin-Botton
- DanStem and StemPhys, University of Copenhagen, 3B Blegdamsvej, Copenhagen N DK-2200, Denmark
| |
Collapse
|
33
|
From morphogen to morphogenesis and back. Nature 2017; 541:311-320. [DOI: 10.1038/nature21348] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 11/18/2016] [Indexed: 12/11/2022]
|
34
|
Nicolás-Pérez M, Kuchling F, Letelier J, Polvillo R, Wittbrodt J, Martínez-Morales JR. Analysis of cellular behavior and cytoskeletal dynamics reveal a constriction mechanism driving optic cup morphogenesis. eLife 2016; 5. [PMID: 27797321 PMCID: PMC5110244 DOI: 10.7554/elife.15797] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022] Open
Abstract
Contractile actomyosin networks have been shown to power tissue morphogenesis. Although the basic cellular machinery generating mechanical tension appears largely conserved, tensions propagate in unique ways within each tissue. Here we use the vertebrate eye as a paradigm to investigate how tensions are generated and transmitted during the folding of a neuroepithelial layer. We record membrane pulsatile behavior and actomyosin dynamics during zebrafish optic cup morphogenesis by live imaging. We show that retinal neuroblasts undergo fast oscillations and that myosin condensation correlates with episodic contractions that progressively reduce basal feet area. Interference with lamc1 function impairs basal contractility and optic cup folding. Mapping of tensile forces by laser cutting uncover a developmental window in which local ablations trigger the displacement of the entire tissue. Our work shows that optic cup morphogenesis is driven by a constriction mechanism and indicates that supra-cellular transmission of mechanical tension depends on ECM attachment. DOI:http://dx.doi.org/10.7554/eLife.15797.001 Tissues and organs form into their final shapes because the cells in a developing embryo generate forces that alter their shape and position. Networks of fibres made from actin and myosin proteins generate these forces, and because the fibres can assemble in many different ways inside cells, they allow the cells to move and change shape in many different ways. Forces in some tissues can cause flat sheets of cells to bend. These sheets of cells are attached on one side (their “basal” surface) to a collection of membranes and molecules that are known as the extracellular matrix. When the cells in the sheet progressively shrink at their basal surface, causing the sheet to bend towards the extracellular matrix, this is known as basal constriction. Nicolás-Pérez et al. have used high-resolution imaging to record how basal constriction helps the optic cup – the main chamber of the eye – to form in zebrafish embryos. This imaging confirmed that a sheet of precursor cells progressively bends towards its basal surface to form the curved shape of the eyeball. Further analysis revealed that this basal constriction happens when myosin fibres accumulate in clusters along the basal surface of some of the precursor cells. The resulting contraction of the basal surface of the cells relies both on the tension generated by myosin inside the cell and on the cells being attached properly to the extracellular matrix. Using a laser beam, Nicolás-Pérez et al. also destroyed small parts of the basal surface of the retina. This procedure allows the mechanical tension distribution throughout the developing eye to be mapped. Laser ablations revealed a narrow time window during development when destroying small parts of the basal surface can cause the entire sheet of cells to relax, preventing it from curving to form the shape of the eye. Sheets of precursor cells are important building blocks of the nervous system, yet researchers only have limited knowledge of the processes that enable them to fold or bend into a final shape. As such, the findings of Nicolás-Pérez et al. will contribute to a wider understanding of how cells and tissues behave while the brain is forming. DOI:http://dx.doi.org/10.7554/eLife.15797.002
Collapse
Affiliation(s)
| | - Franz Kuchling
- Centro Andaluz de Biología del Desarrollo, Seville, Spain.,Centre for Organismal Studies, COS, University of Heidelberg, Heidelberg, Germany
| | | | - Rocío Polvillo
- Centro Andaluz de Biología del Desarrollo, Seville, Spain
| | - Jochen Wittbrodt
- Centre for Organismal Studies, COS, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
35
|
Specification of embryonic stem cell-derived tissues into eye fields by Wnt signaling using rostral diencephalic tissue-inducing culture. Mech Dev 2016; 141:90-99. [PMID: 27151576 DOI: 10.1016/j.mod.2016.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/26/2016] [Accepted: 05/01/2016] [Indexed: 01/13/2023]
Abstract
The eyes are subdivided from the rostral diencephalon in early development. How the neuroectoderm regulates this subdivision, however, is largely unknown. Taking advantage of embryonic stem cell (ESC) culture using a Rax reporter line to monitor rostral diencephalon formation, we found that ESC-derived tissues at day 7 grown in Glasgow Minimum Expression Media (GMEM) containing knockout serum replacement (KSR) exhibited higher levels of expression of axin2, a Wnt target gene, than those grown in chemically defined medium (CDM). Surprisingly, Wnt agonist facilitated eye field-like tissue specification in CDM. In contrast, the addition of Wnt antagonist diminished eye field tissue formation in GMEM+KSR. Furthermore, the morphological formation of the eye tissue anlage, including the optic vesicle, was accompanied by Wnt signaling activation. Additionally, using CDM culture, we developed an efficient method for generating Rax+/Chx10+ retinal progenitors, which could become fully stratified retina. Here we provide a new avenue for exploring the mechanisms of eye field specification in vitro.
