1
|
de Wallau MB, Xavier AC, Moreno CA, Kim CA, Mendes EL, Ribeiro EM, Oliveira A, Félix TM, Fett-Conte AC, Bonadia LC, Correia-Costa GR, Monlleó IL, Gil-da-Silva-Lopes VL, Vieira TP. 22q11.2 Deletion Syndrome: Influence of Parental Origin on Clinical Heterogeneity. Genes (Basel) 2024; 15:518. [PMID: 38674452 PMCID: PMC11050591 DOI: 10.3390/genes15040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
22q11.2 deletion syndrome (22q11.2DS) shows significant clinical heterogeneity. This study aimed to explore the association between clinical heterogeneity in 22q11.2DS and the parental origin of the deletion. The parental origin of the deletion was determined for 61 individuals with 22q11.2DS by genotyping DNA microsatellite markers and single-nucleotide polymorphisms (SNPs). Among the 61 individuals, 29 (47.5%) had a maternal origin of the deletion, and 32 (52.5%) a paternal origin. Comparison of the frequency of the main clinical features between individuals with deletions of maternal or paternal origin showed no statistically significant difference. However, Truncus arteriosus, pulmonary atresia, seizures, and scoliosis were only found in patients with deletions of maternal origin. Also, a slight difference in the frequency of other clinical features between groups of maternal or paternal origin was noted, including congenital heart disease, endocrinological alterations, and genitourinary abnormalities, all of them more common in patients with deletions of maternal origin. Although parental origin of the deletion does not seem to contribute to the phenotypic variability of most clinical signs observed in 22q11.2DS, these findings suggest that patients with deletions of maternal origin could have a more severe phenotype. Further studies with larger samples focusing on these specific features could corroborate these findings.
Collapse
Affiliation(s)
- Melissa Bittencourt de Wallau
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| | | | - Carolina Araújo Moreno
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| | - Chong Ae Kim
- Instituto da Criança, Hospital de Clínicas, FMUSP, São Paulo 05403-000, São Paulo, Brazil;
| | - Elaine Lustosa Mendes
- Serviço de Genética do Hospital de Clínicas da UFPR, Curitiba 80060-900, Paraná, Brazil;
| | - Erlane Marques Ribeiro
- Serviço de Genética do Hospital Infantil Albert Sabin—HIAS, Fortaleza 60410-794, Ceará, Brazil;
| | - Amanda Oliveira
- Centro de Atenção aos Defeitos da Face—CADEFI, Recife 50060-293, Pernambuco, Brazil;
| | - Têmis Maria Félix
- Serviço de Genética Médica do Hospital de Clínicas de Porto Alegre—HCPA, Porto Alegre 90035-903, Rio Grande do Sul, Brazil;
| | - Agnes Cristina Fett-Conte
- Serviço de Genética da Faculdade de Medicina de São José do Rio Preto (FAMERP/FUNFARME), São José do Rio Preto 15090-000, São Paulo, Brazil;
| | - Luciana Cardoso Bonadia
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| | - Gabriela Roldão Correia-Costa
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| | - Isabella Lopes Monlleó
- Serviço de Genética Médica do Hospital Universitário Prof. Alberto Antunes (HUPAA), Faculdade de Medicina, Universidade Federal de Alagoas (UFAL), Maceió 57072-900, Alagoas, Brazil;
| | - Vera Lúcia Gil-da-Silva-Lopes
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| | - Társis Paiva Vieira
- Medical Genetics and Genomic Medicine, Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas 13083-887, São Paulo, Brazil; (M.B.d.W.); (C.A.M.); (L.C.B.); (G.R.C.-C.); (V.L.G.-d.-S.-L.)
| |
Collapse
|
2
|
Green RM, Lo Vercio LD, Dauter A, Barretto EC, Devine J, Vidal-García M, Marchini M, Robertson S, Zhao X, Mahika A, Shakir MB, Guo S, Boughner JC, Dean W, Lander AD, Marcucio RS, Forkert ND, Hallgrímsson B. Quantifying the relationship between cell proliferation and morphology during development of the face. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.12.540515. [PMID: 37214859 PMCID: PMC10197725 DOI: 10.1101/2023.05.12.540515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Morphogenesis requires highly coordinated, complex interactions between cellular processes: proliferation, migration, and apoptosis, along with physical tissue interactions. How these cellular and tissue dynamics drive morphogenesis remains elusive. Three dimensional (3D) microscopic imaging poses great promise, and generates elegant images. However, generating even moderate through-put quantified images is challenging for many reasons. As a result, the association between morphogenesis and cellular processes in 3D developing tissues has not been fully explored. To address this critical gap, we have developed an imaging and image analysis pipeline to enable 3D quantification of cellular dynamics along with 3D morphology for the same individual embryo. Specifically, we focus on how 3D distribution of proliferation relates to morphogenesis during mouse facial development. Our method involves imaging with light-sheet microscopy, automated segmentation of cells and tissues using machine learning-based tools, and quantification of external morphology via geometric morphometrics. Applying this framework, we show that changes in proliferation are tightly correlated to changes in morphology over the course of facial morphogenesis. These analyses illustrate the potential of this pipeline to investigate mechanistic relationships between cellular dynamics and morphogenesis during embryonic development.
Collapse
Affiliation(s)
- Rebecca M Green
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lucas D Lo Vercio
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Andreas Dauter
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Elizabeth C Barretto
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Jay Devine
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Marta Vidal-García
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | | | - Samuel Robertson
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Xiang Zhao
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Anandita Mahika
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - M Bilal Shakir
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Sienna Guo
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Julia C Boughner
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Wendy Dean
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Nils D Forkert
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Benedikt Hallgrímsson
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Wang D, Wang A, Wu F, Qiu X, Li Y, Chu J, Huang WC, Xu K, Gong X, Li S. Sox10 + adult stem cells contribute to biomaterial encapsulation and microvascularization. Sci Rep 2017; 7:40295. [PMID: 28071739 PMCID: PMC5223127 DOI: 10.1038/srep40295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/05/2016] [Indexed: 01/06/2023] Open
Abstract
Implanted biomaterials and biomedical devices generally induce foreign body reaction and end up with encapsulation by a dense avascular fibrous layer enriched in extracellular matrix. Fibroblasts/myofibroblasts are thought to be the major cell type involved in encapsulation, but it is unclear whether and how stem cells contribute to this process. Here we show, for the first time, that Sox10+ adult stem cells contribute to both encapsulation and microvessel formation. Sox10+ adult stem cells were found sparsely in the stroma of subcutaneous loose connective tissues. Upon subcutaneous biomaterial implantation, Sox10+ stem cells were activated and recruited to the biomaterial scaffold, and differentiated into fibroblasts and then myofibroblasts. This differentiation process from Sox10+ stem cells to myofibroblasts could be recapitulated in vitro. On the other hand, Sox10+ stem cells could differentiate into perivascular cells to stabilize newly formed microvessels. Sox10+ stem cells and endothelial cells in three-dimensional co-culture self-assembled into microvessels, and platelet-derived growth factor had chemotactic effect on Sox10+ stem cells. Transplanted Sox10+ stem cells differentiated into smooth muscle cells to stabilize functional microvessels. These findings demonstrate the critical role of adult stem cells in tissue remodeling and unravel the complexity of stem cell fate determination.
Collapse
Affiliation(s)
- Dong Wang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,School of Optometry and Vision Science Program, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Aijun Wang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Surgery, University of California, Davis, Sacramento, California 95817, USA
| | - Fan Wu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Xuefeng Qiu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ye Li
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, California 94720, USA.,Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Julia Chu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Wen-Chin Huang
- Department of Bioengineering, University of California, Berkeley, California 94720, USA
| | - Kang Xu
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Xiaohua Gong
- School of Optometry and Vision Science Program, University of California, Berkeley, California 94720, USA
| | - Song Li
- Department of Bioengineering, University of California, Berkeley, California 94720, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
4
|
Nagy N, Goldstein AM. Enteric nervous system development: A crest cell's journey from neural tube to colon. Semin Cell Dev Biol 2017; 66:94-106. [PMID: 28087321 DOI: 10.1016/j.semcdb.2017.01.006] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
Abstract
The enteric nervous system (ENS) is comprised of a network of neurons and glial cells that are responsible for coordinating many aspects of gastrointestinal (GI) function. These cells arise from the neural crest, migrate to the gut, and then continue their journey to colonize the entire length of the GI tract. Our understanding of the molecular and cellular events that regulate these processes has advanced significantly over the past several decades, in large part facilitated by the use of rodents, avians, and zebrafish as model systems to dissect the signals and pathways involved. These studies have highlighted the highly dynamic nature of ENS development and the importance of carefully balancing migration, proliferation, and differentiation of enteric neural crest-derived cells (ENCCs). Proliferation, in particular, is critically important as it drives cell density and speed of migration, both of which are important for ensuring complete colonization of the gut. However, proliferation must be tempered by differentiation among cells that have reached their final destination and are ready to send axonal extensions, connect to effector cells, and begin to produce neurotransmitters or other signals. Abnormalities in the normal processes guiding ENCC development can lead to failure of ENS formation, as occurs in Hirschsprung disease, in which the distal intestine remains aganglionic. This review summarizes our current understanding of the factors involved in early development of the ENS and discusses areas in need of further investigation.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States; Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States; Center for Neurointestinal Health, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
5
|
Elsherif MA, Spinner RJ, Tubbs RS, Persaud TVN, Fogelson JL. The association of a hemivertebra with a large dumbbell-shaped tumor: A potential embryological explanation. Clin Anat 2016; 29:807-10. [PMID: 27006291 DOI: 10.1002/ca.22715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 11/08/2022]
Affiliation(s)
| | | | | | - T V N Persaud
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
6
|
Krisa L, Murray M. The implications of injury in the developing nervous system on upper extremity function. J Hand Ther 2016; 28:101-4; quiz 105. [PMID: 25835256 DOI: 10.1016/j.jht.2015.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/06/2015] [Accepted: 01/08/2015] [Indexed: 02/03/2023]
Abstract
STUDY DESIGN Literature review. PURPOSE The corticospinal system (CS) and peripheral nervous system (PNS) are common sites of damage during the early stages of life. The prenatal or immediately prenatal period is the most common time for damage to occur. Here we briefly review the basic features of the development of the CS and the PNS and the clinical consequences of injury to or improper development of these systems on upper extremity (UE) function. RESULTS The proper development of both the CS and PNS is necessary to achieve adequate function of the (UE). Injury or improper development of these systems can lead to upper extremity dysfunction and limit participation in activities of daily living. CONCLUSIONS Both the PNS and CS play major roles in the proper functioning of the UE. A better understanding of their roles and common developmental disorders is needed to move rehabilitation of motor impairments forward.
