1
|
Wu IC, Liou JW, Yang CH, Chen JH, Chen KY, Hung CH. Self-assembly of gelatin and collagen in the polyvinyl alcohol substrate and its influence on cell adhesion, proliferation, shape, spreading and differentiation. Front Bioeng Biotechnol 2023; 11:1193849. [PMID: 37520293 PMCID: PMC10375239 DOI: 10.3389/fbioe.2023.1193849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Culture substrates display profound influence on biological and developmental characteristic of cells cultured in vitro. This study investigates the influence of polyvinyl alcohol (PVA) substrates blended with different concentration of collagen or/and gelatin on the cell adhesion, proliferation, shape, spreading, and differentiation of stem cells. The collagen/gelatin blended PVA substrates were prepared by air drying. During drying, blended collagen or/and gelatin can self-assemble into macro-scale nucleated particles or branched fibrils in the PVA substrates that can be observed under the optical microscope. These collagen/gelatin blended substrates revealed different surface topography, z-average, roughness, surface adhesion and Young's modulus as examined by the atomic force microscope (AFM). The results of Fourier transform infrared spectroscopy (FTIR) analysis indicated that the absorption of amide I (1,600-1,700 cm-1) and amide II (1,500-1,600 cm-1) groups increased with increasing collagen and gelatin concentration blended and the potential of fibril formation. These collagen or/and gelatin blended PVA substrates showed enhanced NIH-3T3 fibroblast adhesion as comparing with the pure PVA, control tissue culture polystyrene, conventional collagen-coated and gelatin-coated wells. These highly adhesive PVA substrates also exhibit inhibited cell spreading and proliferation. It is also found that the shape of NIH-3T3 fibroblasts can be switched between oval, spindle and flattened shapes depending on the concentration of collagen or/and gelatin blended. For inductive differentiation of stem cells, it is found that number and ration of neural differentiation of rat cerebral cortical neural stem cells increase with the decreasing collagen concentration in the collagen-blended PVA substrates. Moreover, the PVA substrates blended with collagen or collagen and gelatin can efficiently support and conduct human pluripotent stem cells to differentiate into Oil-Red-O- and UCP-1-positive brown-adipocyte-like cells via ectodermal lineage without the addition of mitogenic factors. These results provide a useful and alternative platform for controlling cell behavior in vitro and may be helpful for future application in the field of regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- I-Chi Wu
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Plastic Surgery Division, Surgical Department, Hualien Armed Forces General Hospital, Hualien City, Taiwan
| | - Je-Wen Liou
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City, Taiwan
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City, Taiwan
| | - Jia-Hui Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Surgery, Taipei Tzu Chi Hospital, New Taipei City, Taiwan
| | - Kuan-Yu Chen
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
- Department of Surgery, New Taipei City Hospital, New Taipei City, Taiwan
| | - Chih-Huang Hung
- Institute of Medical Sciences, Tzu Chi University, Hualien City, Taiwan
| |
Collapse
|
2
|
Michels K, Bohnsack BL. Ophthalmological Manifestations of Axenfeld-Rieger Syndrome: Current Perspectives. Clin Ophthalmol 2023; 17:819-828. [PMID: 36926528 PMCID: PMC10013571 DOI: 10.2147/opth.s379853] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Axenfeld-Rieger syndrome (ARS) is a rare congenital disease that is primarily characterized by ocular anterior segment anomalies but is also associated with craniofacial, dental, cardiac, and neurologic abnormalities. Over half of cases are linked with autosomal dominant mutations in either FOXC1 or PITX2, which reflects the molecular role of these genes in regulating neural crest cell contributions to the eye, face, and heart. Within the eye, ARS is classically defined as the combination of posterior embryotoxon with iris bridging strands (Axenfeld anomaly) and iris hypoplasia causing corectopia and pseudopolycoria (Rieger anomaly). Glaucoma due to iridogoniodysgenesis is the main source of morbidity and is typically diagnosed during infancy or childhood in over half of affected individuals. Angle bypass surgery, such as glaucoma drainage devices and trabeculectomies, is often needed to obtain intraocular pressure control. A multi-disciplinary approach including glaucoma specialists and pediatric ophthalmologists produces optimal outcomes as vision is dependent on many factors including glaucoma, refractive error, amblyopia and strabismus. Further, since ophthalmologists often make the diagnosis, it is important to refer patients with ARS to other specialists including dentistry, cardiology, and neurology.
Collapse
Affiliation(s)
- Kristi Michels
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brenda L Bohnsack
- Division of Ophthalmology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
3
|
Seyyar SA, Tıskaoğlu NS, Gürbostan G, Pekpak E, Sayglı O. Increased Endothelial Cell Density in Childhood Patients With Thalassemia Major. Eye Contact Lens 2021; 47:660-663. [PMID: 34173366 DOI: 10.1097/icl.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE We aimed to compare specular microscopy, biometric, refractive, and anterior segment topographic parameters in children with thalassemia major (TM) with normal children of the same age. MATERIAL METHOD This cross-sectional study included 55 eyes of 55 patients with TM and 63 eyes of 63 age-sex-matched healthy children; all patients with TM were receiving treatment with blood transfusions every 3 to 4 weeks and an oral iron chelator deferasirox (DFX) (ICL670) tablet. A comprehensive ophthalmic examination was performed, including refraction (spherical equivalent), best-corrected visual acuity, slit-lamp bio microscopy, specular examination, optical biometry, intraocular pressure, anterior segment topography, and fundus examination. RESULTS Endothelial cell density (ECD) was 3,138 in the TM group and 2,996 in the control group (P=0.003). The mean central corneal thickness was significantly thinner in the TM group (P=0.010). Flat keratometry (K1) was 43.5 D in the study group and 42.9 D in the control group (P=0.039). The mean anterior chamber depth (ACD) was shallower in the TM group (P=0.004); axial length (AL) was significantly shorter in the TM group (P=0.002). CONCLUSION This study shows that there are differences in ECD, AL, keratometry values, ACD, and anterior segment parameters of pediatric patients with TM compared with healthy controls.