Collapse
|
36
|
Ollé-Vila A, Duran-Nebreda S, Conde-Pueyo N, Montañez R, Solé R. A morphospace for synthetic organs and organoids: the possible and the actual. Integr Biol (Camb) 2016; 8:485-503. [PMID: 27032985 DOI: 10.1039/c5ib00324e] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Efforts in evolutionary developmental biology have shed light on how organs are developed and why evolution has selected some structures instead of others. These advances in the understanding of organogenesis along with the most recent techniques of organotypic cultures, tissue bioprinting and synthetic biology provide the tools to hack the physical and genetic constraints in organ development, thus opening new avenues for research in the form of completely designed or merely altered settings. Here we propose a unifying framework that connects the concept of morphospace (i.e. the space of possible structures) with synthetic biology and tissue engineering. We aim for a synthesis that incorporates our understanding of both evolutionary and architectural constraints and can be used as a guide for exploring alternative design principles to build artificial organs and organoids. We present a three-dimensional morphospace incorporating three key features associated to organ and organoid complexity. The axes of this space include the degree of complexity introduced by developmental mechanisms required to build the structure, its potential to store and react to information and the underlying physical state. We suggest that a large fraction of this space is empty, and that the void might offer clues for alternative ways of designing and even inventing new organs.
Collapse
Affiliation(s)
- Aina Ollé-Vila
- ICREA-Complex Systems Lab, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain.
| | | | | | | | | |
Collapse
|
37
|
Oltean A, Huang J, Beebe DC, Taber LA. Tissue growth constrained by extracellular matrix drives invagination during optic cup morphogenesis. Biomech Model Mechanobiol 2016; 15:1405-1421. [PMID: 26984743 DOI: 10.1007/s10237-016-0771-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/05/2016] [Indexed: 12/19/2022]
Abstract
In the early embryo, the eyes form initially as relatively spherical optic vesicles (OVs) that protrude from both sides of the brain tube. Each OV grows until it contacts and adheres to the overlying surface ectoderm (SE) via an extracellular matrix (ECM) that is secreted by the SE and OV. The OV and SE then thicken and bend inward (invaginate) to create the optic cup (OC) and lens vesicle, respectively. While constriction of cell apices likely plays a role in SE invagination, the mechanisms that drive OV invagination are poorly understood. Here, we used experiments and computational modeling to explore the hypothesis that the ECM locally constrains the growing OV, forcing it to invaginate. In chick embryos, we examined the need for the ECM by (1) removing SE at different developmental stages and (2) exposing the embryo to collagenase. At relatively early stages of invagination (Hamburger-Hamilton stage HH14[Formula: see text]), removing the SE caused the curvature of the OV to reverse as it 'popped out' and became convex, but the OV remained concave at later stages (HH15) and invaginated further during subsequent culture. Disrupting the ECM had a similar effect, with the OV popping out at early to mid-stages of invagination (HH14[Formula: see text] to HH14[Formula: see text]). These results suggest that the ECM is required for the early stages but not the late stages of OV invagination. Microindentation tests indicate that the matrix is considerably stiffer than the cellular OV, and a finite-element model consisting of a growing spherical OV attached to a relatively stiff layer of ECM reproduced the observed behavior, as well as measured temporal changes in OV curvature, wall thickness, and invagination depth reasonably well. Results from our study also suggest that the OV grows relatively uniformly, while the ECM is stiffer toward the center of the optic vesicle. These results are consistent with our matrix-constraint hypothesis, providing new insight into the mechanics of OC (early retina) morphogenesis.
