1
|
Pfann M, Ben-Tal Cohen E, Sela-Donenfeld D, Cinnamon Y. Application of the Magnet-Cre optogenetic system in the chicken model. Dev Biol 2025; 523:68-81. [PMID: 40187475 DOI: 10.1016/j.ydbio.2025.04.003] [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: 10/20/2024] [Revised: 03/03/2025] [Accepted: 04/03/2025] [Indexed: 04/07/2025]
Abstract
Chickens serve as an excellent model organism for developmental biology, offering unique opportunities for precise spatiotemporal access to embryos within eggs. Optogenes are light-activated proteins that regulate gene expression, offering a non-invasive method to activate genes at specific locations and developmental stages, advancing developmental biology research. This study employed the Magnet-Cre optogenetic system to control gene expression in developing chicken embryos. Magnet-Cre consists of two light-sensitive protein domains that dimerize upon light activation, each attached to an inactive half of the Cre recombinase enzyme, which becomes active upon dimerization. We developed an all-in-one plasmid containing a green fluorescent protein marker, the Magnet-Cre system, and a light-activated red fluorescent protein gene. This plasmid was electroporated into the neural tube of Hamburger and Hamilton (H&H) stage 14 chicken embryos. Embryo samples were cleared using the CUBIC protocol and imaged with a light sheet microscope to analyze optogenetic activity via red-fluorescent cells. We established a pipeline for Magnet-Cre activation in chicken embryos, demonstrating that a single 3-min exposure to blue light following incubation at 28 °C was sufficient to trigger gene activity within the neural tube, with increased activity upon additional light exposure. Finally, we showed a spatiotemporal control of gene activity using a localized laser light induction. This research lays the groundwork for further advancements in avian developmental biology and poultry research, enabling spatiotemporal control of genes in both embryos and transgenic chickens.
Collapse
Affiliation(s)
- Michael Pfann
- Department of Poultry and Aquaculture Science, Institute of Animal Sciences, Agricultural Research Organization - Volcani Institute, Rishon LeZion, 7505101, Israel; Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Enbal Ben-Tal Cohen
- Department of Poultry and Aquaculture Science, Institute of Animal Sciences, Agricultural Research Organization - Volcani Institute, Rishon LeZion, 7505101, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Yuval Cinnamon
- Department of Poultry and Aquaculture Science, Institute of Animal Sciences, Agricultural Research Organization - Volcani Institute, Rishon LeZion, 7505101, Israel.
| |
Collapse
|
2
|
Liu G, Peng Y, Li Z, Zhu X, Sun H, Chen G, Zuo Q, Niu Y, Song J, Han W, Wei W, Li B, Jin K. An Inducible CYP19A1 Excision Model for Sexual Differentiation in Chicken ( Gallus gallus) via the CRISPR/Cas9 System. Vet Sci 2025; 12:296. [PMID: 40284798 PMCID: PMC12031565 DOI: 10.3390/vetsci12040296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Aromatase, a crucial enzyme for estrogen synthesis, plays a vital role in gender determination and differentiation. This study aimed to establish an inducible knockout model of the chicken CYP19A1 gene, which encodes aromatase, to support gender control in chickens. We selected the most efficient sgRNA target site and constructed an inducible knockout model based on the Tet-on system. The knockout efficiency reached 80% with 20 μg/mL DOX induction in vitro. The encapsulation of the plasmid with PEI and injection into eggs achieved a knockout efficiency of 45% in ovo. qRT-PCR analysis revealed a significant downregulation of female-related genes (CYP19A1, FOXL2, ESR1) and upregulation of male-related genes (DMRT1, SOX9, AMH) in female chicken embryos after induction. Western blotting showed decreased protein expression of CYP19A1 and FOXL2, and increased SOX9 expression in female embryos post-DOX induction. Elisa detection further confirmed lower estradiol levels in the gonads of induced female embryos compared to normal and non-induced females. These findings demonstrate the successful establishment of an inducible knockout system for the CYP19A1 gene in chickens, providing theoretical and technical support for the creation of new breeding materials for gender control.
Collapse
Affiliation(s)
- Guangzheng Liu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yixiu Peng
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zeyu Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xilin Zhu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hongyan Sun
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Guohong Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD 20742, USA;
| | - Wei Han
- Jiangsu Institute of Poultry Sciences/Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou 225003, China;
| | - Wanhong Wei
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China;
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Kai Jin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (G.L.); (Y.P.); (Z.L.); (X.Z.); (H.S.); (G.C.); (Q.Z.); (Y.N.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Jiangsu Institute of Poultry Sciences/Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou 225003, China;
| |
Collapse
|
3
|
Renaux E, Baudouin C, Marchese D, Clovis Y, Lee SK, Gofflot F, Rezsohazy R, Clotman F. Lhx4 surpasses its paralog Lhx3 in promoting the differentiation of spinal V2a interneurons. Cell Mol Life Sci 2024; 81:286. [PMID: 38970652 PMCID: PMC11335214 DOI: 10.1007/s00018-024-05316-x] [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: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/08/2024]
Abstract
Paralog factors are considered to ensure the robustness of biological processes by providing redundant activity in cells where they are co-expressed. However, the specific contribution of each factor is frequently underestimated. In the developing spinal cord, multiple families of transcription factors successively contribute to differentiate an initially homogenous population of neural progenitors into a myriad of neuronal subsets with distinct molecular, morphological, and functional characteristics. The LIM-homeodomain transcription factors Lhx3, Lhx4, Isl1 and Isl2 promote the segregation and differentiation of spinal motor neurons and V2 interneurons. Based on their high sequence identity and their similar distribution, the Lhx3 and Lhx4 paralogs are considered to contribute similarly to these processes. However, the specific contribution of Lhx4 has never been studied. Here, we provide evidence that Lhx3 and Lhx4 are present in the same cell populations during spinal cord development. Similarly to Lhx3, Lhx4 can form multiproteic complexes with Isl1 or Isl2 and the nuclear LIM interactor NLI. Lhx4 can stimulate a V2-specific enhancer more efficiently than Lhx3 and surpasses Lhx3 in promoting the differentiation of V2a interneurons in chicken embryo electroporation experiments. Finally, Lhx4 inactivation in mice results in alterations of differentiation of the V2a subpopulation, but not of motor neuron production, suggesting that Lhx4 plays unique roles in V2a differentiation that are not compensated by the presence of Lhx3. Thus, Lhx4 could be the major LIM-HD factor involved in V2a interneuron differentiation during spinal cord development and should be considered for in vitro differentiation of spinal neuronal populations.
Collapse
Affiliation(s)
- Estelle Renaux
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, 1348, Belgium
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, Brussels, 1200, Belgium
| | - Charlotte Baudouin
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, Brussels, 1200, Belgium
| | - Damien Marchese
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, 1348, Belgium
| | - Yoanne Clovis
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Soo-Kyung Lee
- Pediatric Neuroscience Research Program, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Biological Sciences, University at Buffalo, Buffalo, NY, 14260, USA
| | - Françoise Gofflot
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, 1348, Belgium
| | - René Rezsohazy
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, 1348, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology, Animal Molecular and Cellular Biology, Louvain-la-Neuve, 1348, Belgium.
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, Brussels, 1200, Belgium.
| |
Collapse
|
4
|
Ventriglia S, Kalcheim C. From neural tube to spinal cord: The dynamic journey of the dorsal neuroepithelium. Dev Biol 2024; 511:26-38. [PMID: 38580174 DOI: 10.1016/j.ydbio.2024.04.001] [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: 02/07/2024] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024]
Abstract
In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.
Collapse
Affiliation(s)
- Susanna Ventriglia
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102, P.O.Box 12272, Israel.
| |
Collapse
|
5
|
Kahane N, Dahan-Barda Y, Kalcheim C. A Spatio-Temporal-Dependent Requirement of Sonic Hedgehog in the Early Development of Sclerotome-Derived Vertebrae and Ribs. Int J Mol Sci 2024; 25:5602. [PMID: 38891790 PMCID: PMC11171667 DOI: 10.3390/ijms25115602] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/19/2024] [Indexed: 06/21/2024] Open
Abstract
Derived from axial structures, Sonic Hedgehog (Shh) is secreted into the paraxial mesoderm, where it plays crucial roles in sclerotome induction and myotome differentiation. Through conditional loss-of-function in quail embryos, we investigate the timing and impact of Shh activity during early formation of sclerotome-derived vertebrae and ribs, and of lateral mesoderm-derived sternum. To this end, Hedgehog interacting protein (Hhip) was electroporated at various times between days 2 and 5. While the vertebral body and rib primordium showed consistent size reduction, rib expansion into the somatopleura remained unaffected, and the sternal bud developed normally. Additionally, we compared these effects with those of locally inhibiting BMP activity. Transfection of Noggin in the lateral mesoderm hindered sternal bud formation. Unlike Hhip, BMP inhibition via Noggin or Smad6 induced myogenic differentiation of the lateral dermomyotome lip, while impeding the growth of the myotome/rib complex into the somatic mesoderm, thus affirming the role of the lateral dermomyotome epithelium in rib guidance. Overall, these findings underscore the continuous requirement for opposing gradients of Shh and BMP activity in the morphogenesis of proximal and distal flank skeletal structures, respectively. Future research should address the implications of these early interactions to the later morphogenesis and function of the musculo-skeletal system and of possible associated malformations.
