51
|
Wang D, Lu Z, Xu Y, Wang ZI, Santella A, Bao Z. Cellular structure image classification with small targeted training samples. IEEE ACCESS : PRACTICAL INNOVATIONS, OPEN SOLUTIONS 2019; 7:148967-148974. [PMID: 32832309 PMCID: PMC7442139 DOI: 10.1109/access.2019.2940161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cell shapes provide crucial biological information on complex tissues. Different cell types often have distinct cell shapes, and collective shape changes usually indicate morphogenetic events and mechanisms. The identification and detection of collective cell shape changes in an extensive collection of 3D time-lapse images of complex tissues is an important step in assaying such mechanisms but is a tedious and time-consuming task. Machine learning provides new opportunities to automatically detect cell shape changes. However, it is challenging to generate sufficient training samples for pattern identification through deep learning because of a limited amount of images and annotations. We present a deep learning approach with minimal well-annotated training samples and apply it to identify multicellular rosettes from 3D live images of the Caenorhabditis elegans embryo with fluorescently labeled cell membranes. Our strategy is to combine two approaches, namely, feature transfer and generative adversarial networks (GANs), to boost image classification with small training samples. Specifically, we use a GAN framework and conduct an unsupervised training to capture the general characteristics of cell membrane images with 11,250 unlabelled images. We then transfer the structure of the GAN discriminator into a new Alex-style neural network for further learning with several dozen labeled samples. Our experiments showed that with 10-15 well-labeled rosette images and 30-40 randomly selected nonrosette images our approach can identify rosettes with more than 80% accuracy and capture more than 90% of the model accuracy achieved with a training data et that is five times larger. We also established a public benchmark dataset for rosette detection. This GAN-based transfer approach can be applied to the study of other cellular structures with minimal training samples.
Collapse
Affiliation(s)
- Dali Wang
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN 37934, USA
- Environmental Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN 37831, USA
| | - Zheng Lu
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN 37934, USA
| | - Yichi Xu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Z I Wang
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN 37934, USA
| | - Anthony Santella
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Zhirong Bao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| |
Collapse
|
52
|
Urasaki A, Morishita S, Naka K, Uozumi M, Abe K, Huang L, Watase E, Nakagawa O, Kawakami K, Matsui T, Bessho Y, Inagaki N. Shootins mediate collective cell migration and organogenesis of the zebrafish posterior lateral line system. Sci Rep 2019; 9:12156. [PMID: 31434971 PMCID: PMC6704158 DOI: 10.1038/s41598-019-48585-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2019] [Indexed: 01/23/2023] Open
Abstract
The zebrafish sensory posterior lateral line is an excellent model system to study collective cell migration and organogenesis. Shootin1 is a cytoplasmic protein involved in neuronal polarization and axon guidance. Previous studies have shown that shootin1 couples actin filament retrograde flow with extracellular adhesive substrates at the leading edge of axonal growth cones, thereby producing mechanical force for the migration and guidance of axonal growth cones. However, the functions of shootin in peripheral cells remain unknown. Here we identified two novel shootin family members, shootin2 and shootin3. In zebrafish, shootin1 and shootin3 are expressed in the posterior lateral line primordium (PLLP) and neuromasts during embryonic development. A shootin1 mutant displayed a reduced speed of PLLP migration, while shootin1;shootin3 double mutation inhibited cell proliferation in the PLLP. Furthermore, our results suggest that shootin1 and shootin3 positively regulate the number of neuromasts and the number of cells in deposited neuromasts. Our study demonstrates that shootins mediate collective cell migration of the posterior lateral line primordium and formation of neuromasts in zebrafish.
Collapse
Affiliation(s)
- Akihiro Urasaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.,Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Seiya Morishita
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kosuke Naka
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Minato Uozumi
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Kouki Abe
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Liguo Huang
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Emiko Watase
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-Shinmachi, Suita, Osaka, 564-8565, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, 411-8540, Japan
| | - Takaaki Matsui
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yasumasa Bessho
- Laboratory of Gene Regulation Research, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
53
|
Wenzel D, Praetorius S, Voigt A. Topological and geometrical quantities in active cellular structures. J Chem Phys 2019; 150:164108. [PMID: 31042877 DOI: 10.1063/1.5085766] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- D. Wenzel
- Institute of Scientific Computing, TU Dresden, 01062 Dresden, Germany
| | - S. Praetorius
- Institute of Scientific Computing, TU Dresden, 01062 Dresden, Germany
| | - A. Voigt
- Institute of Scientific Computing, TU Dresden, 01062 Dresden, Germany
- Dresden Center for Computational Materials Science (DCMS), TU Dresden, 01062 Dresden, Germany
- Center for Systems Biology Dresden (CSBD), Pfotenhauerstr. 108, 01307 Dresden, Germany
| |
Collapse
|
54
|
Lawton AK, Engstrom T, Rohrbach D, Omura M, Turnbull DH, Mamou J, Zhang T, Schwarz JM, Joyner AL. Cerebellar folding is initiated by mechanical constraints on a fluid-like layer without a cellular pre-pattern. eLife 2019; 8:e45019. [PMID: 30990415 PMCID: PMC6467563 DOI: 10.7554/elife.45019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/30/2019] [Indexed: 12/28/2022] Open
Abstract
Models based in differential expansion of elastic material, axonal constraints, directed growth, or multi-phasic combinations have been proposed to explain brain folding. However, the cellular and physical processes present during folding have not been defined. We used the murine cerebellum to challenge folding models with in vivo data. We show that at folding initiation differential expansion is created by the outer layer of proliferating progenitors expanding faster than the core. However, the stiffness differential, compressive forces, and emergent thickness variations required by elastic material models are not present. We find that folding occurs without an obvious cellular pre-pattern, that the outer layer expansion is uniform and fluid-like, and that the cerebellum is under radial and circumferential constraints. Lastly, we find that a multi-phase model incorporating differential expansion of a fluid outer layer and radial and circumferential constraints approximates the in vivo shape evolution observed during initiation of cerebellar folding.