Collapse
Affiliation(s)
- Alina Oltean
- Department of Biomedical Engineering, Washington University, One Brookings Drive, Campus Box 1097, Saint Louis, MO, 63130-4899, USA.
| | - Jie Huang
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, 63130, USA
| | - David C Beebe
- Department of Ophthalmology and Visual Sciences, Washington University, Saint Louis, MO, 63130, USA
| | - Larry A Taber
- Department of Biomedical Engineering, Washington University, One Brookings Drive, Campus Box 1097, Saint Louis, MO, 63130-4899, USA
| |
Collapse
|
38
|
Tamate R, Ueki T, Yoshida R. Evolved Colloidosomes Undergoing Cell-like Autonomous Shape Oscillations with Buckling. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201511871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ryota Tamate
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
| | - Takeshi Ueki
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
- National Institute for Materials Science (NIMS); 1-1 Namiki, Tsukuba Ibaraki 305-0044 Japan
| | - Ryo Yoshida
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
| |
Collapse
|
39
|
Tamate R, Ueki T, Yoshida R. Evolved Colloidosomes Undergoing Cell-like Autonomous Shape Oscillations with Buckling. Angew Chem Int Ed Engl 2016; 55:5179-83. [DOI: 10.1002/anie.201511871] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/13/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Ryota Tamate
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
| | - Takeshi Ueki
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
- National Institute for Materials Science (NIMS); 1-1 Namiki, Tsukuba Ibaraki 305-0044 Japan
| | - Ryo Yoshida
- Department of Materials Engineering; School of Engineering; The University of Tokyo; 7-3-1 Hongo, Bunkyo-ku Tokyo 113-8656 Japan
| |
Collapse
|
40
|
Li A, Lai YC, Figueroa S, Yang T, Widelitz RB, Kobielak K, Nie Q, Chuong CM. Deciphering principles of morphogenesis from temporal and spatial patterns on the integument. Dev Dyn 2015; 244:905-20. [PMID: 25858668 PMCID: PMC4520785 DOI: 10.1002/dvdy.24281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/04/2015] [Accepted: 04/03/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND How tissue patterns form in development and regeneration is a fundamental issue remaining to be fully understood. The integument often forms repetitive units in space (periodic patterning) and time (cyclic renewal), such as feathers and hairs. Integument patterns are visible and experimentally manipulatable, helping us reveal pattern formative processes. Variability is seen in regional phenotypic specificities and temporal cycling at different physiological stages. RESULTS Here we show some cellular/molecular bases revealed by analyzing integument patterns. (1) Localized cellular activity (proliferation, rearrangement, apoptosis, differentiation) transforms prototypic organ primordia into specific shapes. Combinatorial positioning of different localized activity zones generates diverse and complex organ forms. (2) Competitive equilibrium between activators and inhibitors regulates stem cells through cyclic quiescence and activation. CONCLUSIONS Dynamic interactions between stem cells and their adjacent niche regulate regenerative behavior, modulated by multi-layers of macro-environmental factors (dermis, body hormone status, and external environment). Genomics studies may reveal how positional information of localized cellular activity is stored. In vivo skin imaging and lineage tracing unveils new insights into stem cell plasticity. Principles of self-assembly obtained from the integumentary organ model can be applied to help restore damaged patterns during regenerative wound healing and for tissue engineering to rebuild tissues. Developmental Dynamics 244:905-920, 2015. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ang Li
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Yung-Chih Lai
- Department of Pathology, University of Southern California, Los Angeles, California
- Center for Developmental Biology and Regenerative Medicine, Taiwan University, Taipei, Taiwan
| | - Seth Figueroa
- Department of Biomedical Engineering, University of California, Irvine, California
| | - Tian Yang
- Department of Cell Biology, College of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Randall B Widelitz
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Krzysztof Kobielak
- Department of Pathology, University of Southern California, Los Angeles, California
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, California
| | - Cheng Ming Chuong
- Department of Pathology, University of Southern California, Los Angeles, California
- Center for Developmental Biology and Regenerative Medicine, Taiwan University, Taipei, Taiwan
- Stem Cell and Regenerative Medicine Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
41
|
Kondo T, Hayashi S. Mechanisms of cell height changes that mediate epithelial invagination. Dev Growth Differ 2015; 57:313-23. [DOI: 10.1111/dgd.12224] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 04/24/2015] [Accepted: 04/26/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Takefumi Kondo
- Laboratory for Morphogenetic Signaling; RIKEN Center for Developmental Biology; Kobe Japan
| | - Shigeo Hayashi
- Laboratory for Morphogenetic Signaling; RIKEN Center for Developmental Biology; Kobe Japan
- Department of Biology; Kobe University Graduate School of Science; Kobe Japan
| |
Collapse
|
42
|
Abstract
Stefano Piccolo looks back at the life and research of his friend and colleague Yoshiki Sasai.