Collapse
Affiliation(s)
| | | | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, Jerusalem 9112102, Israel; (N.K.); (Y.D.-B.)
| |
Collapse
|
6
|
Sanketi BD, Sivakumar A, Kurpios NA. Visualizing and manipulating the production and accumulation of hyaluronan for functional assessment in chicken embryos. STAR Protoc 2023; 4:102200. [PMID: 36989110 PMCID: PMC10074245 DOI: 10.1016/j.xpro.2023.102200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/08/2023] [Accepted: 03/04/2023] [Indexed: 03/29/2023] Open
Abstract
Hyaluronan (HA) accumulates in the extracellular matrix to regulate organ morphogenesis. The spatiotemporal dynamics of its production and function are poorly understood due to its instability. Here, we present a protocol using the embryonic chicken intestine as a binary in vivo system for HA visualization and manipulation. We describe steps for pharmacological manipulation and in ovo electroporation to target HA production and accumulation. We then detail HA-binding protein assay to detect HA accumulation and quantification of tissue morphology changes. For complete details on the use and execution of this protocol, please refer to Sivakumar et al. (2018)1 and Sanketi et al. (2022).2.
Collapse
Affiliation(s)
- Bhargav D Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| | - Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
7
|
Saito D, Tadokoro R, Nagasaka A, Yoshino D, Teramoto T, Mizumoto K, Funamoto K, Kidokoro H, Miyata T, Tamura K, Takahashi Y. Stiffness of primordial germ cells is required for their extravasation in avian embryos. iScience 2022; 25:105629. [PMID: 36465120 PMCID: PMC9713369 DOI: 10.1016/j.isci.2022.105629] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Unlike mammals, primordial germ cells (PGCs) in avian early embryos exploit blood circulation to translocate to the somatic gonadal primordium, but how circulating PGCs undergo extravasation remains elusive. We demonstrate with single-cell level live-imaging analyses that the PGCs are arrested at a specific site in the capillary plexus, which is predominantly governed by occlusion at a narrow path in the vasculature. The occlusion is enabled by a heightened stiffness of the PGCs mediated by actin polymerization. Following the occlusion, PGCs reset their stiffness to soften in order to squeeze through the endothelial lining as they transmigrate. Our discovery also provides a model for the understanding of metastasizing cancer extravasation occurring mainly by occlusion.
Collapse
Affiliation(s)
- Daisuke Saito
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Ryosuke Tadokoro
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
- Department of Bioscience, Okayama University of Science, Okayama, Okayama 700-0005, Japan
| | - Arata Nagasaka
- Division of Histology, Meikai University School of Dentistry, Sakado, Saitama 350-0283, Japan
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Daisuke Yoshino
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Tokyo 184-8588, Japan
| | - Takayuki Teramoto
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Fukuoka 819-0395, Japan
| | - Kanta Mizumoto
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
| | - Kenichi Funamoto
- Institute of Fluid Science, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Hinako Kidokoro
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
- Organization for Research Initiatives and Development, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Koji Tamura
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Kyoto 606-8502, Japan
| |
Collapse
|
8
|
Sanketi BD, Zuela-Sopilniak N, Bundschuh E, Gopal S, Hu S, Long J, Lammerding J, Hopyan S, Kurpios NA. Pitx2 patterns an accelerator-brake mechanical feedback through latent TGFβ to rotate the gut. Science 2022; 377:eabl3921. [PMID: 36137018 PMCID: PMC10089252 DOI: 10.1126/science.abl3921] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The vertebrate intestine forms by asymmetric gut rotation and elongation, and errors cause lethal obstructions in human infants. Rotation begins with tissue deformation of the dorsal mesentery, which is dependent on left-sided expression of the Paired-like transcription factor Pitx2. The conserved morphogen Nodal induces asymmetric Pitx2 to govern embryonic laterality, but organ-level regulation of Pitx2 during gut asymmetry remains unknown. We found Nodal to be dispensable for Pitx2 expression during mesentery deformation. Intestinal rotation instead required a mechanosensitive latent transforming growth factor-β (TGFβ), tuning a second wave of Pitx2 that induced reciprocal tissue stiffness in the left mesentery as mechanical feedback with the right side. This signaling regulator, an accelerator (right) and brake (left), combines biochemical and biomechanical inputs to break gut morphological symmetry and direct intestinal rotation.
Collapse
Affiliation(s)
- Bhargav D Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Noam Zuela-Sopilniak
- Weill Institute for Cell and Molecular Biology and Department of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Elizabeth Bundschuh
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Sharada Gopal
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Joseph Long
- Weill Institute for Cell and Molecular Biology and Department of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology and Department of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Sevan Hopyan
- Program in Developmental and Stem Cell Biology, Research Institute, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
9
|
Kawachi T, Tadokoro R, Takahashi Y. Cell Lineage, Self-Renewal, and Epithelial-to-Mesenchymal Transition during Secondary Neurulation. J Korean Neurosurg Soc 2021; 64:367-373. [PMID: 33906340 PMCID: PMC8128514 DOI: 10.3340/jkns.2021.0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 11/27/2022] Open
Abstract
Secondary neurulation (SN) is a critical process to form the neural tube in the posterior region of the body including the tail. SN is distinct from the anteriorly occurring primary neurulation (PN); whereas the PN proceeds by folding an epithelial neural plate, SN precursors arise from a specified epiblast by epithelial-to-mesenchymal transition (EMT), and undergo self-renewal in the tail bud. They finally differentiate into the neural tube through mesenchymal-to-epithelial transition (MET). We here overview recent progresses in the studies of SN with a particular focus on the regulation of cell lineage, self-renewal, and EMT/MET. Cellular mechanisms underlying SN help to understand the functional diversity of the tail in vertebrates.
Collapse
Affiliation(s)
- Teruaki Kawachi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Japan
- Faculty of Engineering, Okayama University of Science, Okayama, Japan
| | - Ryosuke Tadokoro
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Japan
- Faculty of Engineering, Okayama University of Science, Okayama, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kyoto, Japan
| |
Collapse
|
10
|
Naganathan S, Oates A. Patterning and mechanics of somite boundaries in zebrafish embryos. Semin Cell Dev Biol 2020; 107:170-178. [DOI: 10.1016/j.semcdb.2020.04.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/12/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022]
|
11
|
Chu D, Nguyen A, Smith SS, Vavrušová Z, Schneider RA. Stable integration of an optimized inducible promoter system enables spatiotemporal control of gene expression throughout avian development. Biol Open 2020; 9:bio055343. [PMID: 32917762 PMCID: PMC7561481 DOI: 10.1242/bio.055343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/27/2020] [Indexed: 01/18/2023] Open
Abstract
Precisely altering gene expression is critical for understanding molecular processes of embryogenesis. Although some tools exist for transgene misexpression in developing chick embryos, we have refined and advanced them by simplifying and optimizing constructs for spatiotemporal control. To maintain expression over the entire course of embryonic development we use an enhanced piggyBac transposon system that efficiently integrates sequences into the host genome. We also incorporate a DNA targeting sequence to direct plasmid translocation into the nucleus and a D4Z4 insulator sequence to prevent epigenetic silencing. We designed these constructs to minimize their size and maximize cellular uptake, and to simplify usage by placing all of the integrating sequences on a single plasmid. Following electroporation of stage HH8.5 embryos, our tetracycline-inducible promoter construct produces robust transgene expression in the presence of doxycycline at any point during embryonic development in ovo or in culture. Moreover, expression levels can be modulated by titrating doxycycline concentrations and spatial control can be achieved using beads or gels. Thus, we have generated a novel, sensitive, tunable, and stable inducible-promoter system for high-resolution gene manipulation in vivo.
Collapse
Affiliation(s)
- Daniel Chu
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - An Nguyen
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Spenser S Smith
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Zuzana Vavrušová
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| |
Collapse
|
12
|
Debrulle S, Baudouin C, Hidalgo-Figueroa M, Pelosi B, Francius C, Rucchin V, Ronellenfitch K, Chow RL, Tissir F, Lee SK, Clotman F. Vsx1 and Chx10 paralogs sequentially secure V2 interneuron identity during spinal cord development. Cell Mol Life Sci 2020; 77:4117-4131. [PMID: 31822965 PMCID: PMC11104857 DOI: 10.1007/s00018-019-03408-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 12/01/2022]
Abstract
Paralog factors are usually described as consolidating biological systems by displaying redundant functionality in the same cells. Here, we report that paralogs can also cooperate in distinct cell populations at successive stages of differentiation. In mouse embryonic spinal cord, motor neurons and V2 interneurons differentiate from adjacent progenitor domains that share identical developmental determinants. Therefore, additional strategies secure respective cell fate. In particular, Hb9 promotes motor neuron identity while inhibiting V2 differentiation, whereas Chx10 stimulates V2a differentiation while repressing motor neuron fate. However, Chx10 is not present at the onset of V2 differentiation and in other V2 populations. In the present study, we show that Vsx1, the single paralog of Chx10, which is produced earlier than Chx10 in V2 precursors, can inhibit motor neuron differentiation and promote V2 interneuron production. However, the single absence of Vsx1 does not impact on V2 fate consolidation, suggesting that lack of Vsx1 may be compensated by other factors. Nevertheless, Vsx1 cooperates with Chx10 to prevent motor neuron differentiation in early V2 precursors although these two paralog factors are not produced in the same cells. Hence, this study uncovers an original situation, namely labor division, wherein paralog genes cooperate at successive steps of neuronal development.