Collapse
Affiliation(s)
- Andrew K Lawton
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
| | - Tyler Engstrom
- Department of PhysicsSyracuse UniversitySyracuseUnited States
| | - Daniel Rohrbach
- Lizzi Center for Biomedical EngineeringRiverside ResearchNew YorkUnited States
| | - Masaaki Omura
- Lizzi Center for Biomedical EngineeringRiverside ResearchNew YorkUnited States
- Department of Radiology, Skirball Institute of Biomolecular MedicineNYU School of MedicineNew YorkUnited States
- Graduate School of Science and EngineeringChiba UniversityChibaJapan
| | - Daniel H Turnbull
- Department of Radiology, Skirball Institute of Biomolecular MedicineNYU School of MedicineNew YorkUnited States
| | - Jonathan Mamou
- Lizzi Center for Biomedical EngineeringRiverside ResearchNew YorkUnited States
| | - Teng Zhang
- Department of Mechanical & Aerospace EngineeringSyracuse UniversitySyracuseUnited States
| | - J M Schwarz
- Department of PhysicsSyracuse UniversitySyracuseUnited States
| | - Alexandra L Joyner
- Developmental Biology ProgramSloan Kettering InstituteNew YorkUnited States
- Biochemistry, Cell and Molecular Biology Program, Weill Graduate School of Medical SciencesCornell UniversityNew YorkUnited States
| |
Collapse
|
55
|
Improved Efficiency of Cardiomyocyte-Like Cell Differentiation from Rat Adipose Tissue-Derived Mesenchymal Stem Cells with a Directed Differentiation Protocol. Stem Cells Int 2019; 2019:8940365. [PMID: 31065283 PMCID: PMC6466858 DOI: 10.1155/2019/8940365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/06/2018] [Accepted: 12/20/2018] [Indexed: 01/29/2023] Open
Abstract
Cell-based therapy has become a resource for the treatment of cardiovascular diseases; however, there are some conundrums to achieve. In vitro cardiomyocyte generation could be a solution for scaling options in clinical applications. Variability on cardiac differentiation in previously reported studies from adipose tissue-derived mesenchymal stem cells (ASCs) and the lack of measuring of the cardiomyocyte differentiation efficiency motivate the present study. Here, we improved the ASC-derived cardiomyocyte-like cell differentiation efficiency with a directed cardiomyocyte differentiation protocol: BMP-4 + VEGF (days 0-4) followed by a methylcellulose-based medium with cytokines (IL-6 and IL-3) (days 5-21). Cultures treated with the directed cardiomyocyte differentiation protocol showed cardiac-like cells and “rosette-like structures” from day 7. The percentage of cardiac troponin T- (cTnT-) positive cells was evaluated by flow cytometry to assess the cardiomyocyte differentiation efficiency in a quantitative manner. ASCs treated with the directed cardiomyocyte differentiation protocol obtained a differentiation efficiency of up to 44.03% (39.96%±3.78) at day 15 without any enrichment step. Also, at day 21 we observed by immunofluorescence the positive expression of early, late, and cardiac maturation differentiation markers (Gata-4, cTnT, cardiac myosin heavy chain (MyH), and the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCa2)) in cultures treated with the directed cardiomyocyte differentiation protocol. Unlike other protocols, the use of critical factors of embryonic cardiomyogenesis coupled with a methylcellulose-based medium containing previously reported cardiogenic cytokines (IL-6 and IL-3) seems to be favorable for in vitro cardiomyocyte generation. This novel efficient culture protocol makes ASC-derived cardiac differentiation more efficient. Further investigation is needed to identify an ASC-derived cardiomyocyte surface marker for cardiac enrichment.
Collapse
|
56
|
Fan L, Kovacevic I, Heiman MG, Bao Z. A multicellular rosette-mediated collective dendrite extension. eLife 2019; 8:38065. [PMID: 30767892 PMCID: PMC6400498 DOI: 10.7554/elife.38065] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
Coordination of neurite morphogenesis with surrounding tissues is crucial to the establishment of neural circuits, but the underlying cellular and molecular mechanisms remain poorly understood. We show that neurons in a C. elegans sensory organ, called the amphid, undergo a collective dendrite extension to form the sensory nerve. The amphid neurons first assemble into a multicellular rosette. The vertex of the rosette, which becomes the dendrite tips, is attached to the anteriorly migrating epidermis and carried to the sensory depression, extruding the dendrites away from the neuronal cell bodies. Multiple adhesion molecules including DYF-7, SAX-7, HMR-1 and DLG-1 function redundantly in rosette-to-epidermis attachment. PAR-6 is localized to the rosette vertex and dendrite tips, and promotes DYF-7 localization and dendrite extension. Our results suggest a collective mechanism of neurite extension that is distinct from the classical pioneer-follower model and highlight the role of mechanical cues from surrounding tissues in shaping neurites.
Collapse
Affiliation(s)
- Li Fan
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Ismar Kovacevic
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| | - Maxwell G Heiman
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, United States.,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, United States
| | - Zhirong Bao
- Developmental Biology Program, Sloan Kettering Institute, New York, United States
| |
Collapse
|
57
|
Calamari ZT, Kuang-Hsien Hu J, Klein OD. Tissue Mechanical Forces and Evolutionary Developmental Changes Act Through Space and Time to Shape Tooth Morphology and Function. Bioessays 2018; 40:e1800140. [PMID: 30387177 PMCID: PMC6516060 DOI: 10.1002/bies.201800140] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/06/2018] [Indexed: 12/24/2022]
Abstract
Efforts from diverse disciplines, including evolutionary studies and biomechanical experiments, have yielded new insights into the genetic, signaling, and mechanical control of tooth formation and functions. Evidence from fossils and non-model organisms has revealed that a common set of genes underlie tooth-forming potential of epithelia, and changes in signaling environments subsequently result in specialized dentitions, maintenance of dental stem cells, and other phenotypic adaptations. In addition to chemical signaling, tissue forces generated through epithelial contraction, differential growth, and skeletal constraints act in parallel to shape the tooth throughout development. Here recent advances in understanding dental development from these studies are reviewed and important gaps that can be filled through continued application of evolutionary and biomechanical approaches are discussed.
Collapse
Affiliation(s)
- Zachary T. Calamari
- Department of Natural Sciences, Baruch College, City University of New York, New York City, New York, 10010, USA
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Jimmy Kuang-Hsien Hu
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, California, 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
58
|
Sequential formation and resolution of multiple rosettes drive embryo remodelling after implantation. Nat Cell Biol 2018; 20:1278-1289. [PMID: 30323188 DOI: 10.1038/s41556-018-0211-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/06/2018] [Indexed: 11/08/2022]
Abstract
The morphogenetic remodelling of embryo architecture after implantation culminates in pro-amniotic cavity formation. Despite its key importance, how this transformation occurs remains unknown. Here, we apply high-resolution imaging of embryos developing in vivo and in vitro, spatial RNA sequencing and 3D trophoblast stem cell models to determine the sequence and mechanisms of these remodelling events. We show that cavitation of the embryonic tissue is followed by folding of extra-embryonic tissue to mediate the formation of a second extra-embryonic cavity. Concomitantly, at the boundary between embryonic and extra-embryonic tissues, a hybrid 3D rosette forms. Resolution of this rosette enables the embryonic cavity to invade the extra-embryonic tissue. Subsequently, β1-integrin signalling mediates the formation of multiple extra-embryonic 3D rosettes. Podocalyxin exocytosis leads to their polarized resolution, permitting the extension of embryonic and extra-embryonic cavities and their fusion into a unified pro-amniotic cavity. These morphogenetic transformations of embryogenesis reveal a previously unappreciated mechanism for lumen expansion and fusion.
Collapse
|
59
|
Neural stem cells deriving from chick embryonic hindbrain recapitulate hindbrain development in culture. Sci Rep 2018; 8:13920. [PMID: 30224755 PMCID: PMC6141497 DOI: 10.1038/s41598-018-32203-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
Neural stem cells (NSCs) are self-renewing multipotent cells that line the neural-tube and generate all the nervous system. Understanding NSC biology is fundamental for neurodevelopmental research and therapy. Many studies emphasized the need to culture NSCs, which are typically purified from mammalian embryonic/adult brains. These sources are somewhat limited in terms of quantity, availability and animal ethical guidelines. Therefore, new sources are needed. The chick is a powerful system for experimental embryology which contributed enormously to neurodevelopmental concepts. Its accessibility, genetic/molecular manipulations, and homology to other vertebrates, makes it valuable for developmental biology research. Recently, we identified a population of NSCs in the chick hindbrain. It resides in rhombomere-boundaries, expresses Sox2 and generates progenitors and neurons. Here, we investigated whether these cells can recapitulate hindbrain development in culture. By developing approaches to propagate and image cells, manipulate their growth-conditions and separate them into subpopulations, we demonstrate the ordered formation of multipotent and self-renewing neurospheres that maintain regional identity and display differential stem/differentiation/proliferation properties. Live imaging revealed new cellular dynamics in the culture. Collectively, these NSC cultures reproduce major aspects of hindbrain development in-vitro, proposing the chick as a model for culturing hindbrain-NSCs that can be directly applied to other neural-tube domains and species.