Collapse
Affiliation(s)
- Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, Via Colombo 3, Padova 35126, Italy
| |
Collapse
|
43
|
Gjorevski N, Ranga A, Lutolf MP. Bioengineering approaches to guide stem cell-based organogenesis. Development 2014; 141:1794-804. [PMID: 24757002 DOI: 10.1242/dev.101048] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During organogenesis, various molecular and physical signals are orchestrated in space and time to sculpt multiple cell types into functional tissues and organs. The complex and dynamic nature of the process has hindered studies aimed at delineating morphogenetic mechanisms in vivo, particularly in mammals. Recent demonstrations of stem cell-driven tissue assembly in culture offer a powerful new tool for modeling and dissecting organogenesis. However, despite the highly organotypic nature of stem cell-derived tissues, substantial differences set them apart from their in vivo counterparts, probably owing to the altered microenvironment in which they reside and the lack of mesenchymal influences. Advances in the biomaterials and microtechnology fields have, for example, afforded a high degree of spatiotemporal control over the cellular microenvironment, making it possible to interrogate the effects of individual microenvironmental components in a modular fashion and rapidly identify organ-specific synthetic culture models. Hence, bioengineering approaches promise to bridge the gap between stem cell-driven tissue formation in culture and morphogenesis in vivo, offering mechanistic insight into organogenesis and unveiling powerful new models for drug discovery, as well as strategies for tissue regeneration in the clinic. We draw on several examples of stem cell-derived organoids to illustrate how bioengineering can contribute to tissue formation ex vivo. We also discuss the challenges that lie ahead and potential ways to overcome them.
Collapse
Affiliation(s)
- Nikolche Gjorevski
- Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne CH-1015, Switzerland
| | | | | |
Collapse
|
44
|
Affiliation(s)
| | - Celeste M. Nelson
- Departments of 1Chemical & Biological Engineering and
- Molecular Biology, Princeton University, Princeton, New Jersey 08544;
| |
Collapse
|
45
|
Karus M, Blaess S, Brüstle O. Self-organization of neural tissue architectures from pluripotent stem cells. J Comp Neurol 2014; 522:2831-44. [PMID: 24737617 DOI: 10.1002/cne.23608] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 12/23/2022]
Abstract
Despite being a subject of intensive research, the mechanisms underlying the formation of neural tissue architectures during development of the central nervous system remain largely enigmatic. So far, studies into neural pattern formation have been restricted mainly to animal experiments. With the advent of pluripotent stem cells it has become possible to explore early steps of nervous system development in vitro. These studies have unraveled a remarkable propensity of primitive neural cells to self-organize into primitive patterns such as neural tube-like rosettes in vitro. Data from more advanced 3D culture systems indicate that this intrinsic propensity for self-organization can even extend to the formation of complex architectures such as a multilayered cortical neuroepithelium or an entire optic cup. These novel experimental paradigms not only demonstrate the enormous self-organization capacity of neural stem cells, they also provide exciting prospects for studying the earliest steps of human neural tissue development and the pathogenesis of brain malformations in reductionist in vitro paradigms.
Collapse
Affiliation(s)
- Michael Karus
- Institute of Reconstructive Neurobiology, University of Bonn LIFE&BRAIN Center, and LIFE&BRAIN GmbH, 53127, Bonn, Germany
| | | | | |
Collapse
|
46
|
Abstract
Pluripotent stem cells (PSCs) have the ability to spontaneously generate structured tissues in vitro reminiscent of embryonic tissue development. Recently, complex organoids such as cortical tissues, cerebral brain organoids, optical cups, intestinal tissues, and liver buds have been generated from PSCs derived from healthy individuals and patients with genetic diseases, providing powerful tools to understand morphogenesis and disease pathology. This article highlights recent advances in the state-of-art generation of organoids from PSCs, possible signaling pathways and mechanisms involved in organogenesis, and the understanding of extracellular microenvironment. Challenges involved in the organoid generation such as increasing organoid size, enhancing the tissue complexity, and improving functional maturation are also discussed.