Collapse
Affiliation(s)
- Stéphanie Debrulle
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Charlotte Baudouin
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Maria Hidalgo-Figueroa
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
- Neuropsychopharmacology and Psychobiology Research Group, Area of Psychobiology, Department of Psychology, Instituto de Investigación E Innovación en Ciencias Biomédicas de Cádiz (INiBICA), University of Cadiz, Cadiz, Spain
| | - Barbara Pelosi
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Cédric Francius
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
- PAREXEL International, Paris, France
| | - Vincent Rucchin
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | | | - Robert L Chow
- Department of Biology, University of Victoria, Victoria, Canada
| | - Fadel Tissir
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Soo-Kyung Lee
- Oregon Health and Science University, Papé Family Pediatric Research Institute and Vollum Institute, Portland, USA
| | - Frédéric Clotman
- Université Catholique de Louvain, Institute of Neuroscience, Brussels, Belgium.
| |
Collapse
|
13
|
Kawachi T, Shimokita E, Kudo R, Tadokoro R, Takahashi Y. Neural-fated self-renewing cells regulated by Sox2 during secondary neurulation in chicken tail bud. Dev Biol 2020; 461:160-171. [DOI: 10.1016/j.ydbio.2020.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 12/24/2022]
|
14
|
Guioli S, Zhao D, Nandi S, Clinton M, Lovell-Badge R. Oestrogen in the chick embryo can induce chromosomally male ZZ left gonad epithelial cells to form an ovarian cortex that can support oogenesis. Development 2020; 147:dev181693. [PMID: 32001442 PMCID: PMC7055392 DOI: 10.1242/dev.181693] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/16/2020] [Indexed: 12/25/2022]
Abstract
In chickens, the embryonic ovary differentiates into two distinct domains before meiosis: a steroidogenic core (the female medulla), overlain by the germ cell niche (the cortex). The differentiation of the medulla is a cell-autonomous process based on chromosomal sex identity (CASI). In order to address the extent to which cortex differentiation depends on intrinsic or extrinsic factors, we generated models of gonadal intersex by mixing ZW (female) and ZZ (male) cells in gonadal chimeras, or by altering oestrogen levels of ZW and ZZ embryos. We found that CASI does not apply to the embryonic cortex. Both ZW and ZZ cells can form the cortex and this can happen independently of the phenotypic sex of the medulla as long as oestrogen is provided. We also show that the cortex-promoting activity of oestrogen signalling is mediated via estrogen receptor alpha within the left gonad epithelium. However, the presence of a medulla with an 'intersex' or male phenotype may compromise germ cell progression into meiosis, causing cortical germ cells to remain in an immature state in the embryo.
Collapse
Affiliation(s)
| | - Debiao Zhao
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Sunil Nandi
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Michael Clinton
- The Roslin Institute and R(D)SVS, Gene Function and Development, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | | |
Collapse
|
15
|
McLennan R, Kulesa PM. In Ovo Electroporation of Plasmid DNA and Morpholinos into Specific Tissues During Early Embryogenesis. Methods Mol Biol 2019; 1976:71-82. [PMID: 30977066 DOI: 10.1007/978-1-4939-9412-0_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In ovo electroporation enables transfection of non-viral plasmid DNA and/or morpholinos to fluorescently label and/or perturb gene function in cells of interest. However, targeted electroporation into specific subregions of the embryo can be challenging due to placement and size limitations of the electrodes. Here we describe the basic techniques for in ovo electroporation in the chick embryo and suggest parameters to electroporate cells within different target tissues that with some modifications may be applicable to a wide range of developmental stages and other embryo model organisms.
Collapse
Affiliation(s)
| | - Paul M Kulesa
- Stowers Institute for Medical Research, Kansas City, MO, USA.,Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
16
|
Miller SR, Benito C, Mirsky R, Jessen KR, Baker CVH. Neural crest Notch/Rbpj signaling regulates olfactory gliogenesis and neuronal migration. Genesis 2018; 56:e23215. [PMID: 30134068 PMCID: PMC6099236 DOI: 10.1002/dvg.23215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
Abstract
The neural crest-derived ensheathing glial cells of the olfactory nerve (OECs) are unique in spanning both the peripheral and central nervous systems: they ensheathe bundles of axons projecting from olfactory receptor neurons in the nasal epithelium to their targets in the olfactory bulb. OECs are clinically relevant as a promising autologous cell transplantation therapy for promoting central nervous system repair. They are also important for fertility, being required for the migration of embryonic gonadotropin-releasing hormone (GnRH) neurons from the olfactory placode along terminal nerve axons to the medial forebrain, which they enter caudal to the olfactory bulbs. Like Schwann cell precursors, OEC precursors associated with the developing olfactory nerve express the glial marker myelin protein zero and the key peripheral glial transcription factor Sox10. The transition from Schwann cell precursors to immature Schwann cells is accelerated by canonical Notch signaling via the Rbpj transcription factor. Here, we aimed to test the role of Notch/Rbpj signaling in developing OECs by blocking the pathway in both chicken and mouse. Our results suggest that Notch/Rbpj signaling prevents the cranial neural crest cells that colonize the olfactory nerve from differentiating as neurons, and at later stages contributes to the guidance of GnRH neurons.
Collapse
Affiliation(s)
- Sophie R. Miller
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeCB2 3DYUnited Kingdom
| | - Cristina Benito
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Kristján R. Jessen
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Clare V. H. Baker
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeCB2 3DYUnited Kingdom
| |
Collapse
|
17
|
Singh PNP, Shea CA, Sonker SK, Rolfe RA, Ray A, Kumar S, Gupta P, Murphy P, Bandyopadhyay A. Precise spatial restriction of BMP signaling in developing joints is perturbed upon loss of embryo movement. Development 2018; 145:dev.153460. [PMID: 29467244 DOI: 10.1242/dev.153460] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 02/09/2018] [Indexed: 12/16/2022]
Abstract
Dynamic mechanical loading of synovial joints is necessary for normal joint development, as evidenced in certain clinical conditions, congenital disorders and animal models where dynamic muscle contractions are reduced or absent. Although the importance of mechanical forces on joint development is unequivocal, little is known about the molecular mechanisms involved. Here, using chick and mouse embryos, we observed that molecular changes in expression of multiple genes analyzed in the absence of mechanical stimulation are consistent across species. Our results suggest that abnormal joint development in immobilized embryos involves inappropriate regulation of Wnt and BMP signaling during definition of the emerging joint territories, i.e. reduced β-catenin activation and concomitant upregulation of pSMAD1/5/8 signaling. Moreover, dynamic mechanical loading of the developing knee joint activates Smurf1 expression; our data suggest that Smurf1 insulates the joint region from pSMAD1/5/8 signaling and is essential for maintenance of joint progenitor cell fate.