Collapse
|
60
|
Blanchard GB, Fletcher AG, Schumacher LJ. The devil is in the mesoscale: Mechanical and behavioural heterogeneity in collective cell movement. Semin Cell Dev Biol 2018; 93:46-54. [PMID: 29940338 DOI: 10.1016/j.semcdb.2018.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/15/2022]
Abstract
Heterogeneity within cell populations can be an important aspect affecting their collective movement and tissue-mechanical properties, determining for example their effective viscoelasticity. Differences in cell-level properties and behaviour within a group of moving cells can give rise to unexpected and non-intuitive behaviours at the tissue level. Such emergent phenomena often manifest themselves through spatiotemporal patterns at an intermediate 'mesoscale' between cell and tissue scales, typically involving tens of cells. Focussing on the development of embryonic animal tissues, we review recent evidence for the importance of heterogeneity at the mesoscale for collective cell migration and convergence and extension movements. We further discuss approaches to incorporate heterogeneity into computational models to complement experimental investigations.
Collapse
Affiliation(s)
- Guy B Blanchard
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK.
| | - Alexander G Fletcher
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield, S3 7RH, UK; Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| | - Linus J Schumacher
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
61
|
Smith EE, Angstadt S, Monteiro N, Zhang W, Khademhosseini A, Yelick PC. Bioengineered Tooth Buds Exhibit Features of Natural Tooth Buds. J Dent Res 2018; 97:1144-1151. [PMID: 29879370 DOI: 10.1177/0022034518779075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tooth loss is a significant health issue currently affecting millions of people worldwide. Artificial dental implants, the current gold standard tooth replacement therapy, do not exhibit many properties of natural teeth and can be associated with complications leading to implant failure. Here we propose bioengineered tooth buds as a superior alternative tooth replacement therapy. We describe improved methods to create highly cellularized bioengineered tooth bud constructs that formed hallmark features that resemble natural tooth buds such as the dental epithelial stem cell niche, enamel knot signaling centers, transient amplifying cells, and mineralized dental tissue formation. These constructs were composed of postnatal dental cells encapsulated within a hydrogel material that were implanted subcutaneously into immunocompromised rats. To our knowledge, this is the first report describing the use of postnatal dental cells to create bioengineered tooth buds that exhibit evidence of these features of natural tooth development. We propose future bioengineered tooth buds as a promising, clinically relevant tooth replacement therapy.
Collapse
Affiliation(s)
- E E Smith
- 1 Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School Medicine, Boston, MA, USA
| | - S Angstadt
- 2 Department of Orthodontics, Tufts University School of Dental Medicine, Boston, MA, USA
| | - N Monteiro
- 2 Department of Orthodontics, Tufts University School of Dental Medicine, Boston, MA, USA
| | - W Zhang
- 2 Department of Orthodontics, Tufts University School of Dental Medicine, Boston, MA, USA
| | - A Khademhosseini
- 3 Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - P C Yelick
- 1 Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School Medicine, Boston, MA, USA.,2 Department of Orthodontics, Tufts University School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
62
|
Chiasson-MacKenzie C, McClatchey AI. Cell-Cell Contact and Receptor Tyrosine Kinase Signaling. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029215. [PMID: 28716887 DOI: 10.1101/cshperspect.a029215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The behavior of cells within tissues is governed by the activities of adhesion receptors that provide spatial cues and transmit forces through intercellular junctions, and by growth-factor receptors, particularly receptor tyrosine kinases (RTKs), that respond to biochemical signals from the environment. Coordination of these two activities is essential for the patterning and polarized migration of cells during morphogenesis and for homeostasis in mature tissues; loss of this coordination is a hallmark of developing cancer and driver of metastatic progression. Although much is known about the individual functions of adhesion and growth factor receptors, we have a surprisingly superficial understanding of the mechanisms by which their activities are coordinated.
Collapse
Affiliation(s)
- Christine Chiasson-MacKenzie
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Departments of Pathology, Charlestown, Massachusetts 02129
| | - Andrea I McClatchey
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Departments of Pathology, Charlestown, Massachusetts 02129
| |
Collapse
|
63
|
Medelnik JP, Roensch K, Okawa S, Del Sol A, Chara O, Mchedlishvili L, Tanaka EM. Signaling-Dependent Control of Apical Membrane Size and Self-Renewal in Rosette-Stage Human Neuroepithelial Stem Cells. Stem Cell Reports 2018; 10:1751-1765. [PMID: 29779899 PMCID: PMC5993681 DOI: 10.1016/j.stemcr.2018.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
In the developing nervous system, neural stem cells are polarized and maintain an apical domain facing a central lumen. The presence of apical membrane is thought to have a profound influence on maintaining the stem cell state. With the onset of neurogenesis, cells lose their polarization, and the concomitant loss of the apical domain coincides with a loss of the stem cell identity. Little is known about the molecular signals controlling apical membrane size. Here, we use two neuroepithelial cell systems, one derived from regenerating axolotl spinal cord and the other from human embryonic stem cells, to identify a molecular signaling pathway initiated by lysophosphatidic acid that controls apical membrane size and consequently controls and maintains epithelial organization and lumen size in neuroepithelial rosettes. This apical domain size increase occurs independently of effects on proliferation and involves a serum response factor-dependent transcriptional induction of junctional and apical membrane components.
Collapse
Affiliation(s)
- Jan-Philip Medelnik
- Research Institute for Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| | - Kathleen Roensch
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux 4367, Luxembourg
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux 4367, Luxembourg
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing (ZIH), Technische Universität Dresden, 01062 Dresden, Germany; Systems Biology Group (SysBio), Instituto de Física de Líquidos y Sistemas Biológicos (IFLySIB), CONICET, Universidad Nacional de La Plata (UNLP), B1900BTE, La Plata, Argentina
| | - Levan Mchedlishvili
- DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Elly M Tanaka
- Research Institute for Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria; DFG Research Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| |
Collapse
|
64
|
López-Escobar B, Caro-Vega JM, Vijayraghavan DS, Plageman TF, Sanchez-Alcazar JA, Moreno RC, Savery D, Márquez-Rivas J, Davidson LA, Ybot-González P. The non-canonical Wnt-PCP pathway shapes the mouse caudal neural plate. Development 2018; 145:dev.157487. [PMID: 29636380 DOI: 10.1242/dev.157487] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/03/2018] [Indexed: 01/03/2023]
Abstract
The last stage of neural tube (NT) formation involves closure of the caudal neural plate (NP), an embryonic structure formed by neuromesodermal progenitors and newly differentiated cells that becomes incorporated into the NT. Here, we show in mouse that, as cell specification progresses, neuromesodermal progenitors and their progeny undergo significant changes in shape prior to their incorporation into the NT. The caudo-rostral progression towards differentiation is coupled to a gradual reliance on a unique combination of complex mechanisms that drive tissue folding, involving pulses of apical actomyosin contraction and planar polarised cell rearrangements, all of which are regulated by the Wnt-PCP pathway. Indeed, when this pathway is disrupted, either chemically or genetically, the polarisation and morphology of cells within the entire caudal NP is disturbed, producing delays in NT closure. The most severe disruptions of this pathway prevent caudal NT closure and result in spina bifida. In addition, a decrease in Vangl2 gene dosage also appears to promote more rapid progression towards a neural fate, but not the specification of more neural cells.