Collapse
Affiliation(s)
- Yan Li
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering; Florida State University; Tallahassee, FL USA
| | - Chunhui Xu
- Department of Pediatrics; Emory University School of Medicine and Children's Healthcare of Atlanta; Atlanta, GA USA
| | - Teng Ma
- Department of Chemical and Biomedical Engineering; FAMU-FSU College of Engineering; Florida State University; Tallahassee, FL USA
| |
Collapse
|
47
|
Bozorgmehr JEH. The role of self-organization in developmental evolution. Theory Biosci 2014; 133:145-63. [PMID: 24737046 DOI: 10.1007/s12064-014-0200-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 03/06/2014] [Indexed: 01/09/2023]
Abstract
In developmental and evolutionary biology, particular emphasis has been given to the relationship between transcription factors and the cognate cis-regulatory elements of their target genes. These constitute the gene regulatory networks that control expression and are assumed to causally determine the formation of structures and body plans. Comparative analysis has, however, established a broad sequence homology among species that nonetheless display quite different anatomies. Transgenic experiments have also confirmed that many developmentally important elements are, in fact, functionally interchangeable. Although dependent upon the appropriate degree of gene expression, the actual construction of specific structures appears not directly linked to the functions of gene products alone. Instead, the self-formation of complex patterns, due in large part to epigenetic and non-genetic determinants, remains a persisting theme in the study of ontogeny and regenerative medicine. Recent evidence indeed points to the existence of a self-organizing process, operating through a set of intrinsic rules and forces, which imposes coordination and a holistic order upon cells and tissue. This has been repeatedly demonstrated in experiments on regeneration as well as in the autonomous formation of structures in vitro. The process cannot be wholly attributed to the functional outcome of protein-protein interactions or to concentration gradients of diffusible chemicals. This phenomenon is examined here along with some of the methodological and theoretical approaches that are now used in understanding the causal basis for self-organization in development and its evolution.
Collapse
|
48
|
Sasai Y. Next-generation regenerative medicine: organogenesis from stem cells in 3D culture. Cell Stem Cell 2014; 12:520-30. [PMID: 23642363 DOI: 10.1016/j.stem.2013.04.009] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The behavior of stem cells, when they work collectively, can be much more sophisticated than one might expect from their individual programming. This Perspective covers recent discoveries about the dynamic patterning and structural self-formation of complex organ buds in 3D stem cell culture, including the generation of various neuroectodermal and endodermal tissues. For some tissues, epithelial-mesenchymal interactions can also be manipulated in coculture to guide organogenesis. This new area of stem cell research-the spatiotemporal control of dynamic cellular interactions-will open a new avenue for next-generation regenerative medicine.
Collapse
Affiliation(s)
- Yoshiki Sasai
- Neurogenesis and Organogenesis Group, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| |
Collapse
|
49
|
Hägglund AC, Berghard A, Carlsson L. Canonical Wnt/β-catenin signalling is essential for optic cup formation. PLoS One 2013; 8:e81158. [PMID: 24324671 PMCID: PMC3852023 DOI: 10.1371/journal.pone.0081158] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/09/2013] [Indexed: 12/17/2022] Open
Abstract
A multitude of signalling pathways are involved in the process of forming an eye. Here we demonstrate that β-catenin is essential for eye development as inactivation of β-catenin prior to cellular specification in the optic vesicle caused anophthalmia in mice. By achieving this early and tissue-specific β-catenin inactivation we find that retinal pigment epithelium (RPE) commitment was blocked and eye development was arrested prior to optic cup formation due to a loss of canonical Wnt signalling in the dorsal optic vesicle. Thus, these results show that Wnt/β-catenin signalling is required earlier and play a more central role in eye development than previous studies have indicated. In our genetic model system a few RPE cells could escape β-catenin inactivation leading to the formation of a small optic rudiment. The optic rudiment contained several neural retinal cell classes surrounded by an RPE. Unlike the RPE cells, the neural retinal cells could be β-catenin-negative revealing that differentiation of the neural retinal cell classes is β-catenin-independent. Moreover, although dorsoventral patterning is initiated in the mutant optic vesicle, the neural retinal cells in the optic rudiment displayed almost exclusively ventral identity. Thus, β-catenin is required for optic cup formation, commitment to RPE cells and maintenance of dorsal identity of the retina.
Collapse
Affiliation(s)
| | - Anna Berghard
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Leif Carlsson
- Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
50
|
Apical contractility in growing epithelium supports robust maintenance of smooth curvatures against cell-division-induced mechanical disturbance. J Biomech 2013; 46:1705-13. [DOI: 10.1016/j.jbiomech.2013.03.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/16/2013] [Accepted: 03/31/2013] [Indexed: 01/08/2023]
|