Collapse
Affiliation(s)
- Sevim Ayca Seyyar
- Ophthalmology Department (S.A.S.), Kocaeli Derince Education and Research Hospital; Ophthalmology Department (N.S.T.), Ersin Arslan Education and Research Hospital; and Ophthalmology Department (G.G., E.P., O.S.), Gaziantep University Hospital
| | | | | | | | | |
Collapse
|
4
|
Abstract
Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Shashank Gandhi
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| | - Marianne E Bronner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA; ,
| |
Collapse
|
5
|
Maynard TM, Horvath A, Bernot JP, Karpinski BA, Tavares ALP, Shah A, Zheng Q, Spurr L, Olender J, Moody SA, Fraser CM, LaMantia AS, Lee NH. Transcriptional dysregulation in developing trigeminal sensory neurons in the LgDel mouse model of DiGeorge 22q11.2 deletion syndrome. Hum Mol Genet 2021; 29:1002-1017. [PMID: 32047912 PMCID: PMC7158380 DOI: 10.1093/hmg/ddaa024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/12/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
LgDel mice, which model the heterozygous deletion of genes at human chromosome 22q11.2 associated with DiGeorge/22q11.2 deletion syndrome (22q11DS), have cranial nerve and craniofacial dysfunction as well as disrupted suckling, feeding and swallowing, similar to key 22q11DS phenotypes. Divergent trigeminal nerve (CN V) differentiation and altered trigeminal ganglion (CNgV) cellular composition prefigure these disruptions in LgDel embryos. We therefore asked whether a distinct transcriptional state in a specific population of early differentiating LgDel cranial sensory neurons, those in CNgV, a major source of innervation for appropriate oropharyngeal function, underlies this departure from typical development. LgDel versus wild-type (WT) CNgV transcriptomes differ significantly at E10.5 just after the ganglion has coalesced. Some changes parallel altered proportions of cranial placode versus cranial neural crest-derived CNgV cells. Others are consistent with a shift in anterior-posterior patterning associated with divergent LgDel cranial nerve differentiation. The most robust quantitative distinction, however, is statistically verifiable increased variability of expression levels for most of the over 17 000 genes expressed in common in LgDel versus WT CNgV. Thus, quantitative expression changes of functionally relevant genes and increased stochastic variation across the entire CNgV transcriptome at the onset of CN V differentiation prefigure subsequent disruption of cranial nerve differentiation and oropharyngeal function in LgDel mice.
Collapse
Affiliation(s)
- Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Anelia Horvath
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,McCormick Genomics and Proteomics Center, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - James P Bernot
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Beverly A Karpinski
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Andre L P Tavares
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Ankita Shah
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Qianqian Zheng
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Liam Spurr
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Jacqueline Olender
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Sally A Moody
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Anthony-S LaMantia
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg VA, 24061, USA.,Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Norman H Lee
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
6
|
Guo P, Ji Z, Jiang H, Huang X, Wang C, Pan B. Identification of a novel CYP26A1 mutation in a Chinese family with congenital microtia. Int J Pediatr Otorhinolaryngol 2020; 139:110488. [PMID: 33197841 DOI: 10.1016/j.ijporl.2020.110488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Microtia is defined as a congenital malformation characterized by a small, abnormally shaped auricle, with atresia or stenosis of the auditory canal. This study investigated a mutation of the cytochrome P450, family 26, subfamily A, polypeptide 1(CYP26A1) gene, which is considered important in craniofacial development, in a family affected with microtia. METHODS Whole-exome sequencing (WES) was performed on the proband and his family members to identify disease-associated variants. Computational predictions of the altered protein were analyzed using several bioinformatics tools. The wild-type (WT) and mutant forms of CYP26A1 cDNA were transfected into human embryonic kidney cells, and the mRNA and protein levels were compared using quantitative polymerase chain reaction (qPCR) and Western blot analyses. RESULTS In this two-generation family, the proband and his mother were diagnosed with unilateral microtia. Unilateral microtia and ipsilateral accessory ear were observed in one of the twins, who were sisters of the proband. The father and the other twin showed no abnormal clinical features. A heterozygous mutation of a C to T in the CYP26A1 gene, which leads to truncation of the CYP26A1 protein, was identified in this family. The nonsense mutation cosegregated with patients and was absent in normal members of the family. The prediction software indicated that it was a possibly pathogenic mutation. The structure of the protein varied significantly between the WT and mutant proteins. Functional analysis showed that this mutation caused a significant decrease in both the mRNA and protein levels. CONCLUSIONS Our findings suggest that this mutation of CYP26A1 may be a pathogenic factor leading to the phenotypes of microtia and accessory ear in this family. Further studies are needed to prove the function of this mutation and to explore the possible mechanism by which this variant is involved in the occurrence of microtia.
Collapse
Affiliation(s)
- Peipei Guo
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100032, China.
| | - Zhonglei Ji
- Affiliated Hospital of Weifang Medical University, Shandong, 261031, China
| | - Haiyue Jiang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100032, China
| | - Xin Huang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100032, China
| | - Changchen Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100032, China
| | - Bo Pan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100032, China.
| |
Collapse
|
7
|
Rapizzi E, Benvenuti S, Deledda C, Martinelli S, Sarchielli E, Fibbi B, Luciani P, Mazzanti B, Pantaleo M, Marroncini G, Vannelli GB, Maggi M, Mannelli M, Luconi M, Peri A. A unique neuroendocrine cell model derived from the human foetal neural crest. J Endocrinol Invest 2020; 43:1259-1269. [PMID: 32157664 DOI: 10.1007/s40618-020-01213-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Nowadays, no human neuroendocrine cell models derived from the neural crest are available. In this study, we present non-transformed long-term primary Neural Crest Cells (NCCs) isolated from the trunk region of the neural crest at VIII-XII gestational weeks of human foetuses obtained from voluntary legal abortion. METHODS AND RESULTS In NCC, quantitative real-time RT PCR demonstrated the expression of neural crest specifier genes, such as Snail1, Snail2/SLUG, Sox10, FoxD3, c-Myc, and p75NTR. Moreover, these cell populations expressed stemness markers (such as Nanog and nestin), as well as markers of motility and invasion (TAGLN, MMP9, CXCR4, and CXCR7), and of neuronal/glial differentiation (MAP2, GFAP, SYP, and TAU). Functional analysis demonstrated that these cells not only possessed high migration properties, but most importantly, they expressed markers of sympatho-adrenal lineage, such as ASCL1 and tyrosine hydroxylase (TH). Moreover, the expression of TH increased after the induction with two different protocols of differentiation towards neuronal and sympatho-adrenal phenotypes. Finally, exposure to conditioned culture media from NCC induced a mature phenotype in a neuronal cell model (namely SH-SY5Y), suggesting that NCC may also act like Schwann precursors. CONCLUSION This unique human cell model provides a solid tool for future studies addressing the bases of human neural crest-derived neuroendocrine tumours.