Collapse
Affiliation(s)
- Pratik Narendra Pratap Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Claire A Shea
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Shashank Kumar Sonker
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Rebecca A Rolfe
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Ayan Ray
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Sandeep Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Pankaj Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| | - Paula Murphy
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
18
|
Okamoto Y, Nishimura N, Matsuda K, Ranawakage DC, Kamachi Y, Kondoh H, Uchikawa M. Cooperation of Sall4 and Sox8 transcription factors in the regulation of the chicken Sox3
gene during otic placode development. Dev Growth Differ 2018. [DOI: 10.1111/dgd.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yu Okamoto
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Naoko Nishimura
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Kazunari Matsuda
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| | - Deshani C. Ranawakage
- School of Environmental Science and Engineering; Kochi University of Technology; Kochi Japan
| | - Yusuke Kamachi
- School of Environmental Science and Engineering; Kochi University of Technology; Kochi Japan
| | - Hisato Kondoh
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
- Faculty of Life Sciences; Kyoto Sangyo University; Kyoto Japan
| | - Masanori Uchikawa
- Graduate School of Frontier Biosciences; Osaka University; Osaka Japan
| |
Collapse
|
19
|
Matsubara H, Saito D, Abe G, Yokoyama H, Suzuki T, Tamura K. Upstream regulation for initiation of restricted Shh expression in the chick limb bud. Dev Dyn 2017; 246:417-430. [PMID: 28205287 DOI: 10.1002/dvdy.24493] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/06/2017] [Accepted: 02/10/2017] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The organizing center, which serves as a morphogen source, has crucial functions in morphogenesis in animal development. The center is necessarily located in a certain restricted area in the morphogenetic field, and there are several ways in which an organizing center can be restricted. The organizing center for limb morphogenesis, the ZPA (zone of polarizing activity), specifically expresses the Shh gene and is restricted to the posterior region of the developing limb bud. RESULTS The pre-pattern along the limb anteroposterior axis, provided by anterior Gli3 expression and posterior Hand2 expression, seems insufficient for the initiation of Shh expression restricted to a narrow, small spot in the posterior limb field. Comparison of the spatiotemporal patterns of gene expression between Shh and some candidate genes (Fgf8, Hoxd10, Hoxd11, Tbx2, and Alx4) upstream of Shh expression suggested that a combination of these genes' expression provides the restricted initiation of Shh expression. CONCLUSIONS Taken together with results of functional assays, we propose a model in which positive and negative transcriptional regulatory networks accumulate their functions in the intersection area of their expression regions to provide a restricted spot for the ZPA, the source of morphogen, Shh. Developmental Dynamics 246:417-430, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Haruka Matsubara
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama Aoba-ku, Sendai, 980-8578, Japan
| | - Daisuke Saito
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama Aoba-ku, Sendai, 980-8578, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aobayama Aoba-ku, Sendai, 980-8578, Japan
| | - Gembu Abe
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama Aoba-ku, Sendai, 980-8578, Japan
| | - Hitoshi Yokoyama
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Hirosaki, 036-8561, Japan
| | - Takayuki Suzuki
- Division of Biological Science, Graduate School of Science, Nagoya University, Furo-Cho, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Koji Tamura
- Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama Aoba-ku, Sendai, 980-8578, Japan
| |
Collapse
|
20
|
Miller SR, Perera SN, Baker CVH. Constitutively active Notch1 converts cranial neural crest-derived frontonasal mesenchyme to perivascular cells in vivo. Biol Open 2017; 6:317-325. [PMID: 28183698 PMCID: PMC5374403 DOI: 10.1242/bio.023887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Perivascular/mural cells originate from either the mesoderm or the cranial neural crest. Regardless of their origin, Notch signalling is necessary for their formation. Furthermore, in both chicken and mouse, constitutive Notch1 activation (via expression of the Notch1 intracellular domain) is sufficient in vivo to convert trunk mesoderm-derived somite cells to perivascular cells, at the expense of skeletal muscle. In experiments originally designed to investigate the effect of premature Notch1 activation on the development of neural crest-derived olfactory ensheathing glial cells (OECs), we used in ovo electroporation to insert a tetracycline-inducible NotchΔE construct (encoding a constitutively active mutant of mouse Notch1) into the genome of chicken cranial neural crest cell precursors, and activated NotchΔE expression by doxycycline injection at embryonic day 4. NotchΔE-targeted cells formed perivascular cells within the frontonasal mesenchyme, and expressed a perivascular marker on the olfactory nerve. Hence, constitutively activating Notch1 is sufficient in vivo to drive not only somite cells, but also neural crest-derived frontonasal mesenchyme and perhaps developing OECs, to a perivascular cell fate. These results also highlight the plasticity of neural crest-derived mesenchyme and glia. Summary: Sustained Notch1 activation is sufficient to drive cranial neural crest-derived frontonasal mesenchyme to adopt a perivascular (mural) cell fate in developing chick embryos.
Collapse
Affiliation(s)
- Sophie R Miller
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Surangi N Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Clare V H Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
21
|
Cho HM, Kim PH, Chang HK, Shen YM, Bonsra K, Kang BJ, Yum SY, Kim JH, Lee SY, Choi MC, Kim HH, Jang G, Cho JY. Targeted Genome Engineering to Control VEGF Expression in Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells: Potential Implications for the Treatment of Myocardial Infarction. Stem Cells Transl Med 2017; 6:1040-1051. [PMID: 28186692 PMCID: PMC5442764 DOI: 10.1002/sctm.16-0114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/11/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022] Open
Abstract
Human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs) exhibit potency for the regeneration of infarcted hearts. Vascular endothelial growth factor (VEGF) is capable of inducing angiogenesis and can boost stem cell‐based therapeutic effects. However, high levels of VEGF can cause abnormal blood vessel growth and hemangiomas. Thus, a controllable system to induce therapeutic levels of VEGF is required for cell therapy. We generated an inducible VEGF‐secreting stem cell (VEGF/hUCB‐MSC) that controls the expression of VEGF and tested the therapeutic efficacy in rat myocardial infarction (MI) model to apply functional stem cells to MI. To introduce the inducible VEGF gene cassette into a safe harbor site of the hUCB‐MSC chromosome, the transcription activator‐like effector nucleases system was used. After confirming the integration of the cassette into the locus, VEGF secretion in physiological concentration from VEGF/hUCB‐MSCs after doxycycline (Dox) induction was proved in conditioned media. VEGF secretion was detected in mice implanted with VEGF/hUCB‐MSCs grown via a cell sheet system. Vessel formation was induced in mice transplanted with Matrigel containing VEGF/hUCB‐MSCs treated with Dox. Moreover, seeding of the VEGF/hUCB‐MSCs onto the cardiac patch significantly improved the left ventricle ejection fraction and fractional shortening in a rat MI model upon VEGF induction. Induced VEGF/hUCB‐MSC patches significantly decreased the MI size and fibrosis and increased muscle thickness, suggesting improved survival of cardiomyocytes and protection from MI damage. These results suggest that our inducible VEGF‐secreting stem cell system is an effective therapeutic approach for the treatment of MI. Stem Cells Translational Medicine2017;6:1040–1051
Collapse
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Pyung-Hwan Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Kyung Chang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yi-Ming Shen
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwaku Bonsra
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Byung-Jae Kang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - So-Yeong Lee
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Min-Cheol Choi
- Department of Veterinary Radiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
22
|
Melanosome transfer to keratinocyte in the chicken embryonic skin is mediated by vesicle release associated with Rho-regulated membrane blebbing. Sci Rep 2016; 6:38277. [PMID: 27910904 PMCID: PMC5133614 DOI: 10.1038/srep38277] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 11/07/2016] [Indexed: 02/08/2023] Open
Abstract
During skin pigmentation in amniotes, melanin synthesized in the melanocyte is transferred to keratinocytes by a particle called the melanosome. Previous studies, mostly using dissociated cultured cells, have proposed several different models that explain how the melanosome transfer is achieved. Here, using a technique that labels the plasma membrane of melanocytes within a three-dimensional system that mimics natural tissues, we have visualized the plasma membrane of melanocytes with EGFP in chicken embryonic skin. Confocal time-lapse microscopy reveals that the melanosome transfer is mediated, at least in part, by vesicles produced by plasma membrane. Unexpectedly, the vesicle release is accompanied by the membrane blebbing of melanocytes. Blebs that have encapsulated a melanosome are pinched off to become vesicles, and these melanosome-containing vesicles are finally engulfed by neighboring keratinocytes. For both the membrane blebbing and vesicle release, Rho small GTPase is essential. We further show that the membrane vesicle-mediated melanosome transfer plays a significant role in the skin pigmentation. Given that the skin pigmentation in inter-feather spaces in chickens is similar to that in inter-hair spaces of humans, our findings should have important consequences in cosmetic medicine.
Collapse
|
23
|
Tsuchiya S, Chiba M, Kishimoto KN, Nakamura H, Tsuchiya M, Hayashi H. Transfer of the bone morphogenetic protein 4 gene into rat periodontal ligament by in vivo electroporation. Arch Oral Biol 2016; 74:123-132. [PMID: 27940045 DOI: 10.1016/j.archoralbio.2016.11.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 10/07/2016] [Accepted: 11/22/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Regulation of alveolar bone metabolism is required in clinical dentistry. The aim of the present study was to establish a method for gene transfer into the periodontal ligament (PDL) by in vivo electroporation with a plasmid vector and to investigate the effects of BMP-4 transfer into the PDL. DESIGN Plasmids containing mouse BMP-4 cDNA (pCAGGS-BMP4) were transfected into cultured rat PDL cells by in vitro electroporation, and BMP-4 production and secretion were detected by immunocytochemistry and western blotting. Next, pCAGGS-BMP4 was injected into the PDL of rats, and electroporation was performed in vivo, using original paired-needle electrodes. BMP-4 expression was examined by immunohistochemical staining 3, 7, 14, 21, and 28days after electroporation. Control groups were injected with pCAGGS by electroporation, injected with pCAGGS-BMP4 without electroporation, or subjected to neither injection nor electroporation. RESULTS In vitro-transfected rat PDL cells exhibited production and secretion of the mature-form BMP-4. After in vivo electroporation of pCAGGS-BMP4, site-specific BMP-4 expression peaked on day 3, gradually decreased until day 14, and was absent by day 21. We observed no unfavorable effects such as inflammation, degeneration, or necrosis. CONCLUSIONS Gene transfer by electroporation with plasmid DNA vectors has several advantages over other methods, including the non-viral vector, non-immunogenic effects, site-specific expression, simplicity, cost-effectiveness, and limited histological side effects. Our results indicate that the method is useful for gene therapy targeting the periodontal tissue, which regulates alveolar bone remodeling.