Collapse
Affiliation(s)
- Laura Krisa
- Jefferson University School of Health Professions, Department of Occupational Therapy, Philadelphia, PA, USA; Jefferson University School of Health Professions, Department of Physical Therapy, Philadelphia, PA, USA; Drexel University College of Medicine, Department of Neurobiology and Anatomy, Philadelphia, PA, USA.
| | - Marion Murray
- Jefferson University School of Health Professions, Department of Occupational Therapy, Philadelphia, PA, USA; Jefferson University School of Health Professions, Department of Physical Therapy, Philadelphia, PA, USA; Drexel University College of Medicine, Department of Neurobiology and Anatomy, Philadelphia, PA, USA
| |
Collapse
|
7
|
Maeda S, Ueda K, Yamana H, Tashiro-Yamaji J, Ibata M, Mikura A, Okada M, Yasuda E, Shibayama Y, Yoshino M, Kubota T, Yoshida R. Blood supply--susceptible formation of melanin pigment in hair bulb melanocytes of mice. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 3:e328. [PMID: 25878939 PMCID: PMC4387150 DOI: 10.1097/gox.0000000000000284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 01/07/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Allogeneic skin grafts onto C57BL/6 mice are rejected, and the rejected skin is replaced by surrounding skin with black hair. In contrast, syngeneic skin grafts are tolerated, and gray hair grows on the grafts. METHODS To explore the mechanism of gray hair growing on the tolerated skin grafts, we prepared full-thickness skin (2-cm square) autografts, 2 (2 cm + 2 cm) horizontal or vertical parallel incisions, and U-shaped (2 cm × 2 cm × 2 cm) flaps with or without pedicle vessels. The grafts, incisions, and flaps were fixed by suturing with string and protected by a transparent bandage. On day 14 after the operation, the bandages were removed to observe the color of the hair growing on the skin. RESULTS Skin autografts from wild-type or hepatocyte growth factor-transgenic (Tg) C57BL/6 mice survived with gray hair, whereas those from steel factor (Kitl)-Tg C57BL/6 mice survived with black hair. In addition, U-shaped flaps lacking both of the 2 main feeding vessels of wild-type mice had gray hair at the tip of the flaps. Light microscopy after staining with hematoxylin and eosin or dihydroxyphenylalanine showed that the formation of melanin pigment in the follicles, but not in the interadnexal skin, was susceptible to the blood supply. CONCLUSIONS Melanin pigment formation in the hair bulb melanocytes appeared to be susceptible to the blood supply, and melanocytosis was promoted in the follicles and in the epidermis of Kitl-Tg C57BL/6 mice.
Collapse
Affiliation(s)
- Shogo Maeda
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Koichi Ueda
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hidenori Yamana
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Junko Tashiro-Yamaji
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Minenori Ibata
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ayako Mikura
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Masashi Okada
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Emi Yasuda
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Yuro Shibayama
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Miya Yoshino
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Takahiro Kubota
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryotaro Yoshida
- From the Department of Physiology, Osaka Medical College, Takatsuki, Japan; Department of Plastic and Reconstructive Surgery, Osaka Medical College, Takatsuki, Japan; Department of Pathology, Osaka Medical College, Takatsuki, Japan; and Division of Immunology, Department of Molecular and Cellular Biology, School of Life Science, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
8
|
Tang XJ, Liu W, Yang B, Shi L, Yin L, Zhang ZY. Parry-Romberg syndrome with rare maxillofacial deformities: a report on two cases. J Craniomaxillofac Surg 2013; 42:780-3. [PMID: 24359863 DOI: 10.1016/j.jcms.2013.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/20/2013] [Accepted: 11/04/2013] [Indexed: 11/30/2022] Open
Abstract
Parry-Romberg syndrome, also known as progressive hemifacial atrophy, is a rare developmental disorder characterized by progressing unilateral facial atrophy slowly, which may affect the skin, fat, muscle and bone. It can also be associated with different systemic manifestations and deformities. In this article, we present the two cases with Parry-Romberg syndrome. Of them, one has additional and rare facial deformity with rare facial cleft and the other has a special tongue feature.
Collapse
Affiliation(s)
- Xiao-Jun Tang
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China
| | - Wei Liu
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China
| | - Bin Yang
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China
| | - Lei Shi
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China
| | - Lin Yin
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China
| | - Zhi-Yong Zhang
- Department of Maxillofacial Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Shi-Jing-Shan, Beijing 100144, China.
| |
Collapse
|
9
|
Bhat R, Bissell MJ. Of plasticity and specificity: dialectics of the microenvironment and macroenvironment and the organ phenotype. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 3:147-63. [PMID: 24719287 DOI: 10.1002/wdev.130] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 07/30/2013] [Accepted: 08/26/2013] [Indexed: 01/09/2023]
Abstract
The study of biological form and how it arises is the domain of the developmental biologists; but once the form is achieved, the organ poses a fascinating conundrum for all the life scientists: how are form and function maintained in adult organs throughout most of the life of the organism? That they do appears to contradict the inherently plastic nature of organogenesis during development. How do cells with the same genetic information arrive at, and maintain such different architectures and functions, and how do they keep remembering that they are different from each other? It is now clear that narratives based solely on genes and an irreversible regulatory dynamics cannot answer these questions satisfactorily, and the concept of microenvironmental signaling needs to be added to the equation. During development, cells rearrange and differentiate in response to diffusive morphogens, juxtacrine signals, and the extracellular matrix (ECM). These components, which constitute the modular microenvironment, are sensitive to cues from other tissues and organs of the developing embryo as well as from the external macroenvironment. On the other hand, once the organ is formed, these modular constituents integrate and constrain the organ architecture, which ensures structural and functional homeostasis and therefore, organ specificity. We argue here that a corollary of the above is that once the organ architecture is compromised in adults by mutations or by changes in the microenvironment such as aging or inflammation, that organ becomes subjected to the developmental and embryonic circuits in search of a new identity. But since the microenvironment is no longer embryonic, the confusion leads to cancer: hence as we have argued, tumors become new evolutionary organs perhaps in search of an elusive homeostasis.
Collapse
Affiliation(s)
- Ramray Bhat
- Department of Cancer & DNA Damage Responses, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | |
Collapse
|
10
|
Dai J, Kuang Y, Fang B, Gong H, Lu S, Mou Z, Sun H, Dong Y, Lu J, Zhang W, Zhang J, Wang Z, Wang X, Shen G. The effect of overexpression of Dlx2 on the migration, proliferation and osteogenic differentiation of cranial neural crest stem cells. Biomaterials 2013; 34:1898-910. [DOI: 10.1016/j.biomaterials.2012.11.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 11/27/2012] [Indexed: 11/24/2022]
|
11
|
Maeda H, Wada N, Tomokiyo A, Monnouchi S, Akamine A. Prospective potency of TGF-β1 on maintenance and regeneration of periodontal tissue. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:283-367. [PMID: 23809439 DOI: 10.1016/b978-0-12-407696-9.00006-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Periodontal ligament (PDL) tissue, central in the periodontium, plays crucial roles in sustaining tooth in the bone socket. Irreparable damages of this tissue provoke tooth loss, causing a decreased quality of life. The question arises as to how PDL tissue is maintained or how the lost PDL tissue can be regenerated. Stem cells included in PDL tissue (PDLSCs) are widely accepted to have the potential to maintain or regenerate the periodontium, but PDLSCs are very few in number. In recent studies, undifferentiated clonal human PDL cell lines were developed to elucidate the applicable potentials of PDLSCs for the periodontal regenerative medicine based on cell-based tissue engineering. In addition, it has been suggested that transforming growth factor-beta 1 is an eligible factor for the maintenance and regeneration of PDL tissue.
Collapse
Affiliation(s)
- Hidefumi Maeda
- Department of Endodontology, Kyushu University Hospital, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
12
|
Casini P, Nardi I, Ori M. Hyaluronan is required for cranial neural crest cells migration and craniofacial development. Dev Dyn 2011; 241:294-302. [PMID: 22184056 DOI: 10.1002/dvdy.23715] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2011] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Hyaluronan is a crucial glycosaminoglycan of the vertebrate embryonic extracellular matrix able to influence cell behaviour, both by assembling the pericellular matrices and by activating signal transducing receptors such as CD44. RESULTS We showed that the hyaluronan synthases, Has1 and Has2, and CD44 display a dynamic expression pattern during cranial neural crest cells (NCC) development. By knocking down Has1 and Has2 gene functions, we revealed that hyaluronan synthesized by Has1 and Has2 is necessary for the proper development of the visceral skeleton. CONCLUSIONS The data suggest that hyaluronan helps to maintain the active migratory behaviour of cranial NCC, and that its presence around pre-chondrogenic NCC is crucial for their survival. CD44 knock down also suggests that the role of hyaluronan in cranial NCC migration could be mediated, at least in part, by the activation of CD44. These findings contribute to the unveiling of the functional relation between NCC and their extracellular environment during craniofacial development.