Collapse
Affiliation(s)
- E Rapizzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - S Benvenuti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - C Deledda
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - S Martinelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - E Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - B Fibbi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - P Luciani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - B Mazzanti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - M Pantaleo
- Genetics and Molecular Medicine Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - G Marroncini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - G B Vannelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - M Maggi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
- Istituto Nazionale Biostrutture e Biosistemi (INBB), viale delle Medaglie d'Oro 305, 00136, Rome, Italy
| | - M Mannelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| | - M Luconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy.
- Istituto Nazionale Biostrutture e Biosistemi (INBB), viale delle Medaglie d'Oro 305, 00136, Rome, Italy.
| | - A Peri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
8
|
Moon KR, van Dijk D, Wang Z, Gigante S, Burkhardt DB, Chen WS, Yim K, Elzen AVD, Hirn MJ, Coifman RR, Ivanova NB, Wolf G, Krishnaswamy S. Visualizing structure and transitions in high-dimensional biological data. Nat Biotechnol 2019; 37:1482-1492. [PMID: 31796933 PMCID: PMC7073148 DOI: 10.1038/s41587-019-0336-3] [Citation(s) in RCA: 410] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/29/2019] [Indexed: 01/12/2023]
Abstract
The high-dimensional data created by high-throughput technologies require visualization tools that reveal data structure and patterns in an intuitive form. We present PHATE, a visualization method that captures both local and global nonlinear structure using an information-geometric distance between data points. We compare PHATE to other tools on a variety of artificial and biological datasets, and find that it consistently preserves a range of patterns in data, including continual progressions, branches and clusters, better than other tools. We define a manifold preservation metric, which we call denoised embedding manifold preservation (DEMaP), and show that PHATE produces lower-dimensional embeddings that are quantitatively better denoised as compared to existing visualization methods. An analysis of a newly generated single-cell RNA sequencing dataset on human germ-layer differentiation demonstrates how PHATE reveals unique biological insight into the main developmental branches, including identification of three previously undescribed subpopulations. We also show that PHATE is applicable to a wide variety of data types, including mass cytometry, single-cell RNA sequencing, Hi-C and gut microbiome data.
Collapse
Affiliation(s)
- Kevin R Moon
- Department of Mathematics and Statistics, Utah State University, Logan, UT, USA
| | - David van Dijk
- Cardiovascular Research Center, section Cardiology, Department of Internal Medicine, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
| | - Zheng Wang
- School of Basic Medicine, Qingdao University, Qingdao, China
- Yale Stem Cell Center, Department of Genetics, Yale University, New Haven, CT, USA
| | - Scott Gigante
- Computational Biology and Bioinformatics Program, Yale University, New Haven, CT, USA
| | | | - William S Chen
- Department of Genetics, Yale University, New Haven, CT, USA
| | - Kristina Yim
- Department of Genetics, Yale University, New Haven, CT, USA
| | | | - Matthew J Hirn
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Mathematics, Michigan State University, East Lansing, MI, USA
| | - Ronald R Coifman
- Applied Mathematics Program, Yale University, New Haven, CT, USA
| | - Natalia B Ivanova
- Department of Genetics, Center for Molecular Medicine, University of Georgia, Athens, GA, USA.
| | - Guy Wolf
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Quebec, Canada.
- Mila-Quebec Artificial Intelligence Institute, Montréal, Quebec, Canada.
| | - Smita Krishnaswamy
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Department of Genetics, Yale University, New Haven, CT, USA.
| |
Collapse
|
9
|
Jiao YH, Liu M, Wang G, Li HY, Liu JS, Yang X, Yang WD. EMT is the major target for okadaic acid-suppressed the development of neural crest cells in chick embryo. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 180:192-201. [PMID: 31085430 DOI: 10.1016/j.ecoenv.2019.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 06/09/2023]
Abstract
As a main marine phycotoxin, okadaic acid (OA) is mainly responsible for diarrheic shellfish poisoning (DSP), through specifically inhibiting phosphatase (PP1 and PP2A). It has been shown that isotope labelled-OA could cross the placental barrier in mice. However, it remains obscure how OA exposure could affect the formation of neural crest cells (NCCs), especially cranial NCCs in early embryo development. Here, we explored the effects of OA exposure on the generation of neural crest cells during embryonic development using the classic chick embryo model. We found that OA exposure at 100 nM (80.5 μg/L) could cause craniofacial bone defects in the developing chick embryo and delay the development of early chick embryos. Immunofluorescent staining of HNK-1, Pax7, and Ap-2α demonstrated that cranial NCC generation was inhibited by OA exposure. Double immunofluorescent staining with Ap-2α/PHIS3 or Pax7/c-Caspase3 manifested that both NCC proliferation and apoptosis were restrained by OA exposure. Furthermore, the expression of Msx1 and BMP4 were down-regulated in the developing chick embryonic neural tubes, which could contribute the inhibitive production of NCCs. We also discovered that expression of EMT-related adhesion molecules, such as Cadherin 6B (Cad6B) and E-cadherin, was altered following OA exposure. In sum, OA exposure negatively affected the development of embryonic neural crest cells, which in turn might result in cranial bone malformation.