Collapse
Affiliation(s)
- Beatriz López-Escobar
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - José Manuel Caro-Vega
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | | | | | - José A Sanchez-Alcazar
- Centro Andaluz de Biología del Desarrollo (CABD), and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC, Sevilla 41013, Spain
| | - Roberto Carlos Moreno
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Dawn Savery
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Javier Márquez-Rivas
- Unidad de Gestión Clínica de Neurocirugía, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain
| | - Lance A Davidson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Patricia Ybot-González
- Grupo de Neurodesarrollo, Hospital Universitario Virgen del Rocio/Instituto de Biomedicina de Sevilla (IBIS)/CSIC/Universidad de Sevilla, Sevilla 41013, Spain .,Unidad de Gestión Clínica de Neurología y Neurofisiología, Hospital Universitario Virgen Macarena, Sevilla 41009, Spain
| |
Collapse
|
65
|
Hříbková H, Grabiec M, Klemová D, Slaninová I, Sun YM. Five steps to form neural rosettes: structure and function. J Cell Sci 2018; 131:jcs.206896. [DOI: 10.1242/jcs.206896] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/18/2017] [Indexed: 12/25/2022] Open
Abstract
Neural rosette formation is a critical morphogenetic process during neural development, whereby neural stem cells are enclosed in rosette niches to equipoise proliferation and differentiation. How neural rosettes form and provide a regulatory micro-environment remains to be elucidated. We employed the human embryonic stem cell-based neural rosette system to investigate the structural development and function of neural rosettes. Our study shows that neural rosette formation consists of 5 types of cell movements: intercalation, constriction, polarization, elongation, and lumen formation. Ca2+ signaling plays a pivotal role in the five steps by regulating the actions of the cytoskeletal complexes, ACTIN, MYOSIN II, and TUBULIN during intercalation, constriction, and elongation. These in turn control the polarizing elements, ZO-1, PARD3, and β-CATENIN during polarization and lumen formation in neural rosette formation. We further demonstrated that the dismantlement of neural rosettes, mediated by the destruction of cytoskeletal elements, promoted neurogenesis and astrogenesis prematurely, indicating that an intact rosette structure is essential for orderly neural development.
Collapse
Affiliation(s)
- Hana Hříbková
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marta Grabiec
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dobromila Klemová
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Iva Slaninová
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Yuh-Man Sun
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
66
|
Méndez-Olivos EE, Muñoz R, Larraín J. Spinal Cord Cells from Pre-metamorphic Stages Differentiate into Neurons and Promote Axon Growth and Regeneration after Transplantation into the Injured Spinal Cord of Non-regenerative Xenopus laevis Froglets. Front Cell Neurosci 2017; 11:398. [PMID: 29326551 PMCID: PMC5733487 DOI: 10.3389/fncel.2017.00398] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/28/2017] [Indexed: 11/13/2022] Open
Abstract
Mammals are unable to regenerate its spinal cord after a lesion, meanwhile, anuran amphibians are capable of spinal cord regeneration only as larvae, and during metamorphosis, this capability is lost. Sox2/3+ cells present in the spinal cord of regenerative larvae are required for spinal cord regeneration. Here we evaluate the effect of the transplantation of spinal cord cells from regenerative larvae into the resected spinal cord of non-regenerative stages (NR-stage). Donor cells were able to survive up to 60 days after transplantation in the injury zone. During the first 3-weeks, transplanted cells organize in neural tube-like structures formed by Sox2/3+ cells. This was not observed when donor cells come from non-regenerative froglets. Mature neurons expressing NeuN and Neurofilament-H were detected in the grafted tissue 4 weeks after transplantation concomitantly with the appearance of axons derived from the donor cells growing into the host spinal cord, suggesting that Sox2/3+ cells behave as neural stem progenitor cells. We also found that cells from regenerative animals provide a permissive environment that promotes growth and regeneration of axons coming from the host. These results suggest that Sox2/3 cells present in the spinal cord of regenerative stage (R-stage) larvae are most probably neural stem progenitor cells that are able to survive, proliferate, self-organize and differentiate into neurons in the environment of the non-regenerative host. In addition, we have established an experimental paradigm to study the biology of neural stem progenitor cells in spinal cord regeneration.
Collapse
Affiliation(s)
- Emilio E Méndez-Olivos
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rosana Muñoz
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
67
|
The mouse Jhy gene regulates ependymal cell differentiation and ciliogenesis. PLoS One 2017; 12:e0184957. [PMID: 29211732 PMCID: PMC5718522 DOI: 10.1371/journal.pone.0184957] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
During the first postnatal week of mouse development, radial glial cells lining the ventricles of the brain differentiate into ependymal cells, undergoing a morphological change from pseudostratified cuboidal cells to a flattened monolayer. Concomitant with this change, multiple motile cilia are generated and aligned on each nascent ependymal cell. Proper ependymal cell development is crucial to forming the brain tissue:CSF barrier, and to the establishment of ciliary CSF flow, but the mechanisms that regulate this differentiation event are poorly understood. The JhylacZ mouse line carries an insertional mutation in the Jhy gene (formerly 4931429I11Rik), and homozygous JhylacZ/lacZ mice develop a rapidly progressive juvenile hydrocephalus, with defects in ependymal cilia morphology and ultrastructure. Here we show that beyond just defective motile cilia, JhylacZ/lacZ mice display abnormal ependymal cell differentiation. Ventricular ependyma in JhylacZ/lacZ mice retain an unorganized and multi-layered morphology, representative of undifferentiated ependymal (radial glial) cells, and they show altered expression of differentiation markers. Most JhylacZ/lacZ ependymal cells do eventually acquire some differentiated ependymal characteristics, suggesting a delay, rather than a block, in the differentiation process, but ciliogenesis remains perturbed. JhylacZ/lacZ ependymal cells also manifest disruptions in adherens junction formation, with altered N-cadherin localization, and have defects in the polarized organization of the apical motile cilia that do form. Functional studies showed that cilia of JhylacZ/lacZ mice have severely reduced motility, a potential cause for the development of hydrocephalus. This work shows that JHY does not only control ciliogenesis, but is a crucial component of the ependymal differentiation process, with ciliary defects likely a consequence of altered ependymal differentiation.
Collapse
|
68
|
Harris TJC. Sculpting epithelia with planar polarized actomyosin networks: Principles from Drosophila. Semin Cell Dev Biol 2017; 81:54-61. [PMID: 28760393 DOI: 10.1016/j.semcdb.2017.07.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 02/04/2023]
Abstract
Drosophila research has revealed how planar polarized actomyosin networks affect various types of tissue morphogenesis. The networks are positioned by both tissue-wide patterning factors (including Even-skipped, Runt, Engrailed, Invected, Hedgehog, Notch, Wingless, Epidermal Growth Factor, Jun N-terminal kinase, Sex combs reduced and Fork head) and local receptor complexes (including Echinoid, Crumbs and Toll receptors). Networks with differing super-structure and contractile output have been discovered. Their contractility can affect individual cells or can be coordinated across groups of cells, and such contractility can drive or resist physical change. For what seem to be simple tissue changes, multiple types of actomyosin networks can contribute, acting together as contractile elements or braces within the developing structure. This review discusses the positioning and effects of planar polarized actomyosin networks for a number of developmental events in Drosophila, including germband extension, dorsal closure, head involution, tracheal pit formation, salivary gland development, imaginal disc boundary formation, and tissue rotation of the male genitalia and the egg chamber.