Collapse
Affiliation(s)
- Paola Casini
- Unit of Cellular and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
13
|
Banerjee S, Gordon L, Donn TM, Berti C, Moens CB, Burden SJ, Granato M. A novel role for MuSK and non-canonical Wnt signaling during segmental neural crest cell migration. Development 2011; 138:3287-96. [PMID: 21750038 DOI: 10.1242/dev.067306] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Trunk neural crest cells delaminate from the dorsal neural tube as an uninterrupted sheet; however, they convert into segmentally organized streams before migrating through the somitic territory. These neural crest cell streams join the segmental trajectories of pathfinding spinal motor axons, suggesting that interactions between these two cell types might be important for neural crest cell migration. Here, we show that in the zebrafish embryo migration of both neural crest cells and motor axons is temporally synchronized and spatially restricted to the center of the somite, but that motor axons are dispensable for segmental neural crest cell migration. Instead, we find that muscle-specific receptor kinase (MuSK) and its putative ligand Wnt11r are crucial for restricting neural crest cell migration to the center of each somite. Moreover, we find that blocking planar cell polarity (PCP) signaling in somitic muscle cells also results in non-segmental neural crest cell migration. Using an F-actin biosensor we show that in the absence of MuSK neural crest cells fail to retract non-productive leading edges, resulting in non-segmental migration. Finally, we show that MuSK knockout mice display similar neural crest cell migration defects, suggesting a novel, evolutionarily conserved role for MuSK in neural crest migration. We propose that a Wnt11r-MuSK dependent, PCP-like pathway restricts neural crest cells to their segmental path.
Collapse
Affiliation(s)
- Santanu Banerjee
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6058, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Sellheyer K, Krahl D. Cutaneous mesenchymal stem cells: status of current knowledge, implications for dermatopathology. J Cutan Pathol 2009; 37:624-34. [PMID: 20002239 DOI: 10.1111/j.1600-0560.2009.01477.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Stem cell biology is currently making its impact on medicine, which will probably increase over the next decades. It not only influences our therapeutic thinking caused by the enormous plasticity of stem cells but also affects diagnostic and conceptual aspects of dermatopathology. Although our knowledge of the keratinocytic stem cells located within the follicular bulge has exploded exponentially since their discovery in 1990, the concept of cutaneous mesenchymal stem cells (MSCs) is new. Described initially in 2001 in mice, MSCs later were also found in the human dermis. The connective tissue sheath and the papilla of the hair follicle probably represent the anatomical niche for cutaneous MSCs. In line with the cancer stem cell hypothesis, mutations of these cells may be the underlying basis of mesenchymal skin neoplasms, such as dermatofibrosarcoma protuberans. Furthermore, research on cutaneous MSCs may impact our thinking on the interaction of the epithelial component of skin neoplasms with their surrounding stroma. We are only in the early stages to recognize the importance of the potential contributions of cutaneous MSC research to dermatopathology, but it is not inconceivable to assume that they could be tremendous, paralleling the early discovery of the follicular bulge as a stem cell niche.
Collapse
Affiliation(s)
- Klaus Sellheyer
- Department of Dermatology, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | | |
Collapse
|
15
|
Noonan syndrome is associated with enhanced pERK activity, the repression of which can prevent craniofacial malformations. Proc Natl Acad Sci U S A 2009; 106:15436-41. [PMID: 19706403 DOI: 10.1073/pnas.0903302106] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A gain of function mutation in SHP2, a protein phosphatase encoded by PTPN11, causes Noonan syndrome (NS), which is characterized in part by developmental deficits in both the cardiac and skull fields. Previously, we found that expression of the mutated protein SHP2 Q79R in the heart led to a phenotypic presentation that mimicked some aspects of NS and that this was dependent upon activation of the ERK1/2 pathway. To understand the role that ERK1/2 signaling plays in skull development through signaling in the neural crest, we explored the consequences of Q79R expression in neural crest cells, which contribute to a subset of the bony and cartilaginous structures of the skull. Hyperactivation of ERK1/2 led to craniofacial defects that included smaller skull lengths, greater inner canthal distances, and taller frontal bone heights. In proportion to the smaller skull length, mandibular bone length was also reduced. Inhibition of ERK1/2 hyperactivity as a result of Q79R expression was achieved by injection of the MAPK/ERK kinase inhibitor U0126 during pregnancy. The drug effectively decreased the severity of the craniofacial defects and restored normal skull shape and fontanelle closure. X-ray computer-assisted microtomography analysis of the head confirmed that decreasing ERK1/2 activity led to an abrogation of the craniofacial deficits and brain shape changes that presented in the mice. These data show that normal ERK1/2 signaling in the neural crest is imperative for normal craniofacial development and offer insight into how the heart and craniofacial developmental fields might be affected in some congenital syndromic presentations.
Collapse
|
16
|
Cooley MA, Kern CB, Fresco VM, Wessels A, Thompson RP, McQuinn TC, Twal WO, Mjaatvedt CH, Drake CJ, Argraves WS. Fibulin-1 is required for morphogenesis of neural crest-derived structures. Dev Biol 2008; 319:336-45. [PMID: 18538758 DOI: 10.1016/j.ydbio.2008.04.029] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 04/22/2008] [Accepted: 04/24/2008] [Indexed: 11/24/2022]
Abstract
Here we report that mouse embryos homozygous for a gene trap insertion in the fibulin-1 (Fbln1) gene are deficient in Fbln1 and exhibit cardiac ventricular wall thinning and ventricular septal defects with double outlet right ventricle or overriding aorta. Fbln1 nulls also display anomalies of aortic arch arteries, hypoplasia of the thymus and thyroid, underdeveloped skull bones, malformations of cranial nerves and hemorrhagic blood vessels in the head and neck. The spectrum of malformations is consistent with Fbln1 influencing neural crest cell (NCC)-dependent development of these tissues. This is supported by evidence that Fbln1 expression is associated with streams of cranial NCCs migrating adjacent to rhombomeres 2-7 and that Fbln1-deficient embryos display patterning anomalies of NCCs forming cranial nerves IX and X, which derive from rhombomeres 6 and 7. Additionally, Fbln1-deficient embryos show increased apoptosis in areas populated by NCCs derived from rhombomeres 4, 6 and 7. Based on these findings, it is concluded that Fbln1 is required for the directed migration and survival of cranial NCCs contributing to the development of pharyngeal glands, craniofacial skeleton, cranial nerves, aortic arch arteries, cardiac outflow tract and cephalic blood vessels.
Collapse
Affiliation(s)
- Marion A Cooley
- Department of Cell Biology and Anatomy, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Strachan LR, Condic ML. Neural crest motility on fibronectin is regulated by integrin activation. Exp Cell Res 2007; 314:441-52. [PMID: 18036522 DOI: 10.1016/j.yexcr.2007.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 10/10/2007] [Accepted: 10/25/2007] [Indexed: 10/25/2022]
Abstract
Cell migration is essential for proper development of numerous structures derived from embryonic neural crest cells (NCCs). Although recent work has shown that receptor recycling plays an important role in NCC motility on laminin, the molecular mechanisms regulating NCC motility on fibronectin remain unclear. One mechanism by which cells regulate motility is by modulating the affinity of integrin receptors. Here, we provide evidence that cranial and trunk NCCs rely on functional regulation of integrins to migrate efficiently on fibronectin (FN) in vitro. For NCCs cultured on fibronectin, velocity decreases after Mn2+ application (a treatment that activates all surface integrins) while velocity on laminin (LM) is not affected. The distribution of activated integrin beta 1 receptors on the surface of NCCs is also substratum-dependent. Integrin activation affects cranial and trunk NCCs differently when cultured on different concentrations of FN substrata; only cranial NCCs slow in a FN concentration-dependent manner. Furthermore, Mn2+ treatment alters the distribution and number of activated integrin beta 1 receptors on the surface of cranial and trunk NCCs in different ways. We provide a hypothesis whereby a combination of activated surface integrin levels and the degree to which those receptors are clustered determines NCC motility on fibronectin.
Collapse
Affiliation(s)
- L R Strachan
- Department of Neurobiology and Anatomy, University of Utah, School of Medicine, 20 North 1900 East, Salt Lake City, UT 84132-3401, USA
| | | |
Collapse
|
18
|
Clagett-Dame M, McNeill EM, Muley PD. Role of all-trans retinoic acid in neurite outgrowth and axonal elongation. ACTA ACUST UNITED AC 2006; 66:739-56. [PMID: 16688769 DOI: 10.1002/neu.20241] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The vitamin A metabolite, all-trans retinoic acid (atRA) plays essential roles in nervous system development, including neuronal patterning, survival, and neurite outgrowth. Our understanding of how the vitamin A acid functions in neurite outgrowth comes largely from cultured embryonic neurons and model neuronal cell systems including human neuroblastoma cells. Specifically, atRA has been shown to increase neurite outgrowth from embryonic DRG, sympathetic, spinal cord, and olfactory receptor neurons, as well as dissociated cerebra and retina explants. A role for atRA in axonal elongation is also supported by a limited number of studies in vivo, in which a deficiency in retinoid signaling produced either by dietary or genetic means has been shown to alter neurite outgrowth from the spinal cord and hindbrain regions. Human neuroblastoma cells also show enhanced numbers of neurites and longer processes in response to atRA. The mechanism whereby retinoids regulate neurite outgrowth includes, but is not limited to, the regulation of the transcription of neurotrophin receptors. More recent evidence supports a role for atRA in regulating components of other signaling pathways or candidate neurite-regulating factors. Some of these effects, such as that on neuron navigator 2 (NAV2), may be direct, whereas others may be secondary to other atRA-induced changes in the cell. This review focuses on what is currently known about neurite initiation and growth, with emphasis on the manner in which atRA may influence these events.