Collapse
Affiliation(s)
- Yu-Hu Jiao
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Meng Liu
- Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, 510632, China
| | - Guang Wang
- Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, 510632, China
| | - Hong-Ye Li
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jie-Sheng Liu
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xuesong Yang
- Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, 510632, China.
| | - Wei-Dong Yang
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Williams AL, Bohnsack BL. What's retinoic acid got to do with it? Retinoic acid regulation of the neural crest in craniofacial and ocular development. Genesis 2019; 57:e23308. [PMID: 31157952 DOI: 10.1002/dvg.23308] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/23/2019] [Accepted: 05/05/2019] [Indexed: 12/21/2022]
Abstract
Retinoic acid (RA), the active derivative of vitamin A (retinol), is an essential morphogen signaling molecule and major regulator of embryonic development. The dysregulation of RA levels during embryogenesis has been associated with numerous congenital anomalies, including craniofacial, auditory, and ocular defects. These anomalies result from disruptions in the cranial neural crest, a vertebrate-specific transient population of stem cells that contribute to the formation of diverse cell lineages and embryonic structures during development. In this review, we summarize our current knowledge of the RA-mediated regulation of cranial neural crest induction at the edge of the neural tube and the migration of these cells into the craniofacial region. Further, we discuss the role of RA in the regulation of cranial neural crest cells found within the frontonasal process, periocular mesenchyme, and pharyngeal arches, which eventually form the bones and connective tissues of the head and neck and contribute to structures in the anterior segment of the eye. We then review our understanding of the mechanisms underlying congenital craniofacial and ocular diseases caused by either the genetic or toxic disruption of RA signaling. Finally, we discuss the role of RA in maintaining neural crest-derived structures in postembryonic tissues and the implications of these studies in creating new treatments for degenerative craniofacial and ocular diseases.
Collapse
Affiliation(s)
- Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
11
|
Chopra K, Ishibashi S, Amaya E. Zebrafish duox mutations provide a model for human congenital hypothyroidism. Biol Open 2019; 8:bio.037655. [PMID: 30700401 PMCID: PMC6398463 DOI: 10.1242/bio.037655] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Thyroid dyshormonogenesis is a leading cause of congenital hypothyroidism, a highly prevalent but treatable condition. Thyroid hormone (TH) synthesis is dependent on the formation of reactive oxygen species (ROS). In humans, the primary sources for ROS production during thyroid hormone synthesis are the NADPH oxidases DUOX1 and DUOX2. Indeed, mutations in DUOX1 and DUOX2 have been linked with congenital hypothyroidism. Unlike humans, zebrafish has a single orthologue for DUOX1 and DUOX2. In this study, we investigated the phenotypes associated with two nonsense mutant alleles, sa9892 and sa13017, of the single duox gene in zebrafish. Both alleles gave rise to readily observable phenotypes reminiscent of congenital hypothyroidism, from the larval stages through to adulthood. By using various methods to examine external and internal phenotypes, we discovered a strong correlation between TH synthesis and duox function, beginning from an early larval stage, when T4 levels are already noticeably absent in the mutants. Loss of T4 production resulted in growth retardation, pigmentation defects, ragged fins, thyroid hyperplasia/external goiter and infertility. Remarkably, all of these defects associated with chronic congenital hypothyroidism could be rescued with T4 treatment, even when initiated when the fish had already reached adulthood. Our work suggests that these zebrafish duox mutants may provide a powerful model to understand the aetiology of untreated and treated congenital hypothyroidism even in advanced stages of development. This article has an associated First Person interview with the first author of the paper. Summary: Zebrafish harbouring two loss-of-function alleles of the single duox gene exhibit various adult phenotypes reminiscent of human congenital hypothyroidism.
Collapse
Affiliation(s)
- Kunal Chopra
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Shoko Ishibashi
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Enrique Amaya
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
12
|
Defining a critical period in calvarial development for Hedgehog pathway antagonist-induced frontal bone dysplasia in mice. Int J Oral Sci 2019; 11:3. [PMID: 30783111 PMCID: PMC6381108 DOI: 10.1038/s41368-018-0040-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/09/2018] [Accepted: 09/27/2018] [Indexed: 12/19/2022] Open
Abstract
The Hedgehog (Hh) signalling pathway is essential for cellular proliferation and differentiation during embryonic development. Gain and loss of function of Hh signalling are known to result in an array of craniofacial malformations. To determine the critical period for Hh pathway antagonist-induced frontal bone hypoplasia, we examined patterns of dysmorphology caused by Hh signalling inhibition. Pregnant mice received a single oral administration of Hh signalling inhibitor GDC-0449 at 100 mg•kg−1 or 150 mg•kg−1 body weight at preselected time points between embryonic days (E)8.5 and 12.5. The optimal teratogenic concentration of GDC-0449 was determined to be 150 mg•kg−1. Exposure between E9.5 and E10.5 induced frontal bone dysplasia, micrognathia and limb defects, with administration at E10.5 producing the most pronounced effects. This model showed decreased ossification of the frontal bone with downregulation of Hh signalling. The osteoid thickness of the frontal bone was significantly reduced. The amount of neural crest-derived frontal bone primordium was reduced after GDC-0449 exposure owing to a decreased rate of cell proliferation and increased cell death. During embryonic development, the Hedgehog signalling pathway regulates the migration, proliferation and differentiation of cranial neural crest cells in the early frontal bone. The Hedgehog signalling pathway transmits information to embryonic cells for their proper cell differentiation, and increased or reduced function of that signalling results in various craniofacial malformations. A team headed by Weihui Chen at Fujian Medical University in China investigated the patterns of abnormalities caused by inhibition of Hedgehog signalling in pregnant mice at preselected embryonic time points. The team was able to identify the critical period for sensitivity to GDC-0449, a potent Hedgehog signalling inhibitor. The authors believe that their mouse model can be effective in further investigating the mechanisms of craniofacial malformations and will have a profound impact on identifying candidate human disease genes and associated environmental factors.