Collapse
Affiliation(s)
- Tony J C Harris
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
69
|
Yoshida S, Kato T, Kanno N, Nishimura N, Nishihara H, Horiguchi K, Kato Y. Cell type-specific localization of Ephs pairing with ephrin-B2 in the rat postnatal pituitary gland. Cell Tissue Res 2017; 370:99-112. [PMID: 28660300 DOI: 10.1007/s00441-017-2646-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 05/06/2017] [Indexed: 01/20/2023]
Abstract
Sox2-expressing stem/progenitor cells in the anterior lobe of the pituitary gland form two types of micro-environments (niches): the marginal cell layer and dense cell clusters in the parenchyma. In relation to the mechanism of regulation of niches, juxtacrine signaling via ephrin and its receptor Eph is known to play important roles in various niches. The ephrin and Eph families are divided into two subclasses to create ephrin/Eph signaling in co-operation with confined partners. Recently, we reported that ephrin-B2 localizes specifically to both pituitary niches. However, the Ephs interacting with ephrin-B2 in these pituitary niches have not yet been identified. Therefore, the present study aims to identify the Ephs interacting with ephrin-B2 and the cells that produce them in the rat pituitary gland. In situ hybridization and immunohistochemistry demonstrated cell type-specific localization of candidate interacting partners for ephrin-B2, including EphA4 in cells located in the posterior lobe, EphB1 in gonadotropes, EphB2 in corticotropes, EphB3 in stem/progenitor cells and EphB4 in endothelial cells in the adult pituitary gland. In particular, double-immunohistochemistry showed cis-interactions between EphB3 and ephrin-B2 in the apical cell membranes of stem/progenitor cell niches throughout life and trans-interactions between EphB2 produced by corticotropes and ephrin-B2 located in the basolateral cell membranes of stem/progenitor cells in the early postnatal pituitary gland. These data indicate that ephrin-B2 plays a role in pituitary stem/progenitor cell niches by selective interaction with EphB3 in cis and EphB2 in trans.
Collapse
Affiliation(s)
- Saishu Yoshida
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa, Japan.,Institute of Reproduction and Endocrinology, Meiji University, Kanagawa, Japan
| | - Takako Kato
- Institute of Reproduction and Endocrinology, Meiji University, Kanagawa, Japan
| | - Naoko Kanno
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa, Japan
| | - Naoto Nishimura
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa, Japan
| | - Hiroto Nishihara
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa, Japan
| | - Kotaro Horiguchi
- Laboratory of Anatomy and Cell Biology, Department of Health Sciences, Kyorin University, Tokyo, Japan
| | - Yukio Kato
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa, Japan. .,Institute of Reproduction and Endocrinology, Meiji University, Kanagawa, Japan. .,Department of Life Science, School of Agriculture, Meiji University, Kanagawa, Japan.
| |
Collapse
|
70
|
Gao Q, Zhang J, Wang X, Liu Y, He R, Liu X, Wang F, Feng J, Yang D, Wang Z, Meng A, Yan X. The signalling receptor MCAM coordinates apical-basal polarity and planar cell polarity during morphogenesis. Nat Commun 2017; 8:15279. [PMID: 28589943 PMCID: PMC5467231 DOI: 10.1038/ncomms15279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
The apical-basal (AB) polarity and planar cell polarity (PCP) provide an animal cell population with different phenotypes during morphogenesis. However, how cells couple these two patterning systems remains unclear. Here we provide in vivo evidence that melanoma cell adhesion molecule (MCAM) coordinates AB polarity-driven lumenogenesis and c-Jun N-terminal kinase (JNK)/PCP-dependent ciliogenesis. We identify that MCAM is an independent receptor of fibroblast growth factor 4 (FGF4), a membrane anchor of phospholipase C-γ (PLC-γ), an immediate upstream receptor of nuclear factor of activated T-cells (NFAT) and a constitutive activator of JNK. We find that MCAM-mediated vesicular trafficking towards FGF4, while generating a priority-grade transcriptional response of NFAT determines lumenogenesis. We demonstrate that MCAM plays indispensable roles in ciliogenesis through activating JNK independently of FGF signals. Furthermore, mcam-deficient zebrafish and Xenopus exhibit a global defect in left-right (LR) asymmetric establishment as a result of morphogenetic failure of their LR organizers. Therefore, MCAM coordination of AB polarity and PCP provides insight into the general mechanisms of morphogenesis.
Collapse
Affiliation(s)
- Qian Gao
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junfeng Zhang
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiumei Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ying Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Rongqiao He
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingfeng Liu
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fei Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Feng
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongling Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhaoqing Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Anming Meng
- State Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
71
|
Barton DL, Henkes S, Weijer CJ, Sknepnek R. Active Vertex Model for cell-resolution description of epithelial tissue mechanics. PLoS Comput Biol 2017; 13:e1005569. [PMID: 28665934 PMCID: PMC5493290 DOI: 10.1371/journal.pcbi.1005569] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
We introduce an Active Vertex Model (AVM) for cell-resolution studies of the mechanics of confluent epithelial tissues consisting of tens of thousands of cells, with a level of detail inaccessible to similar methods. The AVM combines the Vertex Model for confluent epithelial tissues with active matter dynamics. This introduces a natural description of the cell motion and accounts for motion patterns observed on multiple scales. Furthermore, cell contacts are generated dynamically from positions of cell centres. This not only enables efficient numerical implementation, but provides a natural description of the T1 transition events responsible for local tissue rearrangements. The AVM also includes cell alignment, cell-specific mechanical properties, cell growth, division and apoptosis. In addition, the AVM introduces a flexible, dynamically changing boundary of the epithelial sheet allowing for studies of phenomena such as the fingering instability or wound healing. We illustrate these capabilities with a number of case studies.
Collapse
Affiliation(s)
- Daniel L. Barton
- Division of Physics, School of Science and Engineering, University of Dundee, Dundee, United Kingdom
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Silke Henkes
- Institute of Complex Systems and Mathematical Biology, Department of Physics, University of Aberdeen, Aberdeen, United Kingdom
| | - Cornelis J. Weijer
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Rastko Sknepnek
- Division of Physics, School of Science and Engineering, University of Dundee, Dundee, United Kingdom
- Division of Computational Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
72
|
Abstract
Planar cell polarity (PCP) signaling orients developmental events in vertebrates and invertebrates, including convergent extension (CE). In this issue of Development Cell, Shah and Tanner et al. (2017) report that ROBO/SAX-3 signaling acts in parallel with PCP signaling to drive the CE required for ventral nerve cord assembly in C. elegans.
Collapse
Affiliation(s)
- Martha C Soto
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| |
Collapse
|
73
|
Shah PK, Tanner MR, Kovacevic I, Rankin A, Marshall TE, Noblett N, Tran NN, Roenspies T, Hung J, Chen Z, Slatculescu C, Perkins TJ, Bao Z, Colavita A. PCP and SAX-3/Robo Pathways Cooperate to Regulate Convergent Extension-Based Nerve Cord Assembly in C. elegans. Dev Cell 2017; 41:195-203.e3. [PMID: 28441532 PMCID: PMC5469364 DOI: 10.1016/j.devcel.2017.03.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 02/08/2017] [Accepted: 03/29/2017] [Indexed: 10/19/2022]
Abstract
Formation and resolution of multicellular rosettes can drive convergent extension (CE) type cell rearrangements during tissue morphogenesis. Rosette dynamics are regulated by both planar cell polarity (PCP)-dependent and -independent pathways. Here we show that CE is involved in ventral nerve cord (VNC) assembly in Caenorhabditis elegans. We show that a VANG-1/Van Gogh and PRKL-1/Prickle containing PCP pathway and a Slit-independent SAX-3/Robo pathway cooperate to regulate, via rosette intermediaries, the intercalation of post-mitotic neuronal cell bodies during VNC formation. We show that VANG-1 and SAX-3 are localized to contracting edges and rosette foci and act to specify edge contraction during rosette formation and to mediate timely rosette resolution. Simultaneous loss of both pathways severely curtails CE resulting in a shortened, anteriorly displaced distribution of VNC neurons at hatching. Our results establish rosette-based CE as an evolutionarily conserved mechanism of nerve cord morphogenesis and reveal a role for SAX-3/Robo in this process.