Collapse
Affiliation(s)
- Margaret Clagett-Dame
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, Wisconsin 53706, USA.
| | | | | |
Collapse
|
19
|
Baffi MO, Moran MA, Serra R. Tgfbr2 regulates the maintenance of boundaries in the axial skeleton. Dev Biol 2006; 296:363-74. [PMID: 16824508 PMCID: PMC1800905 DOI: 10.1016/j.ydbio.2006.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 05/27/2006] [Accepted: 06/01/2006] [Indexed: 02/01/2023]
Abstract
Previously, we showed that deletion of the TGF-beta type II receptor (Tgfbr2) in Type II Collagen (Col2a) expressing cells results in defects in the development of the axial skeleton. Defects included a reduction in size and alterations in the shape of specific vertebral elements. Anterior lateral and dorsal elements of the vertebrae were missing or irregularly shaped. Vertebral bodies were only mildly affected, but the intervertebral disc (IVD) was reduced or missing. In this manuscript, we show that alterations in the initiation or proliferation of cartilage are not detected in the axial skeleton. However, the expression domain of Fibromodulin (Fmod), a marker of the IVD, was reduced and the area of the future IVD contained peanut agglutinin (PNA) staining cartilage. Next, we show that the expression domains of Pax1 and Pax9, which are preferentially expressed in the caudal sclerotome, are expanded over the entire rostral to caudal length of the sclerotome segment. Dorsal-ventral patterning was not affected in these mice as accessed by expression of Pax1, Pax9, and Msx1. Proliferation was modestly reduced in the loose cells of the sclerotome. The results suggest that signaling through Tgfbr2 regulates the maintenance of boundaries in the sclerotome and developing axial skeleton.
Collapse
Affiliation(s)
| | | | - Rosa Serra
- *Corresponding author. Fax: +1 205 975 5648. E-mail address: (R. Serra)
| |
Collapse
|
20
|
Lin Y, Yan Z, Liu L, Qiao J, Jing W, Wu L, Chen X, Li Z, Tang W, Zheng X, Tian W. Proliferation and pluripotency potential of ectomesenchymal cells derived from first branchial arch. Cell Prolif 2006; 39:79-92. [PMID: 16542344 PMCID: PMC6496172 DOI: 10.1111/j.1365-2184.2006.00374.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2005] [Accepted: 11/14/2005] [Indexed: 01/08/2023] Open
Abstract
Cranial neural crest-derived ectomesenchymal cells are multipotential progenitors that contribute to various tissue types during embryogenesis. Their potential to be expanded in culture as a monolayer and to be induced into different cell lineages in vitro has not been previously reported in detail. In this study, the ectomesenchymal cells in the first branchial arch were enzymatically isolated from the mandibular processes of BALB/c mice and were maintained in an intact state in a medium containing leukaemia inhibitory factor. Here, we first evaluated the proliferative activity of the cells after the third passage, using bromodeoxyuridine labelling and in situ hybridization of telomerase mRNA. Positive staining for expression of HNK-1, S-100 and vimentin confirmed that the population of stem cells originated from the ectomesenchyme, which did not express cytokeratin. Then we investigated the molecular and cellular characteristics of the ectomesenchymal cells during their differentiation towards neurogenic, endothelial, myogenic and odontogenic lineages. Expression of multiple lineage-specific genes and proteins was detected by utilizing a range of molecular and biochemical approaches when the cells were transferred to inductive medium. Histological and immunohistochemical analysis of the induced cells at various intervals indicated obvious phenotypic alteration and presence of specific proteins for the differentiated lineages, for example nestin, factor VIII, alpha-SMA and dentin sialophosphoprotein (DSPP), respectively. Correlatively, results of reverse transcription-PCR corroborated at mRNA level the expression of the characteristic molecules during differentiation. Therefore, it is suggested that the ectomesenchymal cells derived from the first branchial arch may represent a novel source of multipotential stem cells capable of undergoing expansion and variant differentiation in vitro.
Collapse
Affiliation(s)
- Yunfeng Lin
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
- Key Laboratory of Oral Biomedical Engineering, Ministry of Education. Sichuan University, Chengdu 610041, China
| | - Zhengbin Yan
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lei Liu
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ju Qiao
- Department of Orthodontics, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wei Jing
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Wu
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
- Key Laboratory of Oral Biomedical Engineering, Ministry of Education. Sichuan University, Chengdu 610041, China
| | - Xizhe Chen
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhiyong Li
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wei Tang
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiaohui Zheng
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Weidong Tian
- Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, China
- Key Laboratory of Oral Biomedical Engineering, Ministry of Education. Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Abstract
The embryonic head is populated by two robust mesenchymal populations, paraxial mesoderm and neural crest cells. Although the developmental histories of each are distinct and separate, they quickly establish intimate relations that are variably important for the histogenesis and morphogenesis of musculoskeletal components of the calvaria, midface and branchial regions. This review will focus first on the genesis and organization within nascent mesodermal and crest populations, emphasizing interactions that probably initiate or augment the establishment of lineages within each. The principal goal is an analysis of the interactions between crest and mesoderm populations, from their first contacts through their concerted movements into peripheral domains, particularly the branchial arches, and continuing to stages at which both the differentiation and the integrated three-dimensional assembly of vascular, connective and muscular tissues is evident. Current views on unresolved or contentious issues, including the relevance of head somitomeres, the processes by which crest cells change locations and constancy of cell-cell relations at the crest-mesoderm interface, are addressed.
Collapse
Affiliation(s)
- Drew M Noden
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca 14853, USA.
| | | |
Collapse
|
22
|
Sun Rhodes LS, Merzdorf CS. The zic1 gene is expressed in chick somites but not in migratory neural crest. Gene Expr Patterns 2006; 6:539-45. [PMID: 16451832 DOI: 10.1016/j.modgep.2005.10.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2005] [Revised: 10/18/2005] [Accepted: 10/19/2005] [Indexed: 10/25/2022]
Abstract
Zic transcription factors regulate the expression of neural and neural crest-specific genes and are expressed in the cells of the dorsal neural tube and the premigratory neural crest. Here we characterize zic1 expression in the chick embryo during somite formation and neural crest migration. zic1 is expressed in the dorsomedial portion of epithelial somites and subsequently in the dorsomedial lip of the dermomyotome. Although zic1 is expressed in cells of the nascent myotome, it is absent from differentiated myotome cells that express myosin. As the dorsal root ganglia form, zic1 is expressed at high levels in the dorsal sclerotome and zic1 expression is more pronounced in the caudal regions of the somites. Double-label experiments showed that cells expressing zic1 are not labeled by the HNK-1 antibody specific for migratory neural crest cells. Thus, migrating neural crest cells do not express zic1.
Collapse
Affiliation(s)
- Lisa S Sun Rhodes
- Department of Cell Biology and Neuroscience, Montana State University, 513 Leon Johnson Hall, Bozeman, MT 59717, USA
| | | |
Collapse
|
23
|
Ishii M, Han J, Yen HY, Sucov HM, Chai Y, Maxson RE. Combined deficiencies of Msx1 and Msx2 cause impaired patterning and survival of the cranial neural crest. Development 2005; 132:4937-50. [PMID: 16221730 DOI: 10.1242/dev.02072] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The neural crest is a multipotent, migratory cell population that contributes to a variety of tissues and organs during vertebrate embryogenesis. Here, we focus on the function of Msx1 and Msx2, homeobox genes implicated in several disorders affecting craniofacial development in humans. We show that Msx1/2 mutants exhibit profound deficiencies in the development of structures derived from the cranial and cardiac neural crest. These include hypoplastic and mispatterned cranial ganglia, dysmorphogenesis of pharyngeal arch derivatives and abnormal organization of conotruncal structures in the developing heart. The expression of the neural crest markers Ap-2alpha, Sox10 and cadherin 6 (cdh6) in Msx1/2 mutants revealed an apparent retardation in the migration of subpopulations of preotic and postotic neural crest cells, and a disorganization of neural crest cells paralleling patterning defects in cranial nerves. In addition, normally distinct subpopulations of migrating crest underwent mixing. The expression of the hindbrain markers Krox20 and Epha4 was altered in Msx1/2 mutants, suggesting that defects in neural crest populations may result, in part, from defects in rhombomere identity. Msx1/2 mutants also exhibited increased Bmp4 expression in migratory cranial neural crest and pharyngeal arches. Finally, proliferation of neural crest-derived mesenchyme was unchanged, but the number of apoptotic cells was increased substantially in neural crest-derived cells that contribute to the cranial ganglia and the first pharyngeal arch. This increase in apoptosis may contribute to the mispatterning of the cranial ganglia and the hypoplasia of the first arch.
Collapse
Affiliation(s)
- Mamoru Ishii
- Department of Biochemistry and Molecular Biology, Norris Cancer Hospital, Keck School of Medicine, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA 90089, USA
| | | | | | | | | | | |
Collapse
|
24
|
Leone DP, Relvas JB, Campos LS, Hemmi S, Brakebusch C, Fässler R, Ffrench-Constant C, Suter U. Regulation of neural progenitor proliferation and survival by beta1 integrins. J Cell Sci 2005; 118:2589-99. [PMID: 15928047 DOI: 10.1242/jcs.02396] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells give rise to undifferentiated nestin-positive progenitors that undergo extensive cell division before differentiating into neuronal and glial cells. The precise control of this process is likely to be, at least in part, controlled by instructive cues originating from the extracellular environment. Some of these cues are interpreted by the integrin family of extracellular matrix receptors. Using neurosphere cell cultures as a model system, we show that beta1-integrin signalling plays a crucial role in the regulation of progenitor cell proliferation, survival and migration. Following conditional genetic ablation of the beta1-integrin allele, and consequent loss of beta1-integrin cell surface protein, mutant nestin-positive progenitor cells proliferate less and die in higher numbers than their wild-type counterparts. Mutant progenitor cell migration on different ECM substrates is also impaired. These effects can be partially compensated by the addition of exogenous growth factors. Thus, beta1-integrin signalling and growth factor signalling tightly interact to control the number and migratory capacity of nestin-positive progenitor cells.