Collapse
|
13
|
Davidoff MS. The Pluripotent Microvascular Pericytes Are the Adult Stem Cells Even in the Testis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1122:235-267. [PMID: 30937872 DOI: 10.1007/978-3-030-11093-2_13] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pericytes of the testis are part of the omnipresent population of pericytes in the vertebrate body and are the only true pluripotent adult stem cells able to produce structures typical for the tree primitive germ layers: ectoderm, mesoderm, and endoderm. They originate very early in the embryogenesis from the pluripotent epiblast. The pericytes become disseminated through the whole vertebrate organism by the growing and differentiating blood vessels where they remain in specialized periendothelial vascular niches as resting pluripotent adult stem cells for tissue generation, maintenance, repair, and regeneration. The pericytes are also the ancestors of the perivascular multipotent stromal cells (MSCs). The variable appearance of the pericytes and their progeny reflects the plasticity under the influence of their own epigenetic and the local environmental factors of the host organ. In the testis the pericytes are the ancestors of the neuroendocrine Leydig cells. After activation the pericytes start to proliferate, migrate, and build transit-amplifying cells that transdifferentiate into multipotent stromal cells. These represent progenitors for a number of different cell types in an organ. Finally, it becomes evident that the pericytes are a brilliant achievement of the biological nature aiming to supply every organ with an omnipresent population of pluripotent adult stem cells. Their fascinating features are prerequisites for future therapy concepts supporting cell systems of organs.
Collapse
Affiliation(s)
- Michail S Davidoff
- University Medical Center Hamburg-Eppendorf, Hamburg Museum of Medical History, Hamburg, Germany.
| |
Collapse
|
14
|
Méndez-Maldonado K, Vega-López G, Caballero-Chacón S, Aybar MJ, Velasco I. Activation of Hes1 and Msx1 in Transgenic Mouse Embryonic Stem Cells Increases Differentiation into Neural Crest Derivatives. Int J Mol Sci 2018; 19:E4025. [PMID: 30551562 PMCID: PMC6321090 DOI: 10.3390/ijms19124025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/28/2018] [Accepted: 12/01/2018] [Indexed: 12/11/2022] Open
Abstract
The neural crest (NC) comprises a multipotent cell population that produces peripheral neurons, cartilage, and smooth muscle cells, among other phenotypes. The participation of Hes1 and Msx1 when expressed in mouse embryonic stem cells (mESCs) undergoing NC differentiation is unexplored. In this work, we generated stable mESCs transfected with constructs encoding chimeric proteins in which the ligand binding domain of glucocorticoid receptor (GR), which is translocated to the nucleus by dexamethasone addition, is fused to either Hes1 (HGR) or Msx1 (MGR), as well as double-transgenic cells (HGR+MGR). These lines continued to express pluripotency markers. Upon NC differentiation, all lines exhibited significantly decreased Sox2 expression and upregulated Sox9, Snai1, and Msx1 expression, indicating NC commitment. Dexamethasone was added to induce nuclear translocation of the chimeric proteins. We found that Collagen IIa transcripts were increased in MGR cells, whereas coactivation of HGR+MGR caused a significant increase in Smooth muscle actin (α-Sma) transcripts. Immunostaining showed that activation in HGR+MGR cells induced higher proportions of β-TUBULIN III⁺, α-SMA⁺ and COL2A1⁺ cells. These findings indicate that nuclear translocation of MSX-1, alone or in combination with HES-1, produce chondrocyte-like cells, and simultaneous activation of HES-1 and MSX-1 increases the generation of smooth muscle and neuronal cells.
Collapse
Affiliation(s)
- Karla Méndez-Maldonado
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, México.
- Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM en el Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Ciudad de México 14269, México.
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México; Ciudad Universitaria, Ciudad de México 04510, México.
| | - Guillermo Vega-López
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), San Miguel de Tucumán T4000ILI, Argentina.
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán T4000ILI, Argentina.
| | - Sara Caballero-Chacón
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, México.
| | - Manuel J Aybar
- Instituto Superior de Investigaciones Biológicas (INSIBIO, CONICET-UNT), San Miguel de Tucumán T4000ILI, Argentina.
- Instituto de Biología "Dr. Francisco D. Barbieri", Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán T4000ILI, Argentina.
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, México.
- Laboratorio de Reprogramación Celular del Instituto de Fisiología Celular, UNAM en el Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Ciudad de México 14269, México.
| |
Collapse
|
15
|
Role of FGF signalling in neural crest cell migration during early chick embryo development. ZYGOTE 2018; 26:457-464. [DOI: 10.1017/s096719941800045x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryFibroblast growth factor (FGF) signalling acts as one of modulators that control neural crest cell (NCC) migration, but how this is achieved is still unclear. In this study, we investigated the effects of FGF signalling on NCC migration by blocking this process. Constructs that were capable of inducing Sprouty2 (Spry2) or dominant-negative FGFR1 (Dn-FGFR1) expression were transfected into the cells making up the neural tubes. Our results revealed that blocking FGF signalling at stage HH10 (neurulation stage) could enhance NCC migration at both the cranial and trunk levels in the developing embryos. It was established that FGF-mediated NCC migration was not due to altering the expression of N-cadherin in the neural tube. Instead, we determined that cyclin D1 was overexpressed in the cranial and trunk levels when Sprouty2 was upregulated in the dorsal neural tube. These results imply that the cell cycle was a target of FGF signalling through which it regulates NCC migration at the neurulation stage.
Collapse
|
16
|
Armas P, Calcaterra NB. G-quadruplex in animal development: Contribution to gene expression and genomic heterogeneity. Mech Dev 2018; 154:64-72. [DOI: 10.1016/j.mod.2018.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/18/2018] [Accepted: 05/09/2018] [Indexed: 12/21/2022]
|
17
|
Li Y, Zhang XT, Wang XY, Wang G, Chuai M, Münsterberg A, Yang X. Robo signaling regulates the production of cranial neural crest cells. Exp Cell Res 2017; 361:73-84. [PMID: 28987541 DOI: 10.1016/j.yexcr.2017.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/08/2017] [Accepted: 10/03/2017] [Indexed: 11/27/2022]
Abstract
Slit/Robo signaling plays an important role in the guidance of developing neurons in developing embryos. However, it remains obscure whether and how Slit/Robo signaling is involved in the production of cranial neural crest cells. In this study, we examined Robo1 deficient mice to reveal developmental defects of mouse cranial frontal and parietal bones, which are derivatives of cranial neural crest cells. Therefore, we determined the production of HNK1+ cranial neural crest cells in early chick embryo development after knock-down (KD) of Robo1 expression. Detection of markers for pre-migratory and migratory neural crest cells, PAX7 and AP-2α, showed that production of both was affected by Robo1 KD. In addition, we found that the transcription factor slug is responsible for the aberrant delamination/EMT of cranial neural crest cells induced by Robo1 KD, which also led to elevated expression of E- and N-Cadherin. N-Cadherin expression was enhanced when blocking FGF signaling with dominant-negative FGFR1 in half of the neural tube. Taken together, we show that Slit/Robo signaling influences the delamination/EMT of cranial neural crest cells, which is required for cranial bone development.