Collapse
Affiliation(s)
- Pavak K Shah
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Matthew R Tanner
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Ismar Kovacevic
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Aysha Rankin
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Teagan E Marshall
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Nathaniel Noblett
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Nhan Nguyen Tran
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA
| | - Tony Roenspies
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Jeffrey Hung
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Zheqian Chen
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Cristina Slatculescu
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Theodore J Perkins
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Zhirong Bao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY 10065, USA.
| | - Antonio Colavita
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
74
|
Kozlovskaja-Gumbrienė A, Yi R, Alexander R, Aman A, Jiskra R, Nagelberg D, Knaut H, McClain M, Piotrowski T. Proliferation-independent regulation of organ size by Fgf/Notch signaling. eLife 2017; 6. [PMID: 28085667 PMCID: PMC5235355 DOI: 10.7554/elife.21049] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/23/2016] [Indexed: 12/31/2022] Open
Abstract
Organ morphogenesis depends on the precise orchestration of cell migration, cell shape changes and cell adhesion. We demonstrate that Notch signaling is an integral part of the Wnt and Fgf signaling feedback loop coordinating cell migration and the self-organization of rosette-shaped sensory organs in the zebrafish lateral line system. We show that Notch signaling acts downstream of Fgf signaling to not only inhibit hair cell differentiation but also to induce and maintain stable epithelial rosettes. Ectopic Notch expression causes a significant increase in organ size independently of proliferation and the Hippo pathway. Transplantation and RNASeq analyses revealed that Notch signaling induces apical junctional complex genes that regulate cell adhesion and apical constriction. Our analysis also demonstrates that in the absence of patterning cues normally provided by a Wnt/Fgf signaling system, rosettes still self-organize in the presence of Notch signaling. DOI:http://dx.doi.org/10.7554/eLife.21049.001
Collapse
Affiliation(s)
| | - Ren Yi
- Stowers Institute for Medical Research, Kansas City, United States
| | | | - Andy Aman
- Stowers Institute for Medical Research, Kansas City, United States
| | - Ryan Jiskra
- Stowers Institute for Medical Research, Kansas City, United States
| | - Danielle Nagelberg
- Developmental Genetics Program and Kimmel Center for Stem Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, United States
| | - Holger Knaut
- Developmental Genetics Program and Kimmel Center for Stem Cell Biology, Skirball Institute of Biomolecular Medicine, New York University Langone Medical Center, New York, United States
| | - Melainia McClain
- Stowers Institute for Medical Research, Kansas City, United States
| | | |
Collapse
|
75
|
Gayathri Vegesna NV, Ronchi P, Durdu S, Terjung S, Pepperkok R. Targeted Ablation Using Laser Nanosurgery. Methods Mol Biol 2017; 1563:107-125. [PMID: 28324605 DOI: 10.1007/978-1-4939-6810-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Laser-mediated dissection methods have been used for many years to micro-irradiate biological samples, but recent technological progress has rendered this technique more precise, powerful, and easy to use. Today pulsed lasers can be operated with diffraction limited, sub-micrometer precision to ablate intracellular structures. Here, we discuss laser nanosurgery setups and the instrumentation in our laboratory. We describe how to use this technique to ablate cytoskeletal elements in living cells. We also show how this technique can be used in multicellular organisms, to micropuncture and/or ablate cells of interest and finally how to monitor a successful laser nanosurgery.
Collapse
Affiliation(s)
| | - Paolo Ronchi
- Cell Biology and Cell Biophysics Unit, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany.,Electron Microscopy Core Facility, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Sevi Durdu
- Cell Biology and Cell Biophysics Unit, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Stefan Terjung
- Advanced Light Microscopy Facility, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Cell Biophysics Unit, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany. .,Advanced Light Microscopy Facility, EMBL Heidelberg, Meyerhofstrasse 1, 69117, Heidelberg, Germany.
| |
Collapse
|
76
|
Marciano DK. A holey pursuit: lumen formation in the developing kidney. Pediatr Nephrol 2017; 32:7-20. [PMID: 26902755 PMCID: PMC5495142 DOI: 10.1007/s00467-016-3326-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 02/07/2023]
Abstract
The formation of polarized epithelial tubules is a hallmark of kidney development. One of the fundamental principles in tubulogenesis is that epithelia coordinate the polarity of individual cells with the surrounding cells and matrix. A central feature in this process is the segregation of membranes into spatially and functionally distinct apical and basolateral domains, and the generation of a luminal space at the apical surface. This review examines our current understanding of the cellular and molecular mechanisms that underlie the establishment of apical-basal polarity and lumen formation in developing renal epithelia, including the roles of cell-cell and cell-matrix interactions and polarity complexes. We highlight growing evidence from animal models, and correlate these findings with models of tubulogenesis from other organ systems, and from in vitro studies.
Collapse
Affiliation(s)
- Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. H5.102, Dallas, TX 75390-8856
| |
Collapse
|
77
|
Spencer MA, Jabeen Z, Lubensky DK. Vertex stability and topological transitions in vertex models of foams and epithelia. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2017; 40:2. [PMID: 28083791 DOI: 10.1140/epje/i2017-11489-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/30/2016] [Indexed: 06/06/2023]
Abstract
In computer simulations of dry foams and of epithelial tissues, vertex models are often used to describe the shape and motion of individual cells. Although these models have been widely adopted, relatively little is known about their basic theoretical properties. For example, while fourfold vertices in real foams are always unstable, it remains unclear whether a simplified vertex model description has the same behavior. Here, we study vertex stability and the dynamics of T1 topological transitions in vertex models. We show that, when all edges have the same tension, stationary fourfold vertices in these models do indeed always break up. In contrast, when tensions are allowed to depend on edge orientation, fourfold vertices can become stable, as is observed in some biological systems. More generally, our formulation of vertex stability leads to an improved treatment of T1 transitions in simulations and paves the way for studies of more biologically realistic models that couple topological transitions to the dynamics of regulatory proteins.
Collapse
Affiliation(s)
- Meryl A Spencer
- Department of Physics, University of Michigan, 48103, Ann Arbor, MI, USA.
| | - Zahera Jabeen
- Department of Physics, University of Michigan, 48103, Ann Arbor, MI, USA
| | - David K Lubensky
- Department of Physics, University of Michigan, 48103, Ann Arbor, MI, USA
| |
Collapse
|
78
|
Modeling psychiatric disorders: from genomic findings to cellular phenotypes. Mol Psychiatry 2016; 21:1167-79. [PMID: 27240529 PMCID: PMC4995546 DOI: 10.1038/mp.2016.89] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/15/2022]
Abstract
Major programs in psychiatric genetics have identified >150 risk loci for psychiatric disorders. These loci converge on a small number of functional pathways, which span conventional diagnostic criteria, suggesting a partly common biology underlying schizophrenia, autism and other psychiatric disorders. Nevertheless, the cellular phenotypes that capture the fundamental features of psychiatric disorders have not yet been determined. Recent advances in genetics and stem cell biology offer new prospects for cell-based modeling of psychiatric disorders. The advent of cell reprogramming and induced pluripotent stem cells (iPSC) provides an opportunity to translate genetic findings into patient-specific in vitro models. iPSC technology is less than a decade old but holds great promise for bridging the gaps between patients, genetics and biology. Despite many obvious advantages, iPSC studies still present multiple challenges. In this expert review, we critically review the challenges for modeling of psychiatric disorders, potential solutions and how iPSC technology can be used to develop an analytical framework for the evaluation and therapeutic manipulation of fundamental disease processes.