Collapse
Affiliation(s)
- Dino P Leone
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH Hönggerberg, Zürich
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Greene RM, Pisano MM. Recent advances in understanding transforming growth factor beta regulation of orofacial development. Hum Exp Toxicol 2005; 24:1-12. [PMID: 15727050 DOI: 10.1191/0960327105ht492oa] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Members of the transforming growth factor (TGF) family have emerged as critical contributors to the choreography of cellular and tissue interactions underlying morphogenesis of the orofacial region. The TGFs beta, and their downstream effector molecules, the Smads, play a pivotal role in normal as well as abnormal development of first branchial arch structures. Components of the TGFbeta signal transduction machinery are discussed in relation to regulation of transcription, cell division and tissue differentiation in developing orofacial tissue, as evidence for a functional linkage between the TGFbeta and retinoic acid signal transduction pathways during orofacial development.
Collapse
Affiliation(s)
- Robert M Greene
- University of Louisville Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, ULSD, Louisville, KY 40292, USA.
| | | |
Collapse
|
26
|
Manganas LN, Maletic-Savatic M. Stem cell therapy for central nervous system demyelinating disease. Curr Neurol Neurosci Rep 2005; 5:225-31. [PMID: 15865888 PMCID: PMC4031751 DOI: 10.1007/s11910-005-0050-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent advances in cell-based therapies for demyelinating central nervous system diseases have demonstrated the ability to restore damaged neuronal architecture and function. Demyelinated axons in patients with multiple sclerosis can spontaneously remyelinate over time; however, the rate and extent at which remyelination occurs is inadequate for complete recovery. Previous attempts aimed at regenerating myelin-forming cells have been successful but limited by the multifocal nature of the lesions and the inability to produce large numbers of myelin-producing cells in culture. Stem cell-based therapy can overcome these limitations to some extent and may prove useful in the future treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Louis N. Manganas
- Department of Neurology, Stony Brook University, Stony Brook, NY 11794-8121, Tel: 631-444-8120, Fax:631-444-1474, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, Tel 516-367-6827, Fax 516-367-6805
| | - Mirjana Maletic-Savatic
- Department of Neurology, Stony Brook University, Stony Brook, NY 11794-8121, Tel: 631-444-8120, Fax:631-444-1474, Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, Tel 516-367-6827, Fax 516-367-6805
| |
Collapse
|
27
|
Masuda T, Shiga T. Chemorepulsion and cell adhesion molecules in patterning initial trajectories of sensory axons. Neurosci Res 2005; 51:337-47. [PMID: 15740797 DOI: 10.1016/j.neures.2005.01.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Revised: 01/06/2005] [Accepted: 01/07/2005] [Indexed: 02/02/2023]
Abstract
Research in the past decade has advanced our knowledge of the key role that diffusible cues play in axonal guidance during development. In higher vertebrates, dorsal root ganglion (DRG) neurons extend axons centrally to the spinal cord through the dorsal root entry zone and peripherally to muscle and skin targets. In this review, we focus on the role of proximate "non-target" tissues in the initial stages of DRG axonal growth. In the early stages of development, "non-target" tissues including the dermamyotome, the notochord, and the ventral spinal cord exert chemorepulsion for DRG axons. We describe how semaphorin 3A, chondroitin sulfate proteogrycans, and cell adhesion molecules participate in chemorepulsion and the way they provide spatio-temporal specificity to chemorepulsion. Axon chemorepulsion may act not only to shape DRG axonal trajectories but it also affects a variety of other axonal projections in the peripheral and central nervous system.
Collapse
Affiliation(s)
- Tomoyuki Masuda
- Department of Anatomy, Fukushima Medical University, School of Medicine, Fukushima 960-1295, Japan
| | | |
Collapse
|
28
|
Strachan LR, Condic ML. Cranial neural crest recycle surface integrins in a substratum-dependent manner to promote rapid motility. ACTA ACUST UNITED AC 2004; 167:545-54. [PMID: 15520227 PMCID: PMC2172496 DOI: 10.1083/jcb.200405024] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell migration is essential for proper development of numerous structures derived from embryonic neural crest cells (NCCs). Although the migratory pathways of NCCs have been determined, the molecular mechanisms regulating NCC motility remain unclear. NCC migration is integrin dependent, and recent work has shown that surface expression levels of particular integrin alpha subunits are important determinants of NCC motility in vitro. Here, we provide evidence that rapid cranial NCC motility on laminin requires integrin recycling. NCCs showed both ligand- and receptor-specific integrin regulation in vitro. On laminin, NCCs accumulated internalized laminin but not fibronectin receptors over 20 min, whereas on fibronectin neither type of receptor accumulated internally beyond 2 min. Internalized laminin receptors colocalized with receptor recycling vesicles and were subsequently recycled back to the cell surface. Blocking receptor recycling with bafilomycin A inhibited NCC motility on laminin, indicating that substratum-dependent integrin recycling is essential for rapid cranial neural crest migration.
Collapse
Affiliation(s)
- Lauren R Strachan
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
29
|
Chen X, Kojima SI, Borisy GG, Green KJ. p120 catenin associates with kinesin and facilitates the transport of cadherin-catenin complexes to intercellular junctions. ACTA ACUST UNITED AC 2004; 163:547-57. [PMID: 14610057 PMCID: PMC2173663 DOI: 10.1083/jcb.200305137] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
p120 catenin (p120) is a component of adherens junctions and has been implicated in regulating cadherin-based cell adhesion as well as the activity of Rho small GTPases, but its exact roles in cell–cell adhesion are unclear. Using time-lapse imaging, we show that p120-GFP associates with vesicles and exhibits unidirectional movements along microtubules. Furthermore, p120 forms a complex with kinesin heavy chain through the p120 NH2-terminal head domain. Overexpression of p120, but not an NH2-terminal deletion mutant deficient in kinesin binding, recruits endogenous kinesin to N-cadherin. Disruption of the interaction between N-cadherin and p120, or the interaction between p120 and kinesin, leads to a delayed accumulation of N-cadherin at cell–cell contacts during calcium-initiated junction reassembly. Our analyses identify a novel role of p120 in promoting cell surface trafficking of cadherins via association and recruitment of kinesin.
Collapse
Affiliation(s)
- Xinyu Chen
- Departments of Pathology and Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
30
|
Strachan LR, Condic ML. Neural crest motility and integrin regulation are distinct in cranial and trunk populations. Dev Biol 2003; 259:288-302. [PMID: 12871702 DOI: 10.1016/s0012-1606(03)00187-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The neural crest is a transient cell population that travels long distances through the embryo to form a wide range of derivatives. The extensive migration of the neural crest is highly unusual and incompletely understood. We examined the ability of neural crest cells (NCCs) to migrate under different conditions in vitro. Unlike most motile cell types, avian NCCs migrate efficiently on a wide range of fibronectin concentrations. Strikingly, the migration of NCCs on laminin depends on the axial level from which the crest is derived. On high concentrations of laminin, cranial NCCs migrate at approximately twice the rate of trunk NCCs and show greater persistence, a higher percentage of migratory cells, and a less organized cytoskeleton. The difference in migration between cranial and trunk neural crest is not due to transcriptional differences in integrin mRNA, but rather to differences in posttranslational regulation. Overexpression of a single integrin is sufficient to significantly slow the migration velocity of cranial neural crest cultured on high laminin densities. These results demonstrate that neural crest cells accommodate a wide range of ECM concentrations in vitro and suggest that differences in integrin regulation along the anterior-posterior axis may contribute to differences in neural crest migration and cell fate.
Collapse
Affiliation(s)
- L R Strachan
- Department of Neurobiology and Anatomy, University of Utah, School of Medicine, 20 North 1900 East, Salt Lake City, UT 84132-3401, USA
| | | |
Collapse
|
31
|
Keklikoğlu N. The role played by neural crest cells in the development of syndrome X: a hypothesis. Med Hypotheses 2003; 60:707-10. [PMID: 12710907 DOI: 10.1016/s0306-9877(03)00032-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study evaluated the components and the related hormone releasing organs, the mechanism of action of these hormones, and the origins of the cells and organs in a disorder known as metabolic syndrome X, the prevalence of which is rapidly increasing particularly among the adult population of industrial societies. Metabolic syndrome X, involving carbohydrate and lipid metabolism disorders and cardiovascular disorders caused by the release of primary hormones, alpha and beta cells of the islets of Langerhans of pancreas, adrenal gland medulla's chromaffin cells, sympathetic nervous system neurons and ganglia, adenohypophysis, neurohypophysis and hypothalamus, were evaluated for being neuroectodermal and mainly having neural crest origin. Generally the cells, tissues and organs involved are neuroectodermal and especially have neural crest origins; and to conclude, in prevention, diagnosis and treatment of this disorder the priority is the analysis and understanding of the migration, differentiation and adaptation at the target of neural crest cells.