Collapse
Affiliation(s)
- Shinobu Tsuchiya
- Division of Oral Dysfunction Science, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Miyagi, 980-8575, Japan.
| | - Mirei Chiba
- Division of Oral Physiology, Department of Oral Function and Morphology, Tohoku University Graduate School of Dentistry, Miyagi, 980-8575, Japan.
| | - Koshi N Kishimoto
- Department of Orthopaedic Surgery, Tohoku University Graduate School of Medicine, Miyagi, 980-8575, Japan.
| | - Harukazu Nakamura
- Department of Molecular Neurobiology, Tohoku University Graduate School of Life Sciences and Institute of Development, Aging and Cancer, Miyagi, 980-8575, Japan.
| | - Masahiro Tsuchiya
- Faculty of Health Science, Department of Nursing, Tohoku Fukushi University, Miyagi, 981-8522, Japan; Division of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Miyagi, 980-8575, Japan.
| | - Haruhide Hayashi
- Division of Oral Physiology, Department of Oral Function and Morphology, Tohoku University Graduate School of Dentistry, Miyagi, 980-8575, Japan.
| |
Collapse
|
24
|
Atsuta Y, Takahashi Y. Early formation of the Müllerian duct is regulated by sequential actions of BMP/Pax2 and FGF/Lim1 signaling. Development 2016; 143:3549-3559. [PMID: 27578782 DOI: 10.1242/dev.137067] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022]
Abstract
The Müllerian duct (MD) and Wolffian duct (WD) are embryonic tubular tissues giving rise to female and male reproductive tracts, respectively. In amniote embryos, both MD and WD emerge in both sexes, but subsequently degenerate in the males and females, respectively. Here, by using MD-specific gene manipulations in chicken embryos, we identify the molecular and cellular mechanisms that link early MD specification to tubular invagination. Early (pre-)specification of MD precursors in the coelomic epithelium requires BMP signaling and its downstream target Pax2 in a WD-independent process. Subsequently, the BMP/Pax2 axis induces Lim1 expression, a hallmark of MD specification, for which FGF/ERK and WD-derived signals are also required. Finally, the sequential actions of the BMP/Pax2 and FGF/Lim1 axes culminate in epithelial invagination to form a tubular structure driven by an apical constriction, where apical accumulation of phospho-myosin light chain is positively regulated by FGF/ERK signaling. Our study delineates mechanisms governing the early formation of the MD, and also serves as a model of how an epithelial cell sheet is transformed to a tubular structure, a process seen in a variety of developmental contexts.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan AMED Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan
| |
Collapse
|
25
|
Harada H, Sato T, Nakamura H. Fgf8 signaling for development of the midbrain and hindbrain. Dev Growth Differ 2016; 58:437-45. [PMID: 27273073 DOI: 10.1111/dgd.12293] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 01/31/2023]
Abstract
In this paper, we review how midbrain and hindbrain are specified. Otx2 and Gbx2 are expressed from the early phase of development, and their expression abuts at the midbrain hindbrain boundary (MHB), where Fgf8 expression is induced, and functions as an organizing molecule for the midbrain and hindbrain. Fgf8 induces En1 and Pax2 expression at the region where Otx2 is expressed to specify midbrain. Fgf8 activates Ras-ERK pathway to specify hindbrain. Downstream of ERK, Pea3 specifies isthmus (rhombomere 0, r0), and Irx2 may specify r1, where the cerebellum is formed.
Collapse
Affiliation(s)
- Hidekiyo Harada
- Genetics and Development Division, Toronto Krembil Research Institute, Toronto, Ontario, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tatsuya Sato
- Department of Developmental Neuroscience, Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan.,Frontier Research Institute for Interdisciplinary Science, Tohoku University, Sendai, 980-8578, Japan
| | - Harukazu Nakamura
- Frontier Research Institute for Interdisciplinary Science, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
26
|
Atsuta Y, Takahashi Y. FGF8 coordinates tissue elongation and cell epithelialization during early kidney tubulogenesis. Development 2015; 142:2329-37. [PMID: 26130757 PMCID: PMC4510593 DOI: 10.1242/dev.122408] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
When a tubular structure forms during early embryogenesis, tubular elongation and lumen formation (epithelialization) proceed simultaneously in a spatiotemporally coordinated manner. We here demonstrate, using the Wolffian duct (WD) of early chicken embryos, that this coordination is regulated by the expression of FGF8, which shifts posteriorly during body axis elongation. FGF8 acts as a chemoattractant on the leader cells of the elongating WD and prevents them from epithelialization, whereas static (‘rear’) cells that receive progressively less FGF8 undergo epithelialization to form a lumen. Thus, FGF8 acts as a binary switch that distinguishes tubular elongation from lumen formation. The posteriorly shifting FGF8 is also known to regulate somite segmentation, suggesting that multiple types of tissue morphogenesis are coordinately regulated by macroscopic changes in body growth. Highlighted article: Body axis elongation is regulated by posterior FGF8 signals . In chicken, nephric duct extension also requires this FGF8 signal, while low FGF8 anteriorly triggers duct lumen formation.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
27
|
Saito D, Takahashi Y. Sympatho-adrenal morphogenesis regulated by the dorsal aorta. Mech Dev 2015; 138 Pt 1:2-7. [PMID: 26235279 DOI: 10.1016/j.mod.2015.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 07/24/2015] [Accepted: 07/27/2015] [Indexed: 12/15/2022]
Abstract
The autonomic nervous system, composed of sympathetic- and para-sympathetic neurons, plays essential roles in a variety of physiological functions including homeostasis and responses to external stimuli. We here present an overview of recent findings concerning how the sympathetic nervous system is formed during the early development, paying particular attention to the morphogenesis of those tissues derived from migrating neural crest cells. Neural crest cells, originally multipotent, are progressively specified to sympathetic ganglia neurons and adrenomedullary cells during their migration through the body. Importantly, the dorsal aorta, the first-forming blood vessel, acts as a signaling center for their migration and differentiation. BMP signals emanating from the dorsal aorta are essential for establishing environmental cues that directly act on the migrating cells. The mechanisms underlying these early neuro-vascular interactions provide insights into understanding diseases caused by malfunctions and malformations of the autonomic nervous system.
Collapse
Affiliation(s)
- Daisuke Saito
- Frontier Research Institute for Interdisciplinary Science (FRIS), Tohoku University, Aoba-ku, Sendai 980-8578, Japan
| | - Yoshiko Takahashi
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, Japan; AMED Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
28
|
Omi M, Nakamura H. Engrailed and tectum development. Dev Growth Differ 2015; 57:135-45. [DOI: 10.1111/dgd.12197] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 12/11/2014] [Accepted: 12/18/2014] [Indexed: 11/27/2022]
Affiliation(s)
- Minoru Omi
- Division of Cell Biology and Neuroscience; Department of Morphological and Physiological Sciences; Faculty of Medical Sciences; University of Fukui; Fukui 910-1193 Japan
| | - Harukazu Nakamura
- Frontier Research Institute for Interdisciplinary Science (FRIS); Tohoku University; 6-3, Aramaki aza Aoba, Aoba-ku Sendai 980-8578 Japan
| |
Collapse
|
29
|
Ren Y, Li X, Liu Q, Deng Y, Shi D. An improved Tet-on system to tightly conditionally regulate reporter gene expression. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0422-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Angiogenesis in the developing spinal cord: blood vessel exclusion from neural progenitor region is mediated by VEGF and its antagonists. PLoS One 2015; 10:e0116119. [PMID: 25585380 PMCID: PMC4293145 DOI: 10.1371/journal.pone.0116119] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 12/04/2014] [Indexed: 01/10/2023] Open
Abstract
Blood vessels in the central nervous system supply a considerable amount of oxygen via intricate vascular networks. We studied how the initial vasculature of the spinal cord is formed in avian (chicken and quail) embryos. Vascular formation in the spinal cord starts by the ingression of intra-neural vascular plexus (INVP) from the peri-neural vascular plexus (PNVP) that envelops the neural tube. At the ventral region of the PNVP, the INVP grows dorsally in the neural tube, and we observed that these vessels followed the defined path at the interface between the medially positioned and undifferentiated neural progenitor zone and the laterally positioned differentiated zone. When the interface between these two zones was experimentally displaced, INVP faithfully followed a newly formed interface, suggesting that the growth path of the INVP is determined by surrounding neural cells. The progenitor zone expressed mRNA of vascular endothelial growth factor-A whereas its receptor VEGFR2 and FLT-1 (VEGFR1), a decoy for VEGF, were expressed in INVP. By manipulating the neural tube with either VEGF or the soluble form of FLT-1, we found that INVP grew in a VEGF-dependent manner, where VEGF signals appear to be fine-tuned by counteractions with anti-angiogenic activities including FLT-1 and possibly semaphorins. These results suggest that the stereotypic patterning of early INVP is achieved by interactions between these vessels and their surrounding neural cells, where VEGF and its antagonists play important roles.