Collapse
|
79
|
Migration of Founder Epithelial Cells Drives Proper Molar Tooth Positioning and Morphogenesis. Dev Cell 2016; 35:713-24. [PMID: 26702830 DOI: 10.1016/j.devcel.2015.11.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/19/2015] [Accepted: 11/24/2015] [Indexed: 02/05/2023]
Abstract
The proper positioning of organs during development is essential, yet little is known about the regulation of this process in mammals. Using murine tooth development as a model, we have found that cell migration plays a central role in positioning of the organ primordium. By combining lineage tracing, genetic cell ablation, and confocal live imaging, we identified a migratory population of Fgf8-expressing epithelial cells in the embryonic mandible. These Fgf8-expressing progenitors furnish the epithelial cells required for tooth development, and the progenitor population migrates toward a Shh-expressing region in the mandible, where the tooth placode will initiate. Inhibition of Fgf and Shh signaling disrupted the oriented migration of cells, leading to a failure of tooth development. These results demonstrate the importance of intraepithelial cell migration in proper positioning of an initiating organ.
Collapse
|
80
|
Venero Galanternik M, Navajas Acedo J, Romero-Carvajal A, Piotrowski T. Imaging collective cell migration and hair cell regeneration in the sensory lateral line. Methods Cell Biol 2016; 134:211-56. [PMID: 27312495 DOI: 10.1016/bs.mcb.2016.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The accessibility of the lateral line system and its amenability to long-term in vivo imaging transformed the developing lateral line into a powerful model system to study fundamental morphogenetic events, such as guided migration, proliferation, cell shape changes, organ formation, organ deposition, cell specification and differentiation. In addition, the lateral line is not only amenable to live imaging during migration stages but also during postembryonic events such as sensory organ tissue homeostasis and regeneration. The robust regenerative capabilities of the mature, mechanosensory lateral line hair cells, which are homologous to inner ear hair cells and the ease with which they can be imaged, have brought zebrafish into the spotlight as a model to develop tools to treat human deafness. In this chapter, we describe protocols for long-term in vivo confocal imaging of the developing and regenerating lateral line.
Collapse
Affiliation(s)
- M Venero Galanternik
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| | - J Navajas Acedo
- Stowers Institute for Medical Research, Kansas City, MO, United States
| | - A Romero-Carvajal
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| | - T Piotrowski
- Stowers Institute for Medical Research, Kansas City, MO, United States; University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
81
|
Wernike D, Chen Y, Mastronardi K, Makil N, Piekny A. Mechanical forces drive neuroblast morphogenesis and are required for epidermal closure. Dev Biol 2016; 412:261-77. [PMID: 26923492 DOI: 10.1016/j.ydbio.2016.02.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/24/2016] [Accepted: 02/24/2016] [Indexed: 10/22/2022]
Abstract
Tissue morphogenesis requires myosin-dependent events such as cell shape changes and migration to be coordinated between cells within a tissue, and/or with cells from other tissues. However, few studies have investigated the simultaneous morphogenesis of multiple tissues in vivo. We found that during Caenorhabditis elegans ventral enclosure, when epidermal cells collectively migrate to cover the ventral surface of the embryo, the underlying neuroblasts (neuronal precursor cells) also undergo morphogenesis. We found that myosin accumulates as foci along the junction-free edges of the ventral epidermal cells to form a ring, whose closure is myosin-dependent. We also observed the accumulation of myosin foci and the adhesion junction proteins E-cadherin and α-catenin in the underlying neuroblasts. Myosin may help to reorganize a subset of neuroblasts into a rosette-like pattern, and decrease their surface area as the overlying epidermal cells constrict. Since myosin is required in the neuroblasts for ventral enclosure, we propose that mechanical forces in the neuroblasts influence constriction of the overlying epidermal cells. In support of this model, disrupting neuroblast cell division or altering their fate influences myosin localization in the overlying epidermal cells. The coordination of myosin-dependent events and forces between cells in different tissues could be a common theme for coordinating morphogenetic events during metazoan development.
Collapse
Affiliation(s)
- Denise Wernike
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Yun Chen
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | | | - Neetha Makil
- Department of Biology, Concordia University, Montreal, Quebec, Canada
| | - Alisa Piekny
- Department of Biology, Concordia University, Montreal, Quebec, Canada.
| |
Collapse
|
82
|
Muccioli M, Qaisi D, Herman K, Plageman TF. Lens placode planar cell polarity is dependent on Cdc42-mediated junctional contraction inhibition. Dev Biol 2016; 412:32-43. [PMID: 26902112 DOI: 10.1016/j.ydbio.2016.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
Development of the ocular lens commences with the formation of the lens placode, an epithelial structure that thickens and subsequently bends inward in a process called invagination. Invagination is observed during the development of many embryonic structures, but the spectrum of morphogenetic events driving this process are, in most cases, not fully understood. A characteristic commonly found in embryonic tissues undergoing epithelial reorganization is planar polarity, a property where cells are geometrically and/or molecularly orientated in a specific direction along the plane of an epithelium. Planar polarity is known to drive the morphogenesis of several epithelial structures, however its role during invagination events is less clear. We have found that at the onset of invagination, cells of the lens placode become geometrically planar polarized such that they are orientated toward a central point in the lens placode. Further investigation revealed that this is due to contraction of radially orientated junctions and the elongation of those circumferentially orientated. Radial junctions have an elevated localization of actomyosin and their contraction is dependent on the F-actin and Rho-kinase binding protein, Shroom3. Elongation of circumferential junctions is dependent upon Cdc42, a Rho-GTPase known to regulate polarity via the Par-complex. We determined that Cdc42 and members of the Par-complex inhibit Shroom3-induced contractility and promote anisotropic placode cell geometry through inhibition of junctional contraction. We postulate that invagination of the lens placode requires careful orchestration of these opposing processes which are mediated by the planar polarization of junctional proteins.
Collapse
Affiliation(s)
- Maria Muccioli
- College of Optometry, The Ohio State University, Columbus, OH 43210, United States
| | - Dalya Qaisi
- College of Optometry, The Ohio State University, Columbus, OH 43210, United States
| | - Ken Herman
- College of Optometry, The Ohio State University, Columbus, OH 43210, United States
| | - Timothy F Plageman
- College of Optometry, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
83
|
Persico AM, Arango C, Buitelaar JK, Correll CU, Glennon JC, Hoekstra PJ, Moreno C, Vitiello B, Vorstman J, Zuddas A. Unmet needs in paediatric psychopharmacology: Present scenario and future perspectives. Eur Neuropsychopharmacol 2015; 25:1513-31. [PMID: 26166453 DOI: 10.1016/j.euroneuro.2015.06.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 05/17/2015] [Accepted: 06/12/2015] [Indexed: 12/20/2022]
Abstract
Paediatric psychopharmacology holds great promise in two equally important areas of enormous biomedical and social impact, namely the treatment of behavioural abnormalities in children and adolescents, and the prevention of psychiatric disorders with adolescent- or adult-onset. Yet, in striking contrast, pharmacological treatment options presently available in child and adolescent psychiatry are dramatically limited. The most important currently unmet needs in paediatric psychopharmacology are: the frequent off-label prescription of medications to children and adolescents based exclusively on data from randomized controlled studies involving adult patients; the frequent lack of age-specific dose, long-term efficacy and tolerability/safety data; the lack of effective medications for many paediatric psychiatric disorders, most critically autism spectrum disorder; the scarcity and limitations of randomized placebo-controlled trials in paediatric psychopharmacology; the unexplored potential for the prevention of psychiatric disorders with adolescent- and adult-onset; the current lack of biomarkers to predict treatment response and severe adverse effects; the need for better preclinical data to foster the successful development of novel drug therapies; and the effective dissemination of evidence-based treatments to the general public, to better inform patients and families of the benefits and risks of pharmacological interventions during development. Priorities and strategies are proposed to overcome some of these limitations, including the European Child and Adolescent Clinical Psychopharmacology Network, as an overarching Pan-European infrastructure aimed at reliably carrying out much needed psychopharmacological trials in children and adolescents, in order to fill the identified gaps and improve overall outcomes.