Collapse
Affiliation(s)
- Nurullah Keklikoğlu
- Department of Histology and Embryology, Faculty of Dentistry, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
32
|
Abstract
The prospects for tooth regeneration in the 21st century are compelling. Using the foundations of experimental embryology, developmental and molecular biology, the principles of biomimetics (the mimicking of biological processes), tooth regeneration is becoming a realistic possibility within the next few decades. The cellular, molecular, and developmental "rules" for tooth morphogenesis are rapidly being discovered. The knowledge gained from adult stem cell biology, especially associated with dentin, cartilage, and bone tissue regeneration, provides additional opportunities for eventual tooth organogenesis. The centuries of tooth development using xenotransplantation, allotransplantation, and autotransplantation have resulted in many important insights that can enhance tooth regeneration. In considering the future, several lines of evidence need to be considered: (1) enamel organ epithelia and dental papilla mesenchyme tissues contain stem cells during postnatal stages of life; (2) late cap stage and bell stage tooth organs contain stem cells; (3) odontogenic adult stem cells respond to mechanical as well as chemical "signals"; (4) presumably adult bone marrow as well as dental pulp tissues contain "odontogenic" stem cells; and (5) epithelial-mesenchymal interactions are pre-requisite for tooth regeneration. The authors express "guarded enthusiasm," yet there should be little doubt that adult stem cell-mediated tooth regeneration will be realized in the not too distant future. The prospects for tooth regeneration could be realized in the next few decades and could be rapidly utilized to improve the quality of human life in many nations around the world.
Collapse
Affiliation(s)
- Yang Chai
- School of Dentistry, University of Southern California, Los Angeles 90089-0641, USA
| | | |
Collapse
|
33
|
Quiñones JL, Rosa R, Ruiz DL, García-Arrarás JE. Extracellular matrix remodeling and metalloproteinase involvement during intestine regeneration in the sea cucumber Holothuria glaberrima. Dev Biol 2002; 250:181-97. [PMID: 12297105 DOI: 10.1006/dbio.2002.0778] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The sea cucumber, Holothuria glaberrima, has the capacity to regenerate its internal organs. Intestinal regeneration is accomplished by the thickening of the mesenteric border and the invasion of this thickening by mucosal epithelium from the esophagus and the cloaca. Extracellular matrix (ECM) remodeling has been associated with morphogenetic events during embryonic development and regeneration. We have used immunohistochemical techniques against ECM components to show that differential changes occur in the ECM during early regeneration. Labeling of fibrous collagenous components and muscle-related laminin disappear from the regenerating intestine and mesentery, while fibronectin labeling and 4G7 (an echinoderm ECM component) are continuously present. Western blots confirm a decrease in fibrous collagen content during the first 2 weeks of regeneration. We have also identified five 1,10-phenanthroline-sensitive bands in collagen gelatin zymographs. The gelatinolytic activities of these bands are enhanced during early stages of regeneration, suggesting that the metalloprotease activity is associated with ECM remodeling. Inhibition of MMPs in vivo with 1,10-phenanthroline, p-aminobenzoyl-Gly-Pro-D-Leu-D-Ala hydroxamate or N-CBZ-Pro-Leu-Gly hydroxamate produces a reversible inhibition of intestinal regeneration and ECM remodeling. Our results show that significant changes in ECM content occur during intestine regeneration in the sea cucumber and that the onset of these changes is correlated to the proteolytic activities of MMPs.
Collapse
Affiliation(s)
- José L Quiñones
- Department of Biology, University of Puerto Rico, Río Piedras, Puerto Rico 00931, USA
| | | | | | | |
Collapse
|
34
|
Abstract
Cell migration occurs in many different contexts. Amoebae and other isolated cells migrate in culture. In animals, 'professional' migratory cells of the immune system constantly survey the body for intruders, whereas other cell types perform specific developmentally regulated migrations. One simple model for the latter type of event is migration of border cells during Drosophila oogenesis. Recent findings have shed light on how border cell fate is induced and on how the migration is guided. This article discusses the implications of these studies and compares (invasive) migration through a tissue with what is known about cells crawling on a flat substratum.
Collapse
|
35
|
Peters J, Sechrist J, Luetolf S, Loredo G, Bronner-Fraser M. Spatial expression of the alternatively spliced EIIIB and EIIIA segments of fibronectin in the early chicken embryo. CELL COMMUNICATION & ADHESION 2002; 9:221-38. [PMID: 12699090 DOI: 10.1080/15419060216015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Using domain-specific antibodies, we have analyzed the tissue distribution of fibronectins (FNs) containing the alternatively spliced EIIIB and EIIIA segments relative to total FN in early chicken embryos. The results show a selective loss of EIIIA+ FN staining in the notochordal sheath and in cartilaginous structures between 4.5 and 7.0 days of development. In other regions, EIIIB+ and EIIIA+ FNs are extensively codistributed in and around mesoderm-derived structures (somites, notochord, heart, and blood vessels), in basal laminae of endoderm and ectoderm-derived structures, as well as within the vicinity of neural crest formation and migration. We also noted that EIIIA staining overlaps with spatial patterns of distribution that have previously been described for the alpha4 integrin subunit, a component of the EIIIA receptor alpha4beta1.
Collapse
Affiliation(s)
- John Peters
- Sacramento VA Medical Center, VA Northern California Health Care System, Mather, CA 95655, USA.
| | | | | | | | | |
Collapse
|
36
|
Gilmour DT, Maischein HM, Nüsslein-Volhard C. Migration and function of a glial subtype in the vertebrate peripheral nervous system. Neuron 2002; 34:577-88. [PMID: 12062041 DOI: 10.1016/s0896-6273(02)00683-9] [Citation(s) in RCA: 253] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glia-axon interactions are essential for the development and function of the nervous system. We combine in vivo imaging and genetics to address the mechanism by which the migration of these cells is coordinated during embryonic development. Using stable transgenic lines, we have followed the migration of one subset of glial cells and their target axons in living zebrafish embryos. These cells coalesce at an early stage and remain coupled throughout migration, with axons apparently pathfinding for glia. Mutant analysis demonstrates that axons provide instructive cues that are sufficient to control glial guidance. Furthermore, mutations in the transcription factor Sox10/cls uncouple the migration of axons and glia. Finally, genetic ablation of this glial subtype reveals an essential role in nerve fasciculation.
Collapse
Affiliation(s)
- Darren T Gilmour
- Max Planck Institut für Entwicklungsbiologie, Abteilung Genetik, Spemannstrasse 35, 72076 Tübingen, Germany
| | | | | |
Collapse
|
37
|
Mary S, Charrasse S, Meriane M, Comunale F, Travo P, Blangy A, Gauthier-Rouvière C. Biogenesis of N-cadherin-dependent cell-cell contacts in living fibroblasts is a microtubule-dependent kinesin-driven mechanism. Mol Biol Cell 2002; 13:285-301. [PMID: 11809840 PMCID: PMC65089 DOI: 10.1091/mbc.01-07-0337] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cadherin-mediated cell-cell adhesion is a dynamic process that is regulated during embryonic development, cell migration, and differentiation. Different cadherins are expressed in specific tissues consistent with their roles in cell type recognition. In this study, we examine the formation of N-cadherin-dependent cell-cell contacts in fibroblasts and myoblasts. In contrast to E-cadherin, both endogenous and ectopically expressed N-cadherin shuttles between an intracellular and a plasma membrane pool. Initial formation of N-cadherin-dependent cell-cell contacts results from the recruitment of the intracellular pool of N-cadherin to the plasma membrane. N-cadherin also localizes to the Golgi apparatus and both secretory and endocytotic vesicles. We demonstrate that the intracellular pool of N-cadherin is tightly associated with the microtubule (MT) network and that junction formation requires MTs. In addition, localization of N-cadherin to the cortex is dependent on an intact F-actin cytoskeleton. We show that N-cadherin transport requires the MT network as well as the activity of the MT-associated motor kinesin. In conclusion, we propose that N-cadherin distribution is a regulated process promoted by cell-cell contact formation, which controls the biogenesis and turnover of the junctions through the MT network.
Collapse
Affiliation(s)
- Sophie Mary
- Centre de Recherche de Biochimie Macromoléculaire, Centre National de la Recherche Scientifique Unité Propre de Recherche 1086, 34293 Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
38
|
Lucas RE, Vlangos CN, Das P, Patel PI, Elsea SH. Genomic organisation of the approximately 1.5 Mb Smith-Magenis syndrome critical interval: transcription map, genomic contig, and candidate gene analysis. Eur J Hum Genet 2001; 9:892-902. [PMID: 11840190 DOI: 10.1038/sj.ejhg.5200734] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2001] [Revised: 09/27/2001] [Accepted: 09/28/2001] [Indexed: 01/08/2023] Open
Abstract
Smith-Magenis syndrome (SMS) is a multiple congenital anomalies/mental retardation syndrome associated with an interstitial deletion of chromosome 17 involving band p11.2. SMS is hypothesised to be a contiguous gene syndrome in which the phenotype arises from the haploinsufficiency of multiple, functionally-unrelated genes in close physical proximity, although the true molecular basis of SMS is not yet known. In this study, we have generated the first overlapping and contiguous transcription map of the SMS critical interval, linking the proximal 17p11.2 region near the SMS-REPM and the distal region near D17S740 in a minimum tiling path of 16 BACs and two PACs. Additional clones provide greater coverage throughout the critical region. Not including the repetitive sequences that flank the critical interval, the map is comprised of 13 known genes, 14 ESTs, and six genomic markers, and is a synthesis of Southern hybridisation and polymerase chain reaction data from gene and marker localisation to BACs and PACs and database sequence analysis from the human genome project high-throughput draft sequence. In order to identify possible candidate genes, we performed sequence analysis and determined the tissue expression pattern analysis of 10 novel ESTs that are deleted in all SMS patients. We also present a detailed review of six promising candidate genes that map to the SMS critical region.