Collapse
|
31
|
Sato T, Muroyama Y, Saito T. Control of Gene Expression in Neurons by the Use of In Vivo Electroporation and the Tetracycline System. ELECTROPORATION METHODS IN NEUROSCIENCE 2015. [DOI: 10.1007/978-1-4939-2459-2_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
32
|
The combination of limb-bud removal and in ovo electroporation techniques: A new powerful method to study gene function in motoneurons undergoing lesion-induced cell death. J Neurosci Methods 2015; 239:206-13. [DOI: 10.1016/j.jneumeth.2014.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/24/2014] [Accepted: 10/24/2014] [Indexed: 12/12/2022]
|
33
|
Mazur AJ, Morosan-Puopolo G, Makowiecka A, Malicka-Błaszkiewicz M, Nowak D, Brand-Saberi B. Analysis of gelsolin expression pattern in developing chicken embryo reveals high GSN expression level in tissues of neural crest origin. Brain Struct Funct 2014; 221:515-34. [PMID: 25352156 PMCID: PMC4720725 DOI: 10.1007/s00429-014-0923-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 10/16/2014] [Indexed: 12/20/2022]
Abstract
Gelsolin is one of the most intensively studied actin-binding proteins. However, in the literature comprehensive studies of GSN expression during development have not been performed yet in all model organisms. In zebrafish, gelsolin is a dorsalizing factor that modulates bone morphogenetic proteins signaling pathways, whereas knockout of the gelsolin coding gene, GSN is not lethal in murine model. To study the role of gelsolin in development of higher vertebrates, it is crucial to estimate GSN expression pattern during development. Here, we examined GSN expression in the developing chicken embryo. We applied numerous methods to track GSN expression in developing embryos at mRNA and protein level. We noted a characteristic GSN expression pattern. Although GSN transcripts were present in several cell types starting from early developmental stages, a relatively high GSN expression was observed in eye, brain vesicles, midbrain, neural tube, heart tube, and splanchnic mesoderm. In older embryos, we observed a high GSN expression in the cranial ganglia and dorsal root ganglia. A detailed analysis of 10-day-old chicken embryos revealed high amounts of gelsolin especially within the head region: in the olfactory and optic systems, meninges, nerves, muscles, presumptive pituitary gland, and pericytes, but not oligodendrocytes in the brain. Obtained results suggest that GSN is expressed at high levels in some tissues of ectodermal origin including all neural crest derivatives. Additionally, we describe that silencing of GSN expression in brain vesicles leads to altered morphology of the mesencephalon. This implies gelsolin is crucial for chicken brain development.
Collapse
Affiliation(s)
- Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland.
| | | | - Aleksandra Makowiecka
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Maria Malicka-Błaszkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, ul. Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University of Bochum, Bochum, Germany
| |
Collapse
|
34
|
Omi M, Harada H, Watanabe Y, Funahashi JI, Nakamura H. Role of En2 in the tectal laminar formation of chick embryos. Development 2014; 141:2131-8. [PMID: 24803658 DOI: 10.1242/dev.102905] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The chick optic tectum consists of 16 laminae. Here, we report contribution of En2 to laminar formation in chick optic tecta. En2 is specifically expressed in laminae g-j of stratum griseum et fibrosum superficiale (SGFS). Misexpression of En2 resulted in disappearance of En2-expressing cells from the superficial layers (laminae a-f of SGFS), where endogenous En2 is not expressed. Misexpression of En2 before postmitotic cells had left the ventricular layer indicated that En2-misexpressing cells stopped at the laminae of endogenous En2 expression and that they did not migrate into the superficial layers. Induction of En2 misexpression using a tetracycline-inducible system after the postmitotic cells had reached superficial layers also resulted in disappearance of En2-expressing cells from the superficial layers. Time-lapse analysis showed that En2-misexpressing cells migrated back from the superficial layers towards the middle layers, where En2 is strongly expressed endogenously. Our results suggest a potential role of En2 in regulating cell migration and positioning in the tectal laminar formation.
Collapse
Affiliation(s)
- Minoru Omi
- Department of Molecular Neurobiology, Graduate School of Life Sciences, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | |
Collapse
|
35
|
Interepithelial signaling with nephric duct is required for the formation of overlying coelomic epithelial cell sheet. Proc Natl Acad Sci U S A 2014; 111:6660-5. [PMID: 24753584 DOI: 10.1073/pnas.1316728111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In most organs of the body, epithelial tissues are supported by their own basement membrane and underlying stroma, the latter being regarded as a complex of amorphous cells, extracellular matrices, and soluble factors. We demonstrate here that an epithelial tube can serve as a component of stroma that supports the formation of epithelial cell sheet derived from a different origin. During development of the mesonephros in chicken embryos, the intermediate mesoderm (IMM), which contains the Wolffian duct (WD) and its associated tubules, is overlain by a sheet of epithelial cells derived from lateral plate (coelomic) mesoderm. We describe that in normal embryos, epitheliogenesis of IMM tubes and the adjacent coelomic cell sheet proceed in a coordinated manner. When the WD was surgically ablated, the overlying coelomic epithelium exhibited aberrant morphology accompanied by a punctated basement membrane. Furthermore, the WD-ablated coelomic epithelium became susceptible to latent external stress; electroporation of Rac1 resulted in epithelial-to-mesenchymal transitions (EMTs) within the coelomic epithelium. The distorted coelomic epithelium was rescued by implanting fibronectin-producing cells in place of the WD, suggesting that fibronectin provided by WD has an important role acting interepithelially. This notion was corroborated further by directly visualizing a translocation of EGFP-tagged fibronectin from fibronectin-producing to -receiving epithelia in vivo. Our findings provide a novel insight into interepithelial signaling that also might occur in adult tissues to protect against EMT and suggest a possible new target for anticancer therapeutic strategy.
Collapse
|
36
|
Kulesa PM, McKinney MC, McLennan R. Developmental imaging: the avian embryo hatches to the challenge. ACTA ACUST UNITED AC 2014; 99:121-33. [PMID: 23897596 DOI: 10.1002/bdrc.21036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 05/31/2013] [Indexed: 01/27/2023]
Abstract
The avian embryo provides a multifaceted model to study developmental mechanisms because of its accessibility to microsurgery, fluorescence cell labeling, in vivo imaging, and molecular manipulation. Early two-dimensional planar growth of the avian embryo mimics human development and provides unique access to complex cell migration patterns using light microscopy. Later developmental events continue to permit access to both light and other imaging modalities, making the avian embryo an excellent model for developmental imaging. For example, significant insights into cell and tissue behaviors within the primitive streak, craniofacial region, and cardiovascular and peripheral nervous systems have come from avian embryo studies. In this review, we provide an update to recent advances in embryo and tissue slice culture and imaging, fluorescence cell labeling, and gene profiling. We focus on how technical advances in the chick and quail provide a clearer understanding of how embryonic cell dynamics are beautifully choreographed in space and time to sculpt cells into functioning structures. We summarize how these technical advances help us to better understand basic developmental mechanisms that may lead to clinical research into human birth defects and tissue repair.
Collapse
Affiliation(s)
- Paul M Kulesa
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA.
| | | | | |
Collapse
|
37
|
McIntyre DC, Seay NW, Croce JC, McClay DR. Short-range Wnt5 signaling initiates specification of sea urchin posterior ectoderm. Development 2013; 140:4881-9. [PMID: 24227654 DOI: 10.1242/dev.095844] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The border between the posterior ectoderm and the endoderm is a location where two germ layers meet and establish an enduring relationship that also later serves, in deuterostomes, as the anatomical site of the anus. In the sea urchin, a prototypic deuterostome, the ectoderm-endoderm boundary is established before gastrulation, and ectodermal cells at the boundary are thought to provide patterning inputs to the underlying mesenchyme. Here we show that a short-range Wnt5 signal from the endoderm actively patterns the adjacent boundary ectoderm. This signal activates a unique subcircuit of the ectoderm gene regulatory network, including the transcription factors IrxA, Nk1, Pax2/5/8 and Lim1, which are ultimately restricted to subregions of the border ectoderm (BE). Surprisingly, Nodal and BMP2/4, previously shown to be activators of ectodermal specification and the secondary embryonic axis, instead restrict the expression of these genes to subregions of the BE. A detailed examination showed that endodermal Wnt5 functions as a short-range signal that activates only a narrow band of ectodermal cells, even though all ectoderm is competent to receive the signal. Thus, cells in the BE integrate positive and negative signals from both the primary and secondary embryonic axes to correctly locate and specify the border ectoderm.