Collapse
Affiliation(s)
- Antonio M Persico
- Child & Adolescent NeuroPsychiatry Unit, University Campus Bio-Medico, Rome, Italy; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy.
| | - Celso Arango
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | - Jan K Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, and Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | - Christoph U Correll
- Psychiatry Research, The Zucker Hillside Hospital, North Shore-Long Island Jewish Health System, Glen Oaks, NY, USA
| | - Jeffrey C Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, and Karakter Child and Adolescent Psychiatry University Centre, Nijmegen, The Netherlands
| | - Pieter J Hoekstra
- Department of Psychiatry, University of Groningen, University Medical Center, Groningen, The Netherlands
| | - Carmen Moreno
- Child and Adolescent Psychiatry Department, Hospital General Universitario Gregorio Marañón, School of Medicine Universidad Complutense, IiSGM, CIBERSAM, Madrid, Spain
| | | | - Jacob Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alessandro Zuddas
- Dept. Biomedical Sciences, Child & Adolescent NeuroPsychiatry Unit, University of Cagliari, Cagliari, Italy
| | | |
Collapse
|
84
|
Abstract
Insulin-producing β cells within the vertebrate fetal pancreas acquire their fate in a step-wise manner. Whereas the intrinsic factors dictating the transcriptional or epigenetic status of pancreatic lineages have been intensely examined, less is known about cell-cell interactions that might constitute a niche for the developing β cell lineage. It is becoming increasingly clear that understanding and recapitulating these steps may instruct in vitro differentiation of embryonic stem cells and/or therapeutic regeneration. Indeed, directed differentiation techniques have improved since transitioning from 2D to 3D cultures, suggesting that the 3D microenvironment in which β cells are born is critical. However, to date, it remains unknown whether the changing architecture of the pancreatic epithelium impacts the fate of cells therein. An emerging challenge in the field is to elucidate how progenitors are allocated during key events, such as the stratification and subsequent resolution of the pre-pancreatic epithelium, as well as the formation of lumens and branches. Here, we assess the progenitor epithelium and examine how it might influence the emergence of pancreatic multipotent progenitors (MPCs), which give rise to β cells and other pancreatic lineages.
Collapse
Affiliation(s)
- Leilani Marty-Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas (LMS,OC)
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas (LMS,OC)
| |
Collapse
|
85
|
Li J, Zeng G, Qi Y, Tang X, Zhang J, Wu Z, Liang J, Shi L, Liu H, Zhang P. Xenotransplantation of human adipose-derived stem cells in zebrafish embryos. PLoS One 2015; 10:e0123264. [PMID: 25849455 PMCID: PMC4388839 DOI: 10.1371/journal.pone.0123264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Zebrafish is a widely used animal model with well-characterized background in developmental biology. The fate of human adipose-derived stem cells (ADSCs) after their xenotransplantation into the developing embryos of zebrafish is unknown. Therefore, human ADSCs were firstly isolated, and then transduced with lentiviral vector system carrying a green fluorescent protein (GFP) reporter gene, and followed by detection of their cell viability and the expression of cell surface antigens. These GFP-expressing human ADSCs were transplanted into the zebrafish embryos at 3.3–4.3 hour post-fertilization (hpf). Green fluorescent signal, the proliferation and differentiation of human ADSCs in recipient embryos were respectively examined using fluorescent microscopy and immunohistochemical staining. The results indicated that human ADSCs did not change their cell viability and the expression levels of cell surface antigens after GFP transduction. Microscopic examination demonstrated that green fluorescent signals of GFP expressed in the transplanted cells were observed in the embryos and larva fish at post-transplantation. The positive staining of Ki-67 revealed the survival and proliferation of human ADSCs in fish larvae after transplantation. The expression of CD105 was observable in the xenotransplanted ADSCs, but CD31 expression was undetectable. Therefore, our results indicate that human ADSCs xenotransplanted in the zebrafish embryos not only can survive and proliferate at across-species circumstance, but also seem to maintain their undifferentiation status in a short term. This xenograft model of zebrafish embryos may provide a promising and useful technical platform for the investigation of biology and physiology of stem cells in vivo.
Collapse
Affiliation(s)
- Jin Li
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Guofang Zeng
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Yawei Qi
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Xudong Tang
- Institute of Biochemistry and Molecular Biology, Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jingjing Zhang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Zeyong Wu
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Jie Liang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
| | - Lei Shi
- School of Light Industry and Food Science, South China University of Technology, Guangzhou, Guangdong Province, China
| | - Hongwei Liu
- Department of Plastic Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory for Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
- * E-mail: (HL); (PZ)
| | - Peihua Zhang
- Institute of Plastic Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong Province, China
- * E-mail: (HL); (PZ)
| |
Collapse
|
86
|
Breau MA, Schneider-Maunoury S. Cranial placodes: models for exploring the multi-facets of cell adhesion in epithelial rearrangement, collective migration and neuronal movements. Dev Biol 2014; 401:25-36. [PMID: 25541234 DOI: 10.1016/j.ydbio.2014.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/08/2014] [Accepted: 12/09/2014] [Indexed: 01/16/2023]
Abstract
Key to morphogenesis is the orchestration of cell movements in the embryo, which requires fine-tuned adhesive interactions between cells and their close environment. The neural crest paradigm has provided important insights into how adhesion dynamics control epithelium-to-mesenchyme transition and mesenchymal cell migration. Much less is known about cranial placodes, patches of ectodermal cells that generate essential parts of vertebrate sensory organs and ganglia. In this review, we summarise the known functions of adhesion molecules in cranial placode morphogenesis, and discuss potential novel implications of adhesive interactions in this crucial developmental process. The great repertoire of placodal cell behaviours offers new avenues for exploring the multiple roles of adhesion complexes in epithelial remodelling, collective migration and neuronal movements.
Collapse
Affiliation(s)
- Marie Anne Breau
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France.
| | - Sylvie Schneider-Maunoury
- Sorbonne Universités, UPMC Univ Paris 06, IBPS-UMR7622, F-75005 Paris, France; CNRS, UMR7622, Institut de Biologie Paris-Seine (IBPS) - Laboratoire de Biologie du Développement, F-75005 Paris, France; INSERM, U1156, F-75005 Paris, France
| |
Collapse
|
87
|
Luminal signaling: it's what's on the inside that counts. Dev Cell 2014; 31:519-20. [PMID: 25490261 DOI: 10.1016/j.devcel.2014.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
During organogenesis, FGFs are diffusible communication signals that allow cells to coordinate morphogenesis and establish tissue architecture. Recently in Nature, Durdu et al. (2014) show that epithelial cell clusters secrete FGFs into a microlumen, restricting FGF localization so that participating cells coordinate differentiation and collective migration via luminal signaling.
Collapse
|