Collapse
Affiliation(s)
- R E Lucas
- Genetics Graduate Program, Michigan State University, East Lansing, Michigan, MI 48824, USA
| | | | | | | | | |
Collapse
|
39
|
Prestoz L, Relvas JB, Hopkins K, Patel S, Sowinski P, Price J, ffrench-Constant C. Association between integrin-dependent migration capacity of neural stem cells in vitro and anatomical repair following transplantation. Mol Cell Neurosci 2001; 18:473-84. [PMID: 11922139 DOI: 10.1006/mcne.2001.1037] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In previous transplantation studies using neural stem cell lines immortalized by the temperature-sensitive SV40 large T-antigen, we have shown that animals with experimental hippocampal lesions resulting from four vessel occlusion recover spatial memory functions more effectively when grafted with the MHP36 cell line than with the MHP15 cell line [Gray et al. (1999). Philos. Trans. R. Soc. London Biol. Sci. 354:1407-1421]. In the present study, we have investigated the cellular and molecular basis of these differences in repair capacity both in vivo and in vitro. Using the same model of hippocampal damage we have shown that following transplantation MHP36 cells migrate and align within the damaged CA1 of the ipsilateral hippocampus. MHP15 cells, in contrast, migrate in a more indiscriminate pattern that does not reflect the anatomy of the region. To analyze the migratory properties of these two cell lines in more detail, we performed migration assays at a nonpermissive temperature on the extracellular matrix substrates laminin, fibronectin, and vitronectin. These showed that MHP36 cells have a greater migration potential than the MHP15 cells. While the pattern of cell surface extracellular matrix receptors of the integrin family was identical in both cell lines, the different degrees of migration on vitronectin were both blocked by inhibitors of alphaV integrins. Differences in integrin signaling therefore contribute to the greater migration potential of the repairing MHP36 cell line.
Collapse
Affiliation(s)
- L Prestoz
- Department of Medical Genetics and Cambridge Centre for Brain Repair, University of Cambridge, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
40
|
Christiansen JH, Coles EG, Wilkinson DG. Molecular control of neural crest formation, migration and differentiation. Curr Opin Cell Biol 2000; 12:719-24. [PMID: 11063938 DOI: 10.1016/s0955-0674(00)00158-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Induction, migration and differentiation of the neural crest are crucial for the development of the vertebrate embryo, and elucidation of the underlying mechanisms remains an important challenge. In the past year, a novel signal regulating the formation of neural crest cells has been identified, and advances have been made in uncovering roles for bone morphogenetic protein signals and for a transcription factor in the onset of neural crest migration. There have been new insights into the migration and plasticity of branchial neural crest cells. Important progress has been made in dissecting the roles of bone morphogenetic protein, Wnt and Notch signalling systems and their associated downstream transcription factors in the control of neural crest cell differentiation.
Collapse
Affiliation(s)
- J H Christiansen
- Division of Developmental Neurobiology, National Institute for Medical Research, The Ridgeway, Mill Hill, NW7 1AA, London, UK
| | | | | |
Collapse
|
41
|
Van De Bor V, Walther R, Giangrande A. Some fly sensory organs are gliogenic and require glide/gcm in a precursor that divides symmetrically and produces glial cells. Development 2000; 127:3735-43. [PMID: 10934018 DOI: 10.1242/dev.127.17.3735] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In flies, the choice between neuronal and glial fates depends on the asymmetric division of multipotent precursors, the neuroglioblast of the central nervous system and the IIb precursor of the sensory organ lineage. In the central nervous system, the choice between the two fates requires asymmetric distribution of the glial cell deficient/glial cell missing (glide/gcm) RNA in the neuroglioblast. Preferential accumulation of the transcript in one of the daughter cells results in the activation of the glial fate in that cell, which becomes a glial precursor. Here we show that glide/gcm is necessary to induce glial differentiation in the peripheral nervous system. We also present evidence that glide/gcm RNA is not necessary to induce the fate choice in the peripheral multipotent precursor. Indeed, glide/gcm RNA and protein are first detected in one daughter of IIb but not in IIb itself. Thus, glide/gcm is required in both central and peripheral glial cells, but its regulation is context dependent. Strikingly, we have found that only subsets of sensory organs are gliogenic and express glide/gcm. The ability to produce glial cells depends on fixed, lineage related, cues and not on stochastic decisions. Finally, we show that after glide/gcm expression has ceased, the IIb daughter migrates and divides symmetrically to produce several mature glial cells. Thus, the glide/gcm-expressing cell, also called the fifth cell of the sensory organ, is indeed a glial precursor. This is the first reported case of symmetric division in the sensory organ lineage. These data indicate that the organization of the fly peripheral nervous system is more complex than previously thought.
Collapse
Affiliation(s)
- V Van De Bor
- Institut de Génétique et Biologie Moléculaire et Cellulaire, IGBMC/CNRS/ULP/INSERM - BP 163 67404 ILLKIRCH, c.u. de Strasbourg, France
| | | | | |
Collapse
|
42
|
Xu Q, Mellitzer G, Wilkinson DG. Roles of Eph receptors and ephrins in segmental patterning. Philos Trans R Soc Lond B Biol Sci 2000; 355:993-1002. [PMID: 11128993 PMCID: PMC1692797 DOI: 10.1098/rstb.2000.0635] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Eph receptor tyrosine kinases and their membrane-bound ligands, ephrins, have key roles in patterning and morphogenesis. Interactions between these molecules are promiscuous, but largely fall into two groups: EphA receptors bind to glycosylphosphatidyl inositol-anchored ephrin-A ligands, and EphB receptors bind to transmembrane ephrin-B proteins. Ephrin-B proteins transduce signals, such that bidirectional signalling can occur upon interaction with the Eph receptor. In many tissues, there are complementary and overlapping expression domains of interacting Eph receptors and ephrins. An important role of Eph receptors and ephrins is to mediate cell contact-dependent repulsion, and this has been implicated in the pathfinding of axons and neural crest cells, and the restriction of cell intermingling between hindbrain segments. Studies in an in vitro system show that bidirectional activation is required to prevent intermingling between cell populations, whereas unidirectional activation can restrict cell communication via gap junctions. Recent work indicates that Eph receptors can also upregulate cell adhesion, but the biochemical basis of repulsion versus adhesion responses is unclear. Eph receptors and ephrins have thus emerged as key regulators that, in parallel with cell adhesion molecules, underlie the establishment and maintenance of patterns of cellular organization.
Collapse
Affiliation(s)
- Q Xu
- Division of Developmental Neurobiology, National Institute for Medical Research, London, UK
| | | | | |
Collapse
|
43
|
Stidwill RP, Christen M. Alteration of fibronectin affinity during differentiation modulates the in vitro migration velocities of Hydra nematocytes. CELL MOTILITY AND THE CYTOSKELETON 2000; 41:68-73. [PMID: 9744300 DOI: 10.1002/(sici)1097-0169(1998)41:1<68::aid-cm6>3.0.co;2-b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In the fresh water Cnidarian Hydra nematocytes differentiate from stem cells in the body column of the polyps and are functional in the tentacles to where they migrate as single cells in an amoeboid fashion. The fluorescent vital stain TROMI (tetramethyl-rhodamine-5/6-maleimide) allows to easily discriminate between cells located in the body column and cells mounted in the tentacles. The two cell populations were found to have different in vitro migration properties. These differences appear to be due largely to a differential attachment to fibronectin. Nematocytes from the tentacles show significantly lower in vitro migration velocities on isolated pieces of the organisms extracellular matrix (the mesoglea) and attach more firmly to fibronectin-coated substrates than cells from the body column. Pretreatment of the mesogleae with antibodies against the cell binding domain of fibronectin or addition of RGD-peptides results in an increase of the average migration velocity of cells from the tentacles and a decreased velocity of the cells from the body column. These findings suggest that (1) modulation of the attachment to fibronectin is decisive for the observed differential migration properties of the two cell populations and (2) the in vitro migration of nematocytes is dependent on subtle and transient interactions of cell surface receptors (most probably integrins) and fibronectin.
Collapse
Affiliation(s)
- R P Stidwill
- Department of Zoology, University of Zürich, Switzerland.
| | | |
Collapse
|
44
|
Wilkinson DG. Eph receptors and ephrins: regulators of guidance and assembly. INTERNATIONAL REVIEW OF CYTOLOGY 2000; 196:177-244. [PMID: 10730216 DOI: 10.1016/s0074-7696(00)96005-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent advances have started to elucidate the developmental functions and biochemistry of Eph receptor tyrosine kinases and their membrane-bound ligands, ephrins. Interactions between these molecules are promiscuous, but they largely fall into two groups: EphA receptors bind to GPI-anchored ephrin-A ligands, while EphB receptors bind to ephrin-B proteins that have a transmembrane and cytoplasmic domain. Remarkably, ephrin-B proteins transduce signals, such that bidirectional signaling can occur upon interaction with Eph receptor. In many tissues, specific Eph receptors and ephrins have complementary domains, whereas other family members may overlap in their expression. An important role of Eph receptors and ephrins is to mediate cell-contact-dependent repulsion. Complementary and overlapping gradients of expression underlie establishment of a topographic map of neuronal projections in the retinotectal system. Eph receptors and ephrins also act at boundaries to channel neuronal growth cones along specific pathways, restrict the migration of neural crest cells, and via bidirectional signaling prevent intermingling between hindbrain segments. Intriguingly, Eph receptors and ephrins can also trigger an adhesive response of endothelial cells and are required for the remodeling of blood vessels. Biochemical studies suggest that the extent of multimerization of Eph receptors modulates the cellular response and that the actin cytoskeleton is one major target of the intracellular pathways activated by Eph receptors. Eph receptors and ephrins have thus emerged as key regulators of the repulsion and adhesion of cells that underlie the establishment, maintenance, and remodeling of patterns of cellular organization.