Collapse
Affiliation(s)
- Yan Li
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China; The key Laboratory of Assisted Circulation, Ministry of Health, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Tan Zhang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Xiao-Yu Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee DD1 5EH, UK
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Xuesong Yang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
18
|
Zhang P, Wang G, Lin Z, Wu Y, Zhang J, Liu M, Lee KKH, Chuai M, Yang X. Alcohol exposure induces chick craniofacial bone defects by negatively affecting cranial neural crest development. Toxicol Lett 2017; 281:53-64. [PMID: 28919490 DOI: 10.1016/j.toxlet.2017.09.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 10/25/2022]
Abstract
Excess alcohol consumption during pregnancy could lead to fetal alcohol syndrome (FAS). However, the molecular mechanism leading to craniofacial abnormality, a feature of FAS, is still poorly understood. The cranial neural crest cells (NCCs) contribute to the formation of the craniofacial bones. Therefore, NCCs exposed to ethanol was investigated - using chick embryos and in vitro explant culture as experimental models. We demonstrated that exposure to 2% ethanol induced craniofacial defects, which includes parietal defect, in the developing chick fetus. Immunofluorescent staining revealed that ethanol treatment downregulated Ap-2ɑ, Pax7 and HNK-1 expressions by cranial NCCs. Using double-immunofluorescent stainings for Ap-2ɑ/pHIS3 and Ap-2ɑ/c-Caspase3, we showed that ethanol treatment inhibited cranial NCC proliferation and increased NCC apoptosis, respectively. Moreover, ethanol treatment of the dorsal neuroepithelium increased Laminin, N-Cadherin and Cadherin 6B expressions while Cadherin 7 expression was repressed. In situ hybridization also revealed that ethanol treatment up-regulated Cadherin 6B expression but down-regulated slug, Msx1, FoxD3 and BMP4 expressions. In summary, our experimental results demonstrated that ethanol treatment interferes with the production of cranial NCCs by affecting the proliferation and apoptosis of these cells. In addition, ethanol affected the delamination, epithelial-mesenchymal transition (EMT) and cell migration of cranial NCCs, which may have contributed to the etiology of the craniofacial defects.
Collapse
Affiliation(s)
- Ping Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Guang Wang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Zhuangling Lin
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Yushi Wu
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Jing Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Meng Liu
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China
| | - Kenneth Ka Ho Lee
- Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Biomedical Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Manli Chuai
- Division of Cell and Developmental Biology, University of Dundee, Dundee, DD1 5EH, UK
| | - Xuesong Yang
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Division of Histology & Embryology, Medical College, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
19
|
Williams AL, Bohnsack BL. Multi-Photon Time Lapse Imaging to Visualize Development in Real-time: Visualization of Migrating Neural Crest Cells in Zebrafish Embryos. J Vis Exp 2017. [PMID: 28829423 DOI: 10.3791/56214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Congenital eye and craniofacial anomalies reflect disruptions in the neural crest, a transient population of migratory stem cells that give rise to numerous cell types throughout the body. Understanding the biology of the neural crest has been limited, reflecting a lack of genetically tractable models that can be studied in vivo and in real-time. Zebrafish is a particularly important developmental model for studying migratory cell populations, such as the neural crest. To examine neural crest migration into the developing eye, a combination of the advanced optical techniques of laser scanning microscopy with long wavelength multi-photon fluorescence excitation was implemented to capture high-resolution, three-dimensional, real-time videos of the developing eye in transgenic zebrafish embryos, namely Tg(sox10:EGFP) and Tg(foxd3:GFP), as sox10 and foxd3 have been shown in numerous animal models to regulate early neural crest differentiation and likely represent markers for neural crest cells. Multi-photon time-lapse imaging was used to discern the behavior and migratory patterns of two neural crest cell populations contributing to early eye development. This protocol provides information for generating time-lapse videos during zebrafish neural crest migration, as an example, and can be further applied to visualize the early development of many structures in the zebrafish and other model organisms.
Collapse
Affiliation(s)
- Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan;
| |
Collapse
|
20
|
Zhang S, Su Y, Gao J, Zhang C, Tanaka H. A potential inhibitory function of draxin in regulating mouse trunk neural crest migration. In Vitro Cell Dev Biol Anim 2016; 53:43-53. [PMID: 27649978 PMCID: PMC5258808 DOI: 10.1007/s11626-016-0079-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/25/2016] [Indexed: 11/28/2022]
Abstract
Draxin is a repulsive axon guidance protein that plays important roles in the formation of three commissures in the central nervous system and dorsal interneuron 3 (dI3) in the chick spinal cord. In the present study, we report the expression pattern of mouse draxin in the embryonic mouse trunk spinal cord. In the presence of draxin, the longest net migration length of a migrating mouse trunk neural crest cell was significantly reduced. In addition, the relative number of apolar neural crest cells increased as the draxin treatment time increased. Draxin caused actin cytoskeleton rearrangement in the migrating trunk neural crest cells. Our data suggest that draxin may regulate mouse trunk neural crest cell migration by the rearrangement of cell actin cytoskeleton and by reducing the polarization activity of these cells subsequently.