Collapse
|
38
|
Rowton M, Ramos P, Anderson DM, Rhee JM, Cunliffe HE, Rawls A. Regulation of mesenchymal-to-epithelial transition by PARAXIS during somitogenesis. Dev Dyn 2013; 242:1332-44. [PMID: 24038871 DOI: 10.1002/dvdy.24033] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/15/2013] [Accepted: 08/15/2013] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Dynamic alterations in cell shape, migration, and adhesion play a central role in tissue morphogenesis during embryonic development and congenital disease. The mesenchymal-to-epithelial transition that occurs during vertebrate somitogenesis is required for proper patterning of the axial musculoskeletal system. Somitic MET is initiated in the presomitic mesoderm by PARAXIS-dependent changes in cell adhesion, cell polarity, and the composition of the extracellular matrix. However, the target genes downstream of the transcription factor PARAXIS remain poorly described. RESULTS A genome-wide comparison of gene expression in the anterior presomitic mesoderm and newly formed somites of Paraxis(-/-) embryos resulted in a set of deregulated genes enriched for factors associated with extracellular matrix and cytoskeletal organization and cell-cell and cell-ECM adhesion. The greatest change in expression was seen in fibroblast activation protein alpha (Fap), encoding a dipeptidyl peptidase capable of increasing fibronectin and collagen fiber organization in extracellular matrix. Further, downstream genes in the Wnt and Notch signaling pathways were downregulated, predicting that PARAXIS participates in positive feedback loops in both pathways. CONCLUSIONS These data demonstrate that PARAXIS initiates and stabilizes somite epithelialization by integrating signals from multiple pathways to control the reorganization of the ECM, cytoskeleton, and adhesion junctions during MET.
Collapse
Affiliation(s)
- Megan Rowton
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | | | | | | | | | | |
Collapse
|
39
|
Neural crest and Schwann cell progenitor-derived melanocytes are two spatially segregated populations similarly regulated by Foxd3. Proc Natl Acad Sci U S A 2013; 110:12709-14. [PMID: 23858437 DOI: 10.1073/pnas.1306287110] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Skin melanocytes arise from two sources: either directly from neural crest progenitors or indirectly from neural crest-derived Schwann cell precursors after colonization of peripheral nerves. The relationship between these two melanocyte populations and the factors controlling their specification remains poorly understood. Direct lineage tracing reveals that neural crest and Schwann cell progenitor-derived melanocytes are differentially restricted to the epaxial and hypaxial body domains, respectively. Furthermore, although both populations are initially part of the Foxd3 lineage, hypaxial melanocytes lose Foxd3 at late stages upon separation from the nerve, whereas we recently found that epaxial melanocytes segregate earlier from Foxd3-positive neural progenitors while still residing in the dorsal neural tube. Gain- and loss-of-function experiments in avians and mice, respectively, reveal that Foxd3 is both sufficient and necessary for regulating the balance between melanocyte and Schwann cell development. In addition, Foxd3 is also sufficient to regulate the switch between neuronal and glial fates in sensory ganglia. Together, we propose that differential fate acquisition of neural crest-derived cells depends on their progressive segregation from the Foxd3-positive lineage.
Collapse
|
40
|
Kahane N, Ribes V, Kicheva A, Briscoe J, Kalcheim C. The transition from differentiation to growth during dermomyotome-derived myogenesis depends on temporally restricted hedgehog signaling. Development 2013; 140:1740-50. [PMID: 23533174 DOI: 10.1242/dev.092726] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The development of a functional tissue requires coordination of the amplification of progenitors and their differentiation into specific cell types. The molecular basis for this coordination during myotome ontogeny is not well understood. Dermomytome progenitors that colonize the myotome first acquire myocyte identity and subsequently proliferate as Pax7-expressing progenitors before undergoing terminal differentiation. We show that the dynamics of sonic hedgehog (Shh) signaling is crucial for this transition in both avian and mouse embryos. Initially, Shh ligand emanating from notochord/floor plate reaches the dermomyotome, where it both maintains the proliferation of dermomyotome cells and promotes myogenic differentiation of progenitors that colonized the myotome. Interfering with Shh signaling at this stage produces small myotomes and accumulation of Pax7-expressing progenitors. An in vivo reporter of Shh activity combined with mouse genetics revealed the existence of both activator and repressor Shh activities operating on distinct subsets of cells during the epaxial myotomal maturation. In contrast to observations in mice, in avians Shh promotes the differentiation of both epaxial and hypaxial myotome domains. Subsequently, myogenic progenitors become refractory to Shh; this is likely to occur at the level of, or upstream of, smoothened signaling. The end of responsiveness to Shh coincides with, and is thus likely to enable, the transition into the growth phase of the myotome.
Collapse
Affiliation(s)
- Nitza Kahane
- Department of Medical Neurobiology, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
41
|
Sato Y, Lansford R. Transgenesis and imaging in birds, and available transgenic reporter lines. Dev Growth Differ 2013; 55:406-21. [PMID: 23621574 DOI: 10.1111/dgd.12058] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 03/10/2013] [Accepted: 03/14/2013] [Indexed: 12/17/2022]
Abstract
Avian embryos are important model organism to study higher vertebrate development. Easy accessibility to developing avian embryos enables a variety of experimental applications to understand specific functions of molecules, tissue-tissue interactions, and cell lineages. The whole-mount ex ovo culture technique for avian embryos permits time-lapse imaging analysis for a better understanding of cell behaviors underlying tissue morphogenesis in physiological conditions. To study mechanisms of blood vessel formation and remodeling in developing embryos by using a time-lapse imaging approach, a transgenic quail model, Tg(tie1:H2B-eYFP), was generated. From a cell behavior perspective, Tg(tie1:H2B-eYFP) quail embryos are a suitable model to shed light on how the structure and pattern of blood vessels are established in higher vertebrates. In this manuscript, we give an overview on the biological and technological background of the transgenic quail model and describe procedures for the ex ovo culture of quail embryos and time-lapse imaging analysis.
Collapse
Affiliation(s)
- Yuki Sato
- Priority Organization for Innovation and Excellence, Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811, Japan.
| | | |
Collapse
|
42
|
Atsuta Y, Tadokoro R, Saito D, Takahashi Y. Transgenesis of the Wolffian duct visualizes dynamic behavior of cells undergoing tubulogenesis in vivo. Dev Growth Differ 2013; 55:579-90. [PMID: 23550588 DOI: 10.1111/dgd.12047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 01/18/2013] [Accepted: 01/18/2013] [Indexed: 12/12/2022]
Abstract
Deciphering how the tubulogenesis is regulated is an essential but unsolved issue in developmental biology. Here, using Wolffian duct (WD) formation in chicken embryos, we have developed a novel method that enables gene manipulation during tubulogenesis in vivo. Exploiting that WD arises from a defined site located anteriorly in the embryo (pronephric region), we targeted this region with the enhanced green fluorescent protein (EGFP) gene by the in ovo electroporation technique. EGFP-positive signals were detected in a wide area of elongating WD, where transgenic cells formed an epithelial component in a mosaic manner. Time-lapse live imaging analyses further revealed dynamic behavior of cells during WD elongation: some cells possessed numerous filopodia, and others exhibited cellular tails that repeated elongation and retraction. The retraction of the tail was precisely regulated by Rho activity via actin dynamics. When electroporated with the C3 gene, encoding Rho inhibitor, WD cells failed to contract their tails, resulting in an aberrantly elongated process. We further combined with the Tol2 transposon-mediated gene transfer technique, and could trace EGFP-positive cells at later stages in the ureteric bud sprouting from WD. This is the first demonstration that exogenous gene(s) can directly be introduced into elongating tubular structures in living amniote embryos. This method has opened a way to investigate how a complex tubulogenesis proceeds in higher vertebrates.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Zoology, Graduate School of Science, Kyoto University, Kitashirakawa, Sakyo-ku, Kyoto, 606-8502, Japan
| | | | | | | |
Collapse
|
43
|
Sato T, Muroyama Y, Saito T. Inducible gene expression in postmitotic neurons by an in vivo electroporation-based tetracycline system. J Neurosci Methods 2013; 214:170-6. [PMID: 23357027 DOI: 10.1016/j.jneumeth.2013.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 01/09/2013] [Accepted: 01/16/2013] [Indexed: 01/05/2023]
Abstract
In vivo electroporation has been widely used to transfect foreign genes into neural progenitors and analyze the function of genes of interest in the developing nervous system. However, it has not been thoroughly examined in the conditional regulation of exogenous genes in postmitotic neurons. Here we show that the combination of in vivo electroporation and the newest version of the tetracycline (Tet)-controlled gene regulatory (Tet-On) system efficiently induced gene expression in various types of neurons in mouse embryonic and postnatal tissues. In pyramidal neurons of the cerebral cortex, tetracycline-responsive element (TRE)-driven gene expression was induced in the presence of doxycycline (Dox). The induction occurred in a dose-dependent manner. The Dox-dependent induction was also observed in cerebellar Purkinje cells and spinal cord neurons. Moreover, the TRE-driven inducible expression of mammalian Barh1 (Mbh1) mimicked the phenotype of the ubiquitous expression of Mbh1 in the spinal cord. These results indicate that the combination of the Tet-On system and in vivo electroporation is useful for analyzing gene function specifically in postmitotic neurons.