Collapse
Affiliation(s)
- D G Wilkinson
- Division of Developmental Neurobiology, National Institute for Medical Research, London, United Kingdom
| |
Collapse
|
45
|
Chai Y, Jiang X, Ito Y, Bringas P, Han J, Rowitch DH, Soriano P, McMahon AP, Sucov HM. Fate of the mammalian cranial neural crest during tooth and mandibular morphogenesis. Development 2000; 127:1671-9. [PMID: 10725243 DOI: 10.1242/dev.127.8.1671] [Citation(s) in RCA: 942] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neural crest cells are multipotential stem cells that contribute extensively to vertebrate development and give rise to various cell and tissue types. Determination of the fate of mammalian neural crest has been inhibited by the lack of appropriate markers. Here, we make use of a two-component genetic system for indelibly marking the progeny of the cranial neural crest during tooth and mandible development. In the first mouse line, Cre recombinase is expressed under the control of the Wnt1 promoter as a transgene. Significantly, Wnt1 transgene expression is limited to the migrating neural crest cells that are derived from the dorsal CNS. The second mouse line, the ROSA26 conditional reporter (R26R), serves as a substrate for the Cre-mediated recombination. Using this two-component genetic system, we have systematically followed the migration and differentiation of the cranial neural crest (CNC) cells from E9.5 to 6 weeks after birth. Our results demonstrate, for the first time, that CNC cells contribute to the formation of condensed dental mesenchyme, dental papilla, odontoblasts, dentine matrix, pulp, cementum, periodontal ligaments, chondrocytes in Meckel's cartilage, mandible, the articulating disc of temporomandibular joint and branchial arch nerve ganglia. More importantly, there is a dynamic distribution of CNC- and non-CNC-derived cells during tooth and mandibular morphogenesis. These results are a first step towards a comprehensive understanding of neural crest cell migration and differentiation during mammalian craniofacial development. Furthermore, this transgenic model also provides a new tool for cell lineage analysis and genetic manipulation of neural-crest-derived components in normal and abnormal embryogenesis.
Collapse
Affiliation(s)
- Y Chai
- Center for Craniofacial Molecular Biology School of Dentistry University of Southern California, CSA 103, Los Angeles, CA 90033, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Glover JC. Development of specific connectivity between premotor neurons and motoneurons in the brain stem and spinal cord. Physiol Rev 2000; 80:615-47. [PMID: 10747203 DOI: 10.1152/physrev.2000.80.2.615] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Astounding progress has been made during the past decade in understanding the general principles governing the development of the nervous system. An area of prime physiological interest that is being elucidated is how the neural circuitry that governs movement is established. The concerted application of molecular biological, anatomical, and electrophysiological techniques to this problem is yielding gratifying insight into how motoneuron, interneuron, and sensory neuron identities are determined, how these different neuron types establish specific axonal projections, and how they recognize and synapse upon each other in patterns that enable the nervous system to exercise precise control over skeletal musculature. This review is an attempt to convey to the physiologist some of the exciting discoveries that have been made, within a context that is intended to link molecular mechanism to behavioral realization. The focus is restricted to the development of monosynaptic connections onto skeletal motoneurons. Principal topics include the inductive mechanisms that pattern the placement and differentiation of motoneurons, Ia sensory afferents, and premotor interneurons; the molecular guidance mechanisms that pattern the projection of premotor axons in the brain stem and spinal cord; and the precision with which initial synaptic connections onto motoneurons are established, with emphasis on the relative roles played by cellular recognition versus electrical activity. It is hoped that this review will provide a guide to understanding both the existing literature and the advances that await this rapidly developing topic.
Collapse
Affiliation(s)
- J C Glover
- Department of Anatomy, University of Oslo, Oslo, Norway.
| |
Collapse
|
47
|
Arregui CO, Balsamo J, Lilien J. Regulation of signaling by protein-tyrosine phosphatases: potential roles in the nervous system. Neurochem Res 2000; 25:95-105. [PMID: 10685609 DOI: 10.1023/a:1007595617447] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During neuronal development, cells respond to a variety of environmental cues through cell surface receptors that are coupled to a signaling transduction machinery based on protein tyrosine phosphorylation and dephosphorylation. Receptor and non-receptor tyrosine kinases have received a great deal of attention; however, in the last few years, receptor (plasma membrane associated) and non-receptor protein-tyrosine phosphatases (PTPs) have also been shown to play important roles in development of the nervous system. In many cases PTPs have provocative distribution patterns or have been shown to be associated with specific cell adhesion and growth factor receptors. Additionally, altering PTP expression levels or activity impairs neuronal behavior. In this review we outline what is currently known about the role of PTPs in development, differentiation and neuronal physiology.
Collapse
Affiliation(s)
- C O Arregui
- Department of Biological Sciences, Wayne State University, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
48
|
Abstract
The neural crest is a transient population of multipotent precursor cells named for its site of origin at the crest of the closing neural folds in vertebrate embryos. Following neural tube closure, these cells become migratory and populate diverse regions throughout the embryo where they give rise to most of the neurons and support cells of the peripheral nervous system (PNS), pigment cells, smooth muscle, craniofacial cartilage, and bone. Because of its remarkable ability to generate such diverse derivatives, the neural crest has fascinated developmental biologists for over one hundred years. A great deal has been learned about the migratory pathways neural crest cells follow and the signals that may trigger their differentiation, but until recently comparatively little was known about earlier steps in neural crest development. In the past few years progress has been made in understanding these earlier events, including how the precursors of these multipotent cells are specified in the early embryo and the mechanisms by which they become migratory. In this review, we first examine the mechanisms underlying neural crest induction, paying particular attention to a number of growth factor and transcription factor families that have been implicated in this process. We also discuss when and how the fate of neural crest precursors may diverge from those of nearby neural and epidermal populations. Finally, we review recent advances in our understanding of how neural crest cells become migratory and address the process of neural crest diversification.
Collapse
Affiliation(s)
- C LaBonne
- Division of Biology, California Institute of Technology, Pasadena 91125, USA.
| | | |
Collapse
|
49
|
Nishimura EK, Yoshida H, Kunisada T, Nishikawa SI. Regulation of E- and P-cadherin expression correlated with melanocyte migration and diversification. Dev Biol 1999; 215:155-66. [PMID: 10545227 DOI: 10.1006/dbio.1999.9478] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Melanocytes (Mc) and their progenitors melanoblasts (Mb) are derived from the neural crest and migrate along the dorsolateral pathway to colonize the dermis, the epidermis, and finally the hair matrix. To examine the involvement of cadherins in the migration of Mc lineage cells, we combined flow cytometric analysis of dissociated live cells with immunohistochemical staining of tissue sections to quantify the level of cadherin expression on the surface of Mb/Mc. At 11.5 days postcoitum, Mb are in the dermis and are E-cadherin(-)P-cadherin(-) (E-cad(-)P-cad(-)). During the next 48 h, a 200-fold increase of E-cadherin expression is induced on the surface of Mb prior to their entry into the epidermis, thereby forming a homogeneous E-cad(high)P-cad(-/low) population. The cadherin expression pattern then diversifies, giving rise to three populations, an E-cad(-)P-cad(-) dermal population, E-cad(high)P-cad(low) epidermal population, and E-cad(-)P-cad(med-high) follicular population. In all three populations, the patterns of expression are region-specific, being identical with those of surrounding cells such as keratinocytes and fibroblasts, and are preserved before and after pigmentation. While most of the epidermal Mb/Mc disappear after the neonatal stage in normal mice, forced expression of steel factor in the epidermis of transgenic mice promotes survival of epidermal Mb/Mc, maintaining epidermal-type cadherin expression pattern (E-cad(high)P-cad(low)) throughout the postnatal life. These findings indicate the involvement of extrinsic cues in coordinating the cadherin expression pattern of Mb/Mc and suggest a role for E- and P-cadherins in guiding Mc progenitors to their final destinations.
Collapse
Affiliation(s)
- E K Nishimura
- Faculty of Medicine, Kyoto University, Shogoin Kawaharacho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | | | | | | |
Collapse
|
50
|
Eickholt BJ, Mackenzie SL, Graham A, Walsh FS, Doherty P. Evidence for collapsin-1 functioning in the control of neural crest migration in both trunk and hindbrain regions. Development 1999; 126:2181-9. [PMID: 10207143 DOI: 10.1242/dev.126.10.2181] [Citation(s) in RCA: 168] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Collapsin-1 belongs to the Semaphorin family of molecules, several members of which have been implicated in the co-ordination of axon growth and guidance. Collapsin-1 can function as a selective chemorepellent for sensory neurons, however, its early expression within the somites and the cranial neural tube (Shepherd, I., Luo, Y., Raper, J. A. and Chang, S. (1996) Dev. Biol. 173, 185–199) suggest that it might contribute to the control of additional developmental processes in the chick. We now report a detailed study on the expression of collapsin-1 as well as on the distribution of collapsin-1-binding sites in regions where neural crest cell migration occurs. collapsin-1 expression is detected in regions bordering neural crest migration pathways in both the trunk and hindbrain regions and a receptor for collapsin-1, neuropilin-1, is expressed by migrating crest cells derived from both regions. When added to crest cells in vitro, a collapsin-1-Fc chimeric protein induces morphological changes similar to those seen in neuronal growth cones. In order to test the function of collapsin-1 on the migration of neural crest cells, an in vitro assay was used in which collapsin-1-Fc was immobilised in alternating stripes consisting of collapsin-Fc/fibronectin versus fibronectin alone. Explanted neural crest cells derived from both trunk and hindbrain regions avoided the collapsin-Fc-containing substratum. These results suggest that collapsin-1 signalling can contribute to the patterning of neural crest cell migration in the developing chick.
Collapse
Affiliation(s)
- B J Eickholt
- Molecular Neurobiology Group, GKT School of Medicine, King's College, London Bridge, London SE1 9RT, UK
| | | | | | | | | |
Collapse
|