Collapse
Affiliation(s)
- Sanbing Zhang
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China.
| | - Yuhong Su
- Department of Human Anatomy, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, Hebei Province, 050017, People's Republic of China.
| | - Jinbao Gao
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China
| | - Chenbing Zhang
- Department of Hand and Foot Surgery, The Third Hospital of Shijiazhuang City, 15 South Tiyu Street, Shijiazhuang, 050000, People's Republic of China
| | - Hideaki Tanaka
- Division of Developmental Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
21
|
Lin SC, Gou GH, Hsia CW, Ho CW, Huang KL, Wu YF, Lee SY, Chen YH. Simulated Microgravity Disrupts Cytoskeleton Organization and Increases Apoptosis of Rat Neural Crest Stem Cells Via Upregulating CXCR4 Expression and RhoA-ROCK1-p38 MAPK-p53 Signaling. Stem Cells Dev 2016; 25:1172-93. [PMID: 27269634 DOI: 10.1089/scd.2016.0040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Neural crest stem cells (NCSCs) are a population of multipotent stem cells that are distributed broadly in many tissues and organs and are capable of differentiating into a variety of cell types that are dispersed throughout three germ layers. We are interested in studying the effects of simulated microgravity on the survival and self-renewal of NCSCs. NCSCs extracted from the hair follicle bulge region of the rat whisker pad were cultured in vitro, respectively, in a 2D adherent environment and a 3D suspension environment using the rotatory cell culture system (RCCS) to simulate microgravity. We found that rat NCSCs (rNCSCs) cultured in the RCCS for 24 h showed disrupted organization of filamentous actin, increased globular actin level, formation of plasma membrane blebbing and neurite-like artifact, as well as decreased levels of cortactin and vimentin. Interestingly, ∼70% of RCCS-cultured rNCSCs co-expressed cleaved (active) caspase-3 and neuronal markers microtubule-associated protein 2 (MAP2) and Tuj1 instead of NCSC markers, suggesting stress-induced formation of neurite-like artifact in rNCSCs. In addition, rNCSCs showed increased C-X-C chemokine receptor 4 (CXCR4) expression, RhoA GTPase activation, Rho-associated kinase 1 (ROCK1) and p38 mitogen-activated protein kinase (MAPK) phosphorylation, and p53 expression in the nucleus. Incubation of rNCSCs with the Gα protein inhibitor pertussis toxin or CXCR4 siRNA during RCCS-culturing prevented cytoskeleton disorganization and plasma membrane blebbing, and it suppressed apoptosis of rNCSCs. Taken together, we demonstrate for the first time that simulated microgravity disrupts cytoskeleton organization and increases apoptosis of rNCSCs via upregulating CXCR4 expression and the RhoA-ROCK1-p38 MAPK-p53 signaling pathway.
Collapse
Affiliation(s)
- Shing-Chen Lin
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Guo-Hau Gou
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Ching-Wu Hsia
- 2 Graduate Institute of Medical Sciences, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Cheng-Wen Ho
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,3 Division of Rehabilitation Medicine, Taoyuan Armed Forces General Hospital , Longtan Township, Taoyuan County, Taiwan
| | - Kun-Lun Huang
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan .,4 Department of Undersea and Hyperbaric Medicine, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Yung-Fu Wu
- 5 Department of Medical Research, Tri-Service General Hospital , Neihu District, Taipei City, Taiwan
| | - Shih-Yu Lee
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| | - Yi-Hui Chen
- 1 Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center , Neihu District, Taipei City, Taiwan
| |
Collapse
|
22
|
Chawla B, Schley E, Williams AL, Bohnsack BL. Retinoic Acid and Pitx2 Regulate Early Neural Crest Survival and Migration in Craniofacial and Ocular Development. ACTA ACUST UNITED AC 2016; 107:126-35. [PMID: 27175943 DOI: 10.1002/bdrb.21177] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/21/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Bahaar Chawla
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Elisa Schley
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Antionette L Williams
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| | - Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
23
|
Boddupally K, Wang G, Chen Y, Kobielak A. Lgr5 Marks Neural Crest Derived Multipotent Oral Stromal Stem Cells. Stem Cells 2016; 34:720-31. [PMID: 26865184 DOI: 10.1002/stem.2314] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 10/09/2015] [Accepted: 11/04/2015] [Indexed: 12/22/2022]
Abstract
It has been suggested that multipotent stem cells with neural crest (NC) origin persist into adulthood in oral mucosa. However their exact localization and role in normal homeostasis is unknown. In this study, we discovered that Lgr5 is expressed in NC cells during embryonic development, which give rise to the dormant stem cells in the adult tongue and oral mucosa. Those Lgr5 positive oral stromal stem cells display properties of NC stem cells including clonal growth and multipotent differentiation. RNA sequencing revealed that adult Lgr5+ oral stromal stem cells express high number of neural crest related markers like Sox9, Twist1, Snai1, Myc, Ets1, Crabp1, Epha2, and Itgb1. Using lineage-tracing experiments, we show that these cells persist more than a year in the ventral tongue and some areas of the oral mucosa and give rise to stromal progeny. In vivo transplantation demonstrated that these cells reconstitute the stroma. Our studies show for the first time that Lgr5 is expressed in the NC cells at embryonic day 9.5 (E9.5) and is maintained during embryonic development and postnataly in the stroma of the ventral tongue, and some areas of the oral mucosa and that Lgr5+ cells participate in the maintenance of the stroma.