Collapse
Affiliation(s)
- Tatsuya Sato
- Department of Developmental Biology, Graduate School of Medicine, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba 260-8670, Japan
| | | | | |
Collapse
|
44
|
TrkB downregulation is required for dendrite retraction in developing neurons of chicken nucleus magnocellularis. J Neurosci 2013; 32:14000-9. [PMID: 23035107 DOI: 10.1523/jneurosci.2274-12.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The chick embryo (Gallus domesticus) is one of the most important model systems in vertebrate developmental biology. The development and function of its auditory brainstem circuitry is exceptionally well studied. These circuits represent an excellent system for genetic manipulation to investigate mechanisms controlling neural circuit formation, synaptogenesis, neuronal polarity, and dendritic arborization. The present study investigates the auditory nucleus, nucleus magnocellularis (NM). The neurotrophin receptor TrkB regulates dendritic structure in CNS neurons. TrkB is expressed in NM neurons at E7-E8 when these neurons have dendritic arbors. Downregulation of TrkB occurs after E8 followed by retraction of dendrites and by E18 most NM cells are adendritic. Is cessation of TrkB expression in NM necessary for dendritic retraction? To answer this question we combined focal in ovo electroporation with transposon mediated gene transfer to obtain stable expression of Doxycycline (Dox) regulated transgenes, specifically TrkB coexpressed with EGFP in a temporally controlled manner. Electroporation was performed at E2 and Dox added onto the chorioallointoic membrane from E7.5 to E16. Expression of EGFP had no effect on development of the embryo, or cell morphology and organization of auditory brainstem nuclei. NM cells expressing EGFP and TrkB at E17-E18 had dendrites and biophysical properties uncharacteristic for normal NM cells, indicating that cessation of TrkB expression is essential for dendrite retraction and functional maturation of these neurons. These studies indicate that expression of transposon based plasmids is an effective method to genetically manipulate events in mid to late embryonic brain development in chick.
Collapse
|
45
|
Abstract
In ovo electroporation is a popular technique to study gene function during development. This technique enables precise temporal and spatial genetic manipulation with the added advantages of being quick and inexpensive. In this chapter the transient transfection of a construct into the neural tube of a chicken embryo via in ovo electroporation is described. Modifications of this basic technique and methods to -analyze the resulting electroporated embryos such as qPCR and microarray are also discussed.
Collapse
Affiliation(s)
- Emma K Farley
- Division of Genetics, Genomics and Development Center for Integrative Genomics, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| |
Collapse
|
46
|
Gong P, Yang YP, Yang Y, Feng YP, Li SJ, Peng XL, Gong YZ. Different gene transfer methods at the very early, early, late and whole embryonic stages in chicken. ACTA BIOLOGICA HUNGARICA 2012; 63:453-62. [PMID: 23134602 DOI: 10.1556/abiol.63.2012.4.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
New technologies in gene transfer combined with experimental embryology make the chicken embryo an excellent model system for gene function studies. The techniques of in ovo electroporation, in vitro culture for ex ovo electroporation and retrovirus-mediated gene transfer have already been fully developed in chicken. Yet to our knowledge, there are no definite descriptions on the features and application scopes of these techniques. The survival rates of different in vitro culture methods were compared and the EGFP expression areas of different gene transfer techniques were explored. It was that the optimal timings of removing embryo for EC culture and Petri dish system was at E1.5 and E2.5, respectively; and optimal timing of injecting retrovirus is at E0. Results indicated that the EC culture, in ovo electroporation, the Petri dish system and retrovirus-mediated method are, respectively, suitable for the very early, early, late and whole embryonic stages in chicken. Comparison of different gene transfer methods and establishment of optimal timings are expected to provide a better choice of the efficient method for a particular experiment.
Collapse
Affiliation(s)
- Ping Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, 430070, Wuhan, Hubei, PR China
| | | | | | | | | | | | | |
Collapse
|
47
|
Nakamura H, Funahashi J. Electroporation: past, present and future. Dev Growth Differ 2012; 55:15-9. [PMID: 23157363 DOI: 10.1111/dgd.12012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Revised: 09/20/2012] [Accepted: 09/25/2012] [Indexed: 01/13/2023]
Abstract
Gene transfer by electroporation has become an indispensable method for the study of developmental biology. The technique is applied not only in chick embryos but also in mice and other organisms. Here, a short history and perspectives of electroporation for gene transfer in vertebrates are described.
Collapse
Affiliation(s)
- Harukazu Nakamura
- Department of Molecular Neurobiology, Graduate School of Life Sciences and Institute of Development, Aging and Cancer, Tohoku University, Seiryo-machi 4-1, Aoba-ku, Sendai, Japan.
| | | |
Collapse
|
48
|
Sato Y. Dorsal aorta formation: separate origins, lateral-to-medial migration, and remodeling. Dev Growth Differ 2012; 55:113-29. [PMID: 23294360 DOI: 10.1111/dgd.12010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 09/19/2012] [Accepted: 09/19/2012] [Indexed: 01/12/2023]
Abstract
Blood vessel formation is a highly dynamic tissue-remodeling event that can be observed from early development in vertebrate embryos. Dorsal aortae, the first functional intra-embryonic blood vessels, arise as two separate bilateral vessels in the trunk and undergo lateral-to-medial translocation, eventually fusing into a single large vessel at the midline. After this dramatic remodeling, the dorsal aorta generates hematopoietic stem cells. The dorsal aorta is a good model to use to increase our understanding of the mechanisms controlling the establishment and remodeling of larger blood vessels in vivo. Because of the easy accessibility to the developing circulatory system, quail and chick embryos have been widely used for studies on blood vessel formation. In particular, the mapping of endothelial cell origins has been performed using quail-chick chimera analysis, revealing endothelial, vascular smooth muscle, and hematopoietic cell progenitors of the dorsal aorta. The avian embryo model also allows conditional gene activation/inactivation and direct observation of cell behaviors during dorsal aorta formation. This allows a better understanding of the molecular mechanisms underlying specific morphogenetic events during dynamic dorsal aorta formation from a cell behavior perspective.
Collapse
Affiliation(s)
- Yuki Sato
- Priority Organization for Innovation and Excellence, Kumamoto University, 2-2-1 Honjo, Kumamoto, Japan.
| |
Collapse
|
49
|
Chrysostomou E, Gale JE, Daudet N. Delta-like 1 and lateral inhibition during hair cell formation in the chicken inner ear: evidence against cis-inhibition. Development 2012; 139:3764-74. [DOI: 10.1242/dev.074476] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The formation of the salt-and-pepper mosaic of hair cells and supporting cells in the sensory epithelia of the inner ear is regulated by Notch signalling and lateral inhibition, but the dynamics of this process and precise mode of action of delta-like 1 (Dll1) in this context are unclear. Here, we transfected the chicken inner ear with a fluorescent reporter that includes elements of the mammalian Hes5 promoter to monitor Notch activity in the developing sensory patches. The Hes5 reporter was active in proliferating cells and supporting cells, and Dll1 expression was highest in prospective hair cells with low levels of Notch activity, which occasionally contacted more differentiated hair cells. To investigate Dll1 functions we used constructs in which Dll1 expression was either constitutive, regulated by the Hes5 promoter, or induced by doxycycline. In support of the standard lateral inhibition model, both continuous and Hes5-regulated expression of Dll1 promoted hair cell differentiation cell-autonomously (in cis) and inhibited hair cell formation in trans. However, some hair cells formed despite contacting Dll1-overexpressing cells, suggesting that some progenitor cells are insensitive to lateral inhibition. This is not due to the cis-inhibition of Notch activity by Dll1 itself, as induction of Dll1 did not cell-autonomously reduce the activity of the Hes5 reporter in progenitor and supporting cells. Altogether, our results show that Dll1 functions primarily in trans to regulate hair cell production but also that additional mechanisms operate downstream of lateral inhibition to eliminate patterning errors in the sensory epithelia of the inner ear.
Collapse
Affiliation(s)
- Elena Chrysostomou
- University College London, The Ear Institute, 332 Gray's Inn Road, London WC1X 8EE, UK
| | - Jonathan E. Gale
- University College London, The Ear Institute, 332 Gray's Inn Road, London WC1X 8EE, UK
| | - Nicolas Daudet
- University College London, The Ear Institute, 332 Gray's Inn Road, London WC1X 8EE, UK
| |
Collapse
|
50
|
Saito D, Takase Y, Murai H, Takahashi Y. The dorsal aorta initiates a molecular cascade that instructs sympatho-adrenal specification. Science 2012; 336:1578-81. [PMID: 22723422 DOI: 10.1126/science.1222369] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The autonomic nervous system, which includes the sympathetic neurons and adrenal medulla, originates from the neural crest. Combining avian blood vessel-specific gene manipulation and mouse genetics, we addressed a long-standing question of how neural crest cells (NCCs) generate sympathetic and medullary lineages during embryogenesis. We found that the dorsal aorta acts as a morphogenetic signaling center that coordinates NCC migration and cell lineage segregation. Bone morphogenetic proteins (BMPs) produced by the dorsal aorta are critical for the production of the chemokine stromal cell-derived factor-1 (SDF -1) and Neuregulin 1 in the para-aortic region, which act as chemoattractants for early migration. Later, BMP signaling is directly involved in the sympatho-medullary segregation. This study provides insights into the complex developmental signaling cascade that instructs one of the earliest events of neurovascular interactions guiding embryonic development.
Collapse
Affiliation(s)
- Daisuke Saito
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | | | | | | |
Collapse
|