Collapse
Affiliation(s)
- Keerthi Boddupally
- Department of Otolaryngology, Head & Neck Surgery, University of Southern California, Los Angeles, California, USA.,Department of Biochemistry and Molecular Biology, Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Guangfang Wang
- Department of Otolaryngology, Head & Neck Surgery, University of Southern California, Los Angeles, California, USA.,Department of Biochemistry and Molecular Biology, Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- Norris Medical Library, University of Southern California, Los Angeles, California, USA
| | - Agnieszka Kobielak
- Department of Otolaryngology, Head & Neck Surgery, University of Southern California, Los Angeles, California, USA.,Department of Biochemistry and Molecular Biology, Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
24
|
Rollo BN, Zhang D, Stamp LA, Menheniott TR, Stathopoulos L, Denham M, Dottori M, King SK, Hutson JM, Newgreen DF. Enteric Neural Cells From Hirschsprung Disease Patients Form Ganglia in Autologous Aneuronal Colon. Cell Mol Gastroenterol Hepatol 2015; 2:92-109. [PMID: 28174705 PMCID: PMC4980742 DOI: 10.1016/j.jcmgh.2015.09.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 09/17/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Hirschsprung disease (HSCR) is caused by failure of cells derived from the neural crest (NC) to colonize the distal bowel in early embryogenesis, resulting in absence of the enteric nervous system (ENS) and failure of intestinal transit postnatally. Treatment is by distal bowel resection, but neural cell replacement may be an alternative. We tested whether aneuronal (aganglionic) colon tissue from patients may be colonized by autologous ENS-derived cells. METHODS Cells were obtained and cryopreserved from 31 HSCR patients from the proximal resection margin of colon, and ENS cells were isolated using flow cytometry for the NC marker p75 (nine patients). Aneuronal colon tissue was obtained from the distal resection margin (23 patients). ENS cells were assessed for NC markers immunohistologically and by quantitative reverse-transcription polymerase chain reaction, and mitosis was detected by ethynyl-2'-deoxyuridine labeling. The ability of human HSCR postnatal ENS-derived cells to colonize the embryonic intestine was demonstrated by organ coculture with avian embryo gut, and the ability of human postnatal HSCR aneuronal colon muscle to support ENS formation was tested by organ coculture with embryonic mouse ENS cells. Finally, the ability of HSCR patient ENS cells to colonize autologous aneuronal colon muscle tissue was assessed. RESULTS ENS-derived p75-sorted cells from patients expressed multiple NC progenitor and differentiation markers and proliferated in culture under conditions simulating Wnt signaling. In organ culture, patient ENS cells migrated appropriately in aneural quail embryo gut, and mouse embryo ENS cells rapidly spread, differentiated, and extended axons in patient aneuronal colon muscle tissue. Postnatal ENS cells derived from HSCR patients colonized autologous aneuronal colon tissue in cocultures, proliferating and differentiating as neurons and glia. CONCLUSIONS NC-lineage cells can be obtained from HSCR patient colon and can form ENS-like structures in aneuronal colonic muscle from the same patient.
Collapse
Key Words
- Aganglionosis
- CHIR-99021, 6-[2-[[4-(2,4-dichlorophenyl)-5-(5-methyl-1H-imidazol-2-yl)pyrimidin-2-yl]amino]ethylamino]pyridine-3-carbonitrile
- Cell Therapy
- ENC, enteric neural crest
- ENS, enteric nervous system
- EdU, ethynyl-2′-deoxyuridine
- Enteric Nervous System
- FBS, fetal bovine serum
- GFAP, glial fibrillary acidic protein
- GSK3, glycogen synthase kinase 3
- HNK1, human natural killer-1
- HSCR, Hirschsprung disease
- Hirschsprung Disease
- MTR, MitoTracker Red
- Megacolon
- NC, neural crest
- PBS, phosphate-buffered saline
- PFA, paraformaldehyde
- RCH, Royal Children’s Hospital
- SMA, smooth muscle actin
- SOX10, sex-determining region Y–box 10
- TUJ1, neuron-specific class III β-tubulin
- eGFP, enhanced green fluorescent protein
- nNOS, neuronal nitric oxide synthase
- nTCM, neural tissue culture medium
- qRT-PCR, quantitative reverse transcription and polymerase chain reaction
Collapse
Affiliation(s)
- Benjamin N. Rollo
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia,Correspondence Address correspondence to: Benjamin N. Rollo, PhD, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Flemington Road, Parkville, Victoria 3052, Australia. fax: +61-3-9348-1391.Murdoch Children’s Research InstituteThe Royal Children’s HospitalFlemington RoadParkvilleVictoria 3052Australia
| | - Dongcheng Zhang
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Lincon A. Stamp
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Trevelyan R. Menheniott
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Lefteris Stathopoulos
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Mark Denham
- Stem Cell Laboratory, Department of Biomedicine, Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark
| | - Mirella Dottori
- Centre for Neural Engineering, NICTA, University of Melbourne, Australia
| | - Sebastian K. King
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia,Royal Children’s Hospital, Parkville, Victoria, Australia
| | - John M. Hutson
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia,Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Donald F. Newgreen
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
| |
Collapse
|
25
|
Tumor suppression function of FoxD3 in lung cancer. Ir J Med Sci 2015; 185:547-553. [PMID: 25894280 DOI: 10.1007/s11845-015-1297-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/06/2015] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The transcription factor forkhead box D3 (FoxD3) is a forkhead protein and serves as a transcriptional repressor. The functions of FoxD3 in embryogenesis and in the development of neural crest cells have been diversely studied. Its role associated with tumor development has attracted attention just in recent years. However, little is known about its regulation and functions in the tumorigenesis and progression of lung cancer. MATERIALS AND METHODS We did a pioneer study to identify the function of FoxD3 in lung cancer by knocking down FoxD3 expression in lung adenocarcinoma cell lines. RESULTS The result showed that silencing FoxD3 in lung cancer cell lines stimulated cell growth and inhibited cell apoptosis. Through quantified PCR analysis, we observed marked gene expression increase of insulin-like growth factor receptor 1 and BRAF under the condition of FoxD3 silencing. In contrast, cell cycle negative regulator P21 expression decreased after FoxD3 silencing. CONCLUSIONS Our study is the first to identify the tumor suppressor function of FoxD3 in lung cancer.
Collapse
|
26
|
Vermillion KL, Lidberg KA, Gammill LS. Expression of actin-binding proteins and requirement for actin-depolymerizing factor in chick neural crest cells. Dev Dyn 2014; 243:730-8. [PMID: 24868596 DOI: 10.1002/dvdy.24105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Neural crest cells are multipotent cells that migrate extensively throughout vertebrate embryos to form diverse lineages. Cell migration requires polarized, organized actin networks that provide the driving force for motility. Actin-binding proteins that regulate neural crest cell migration are just beginning to be defined. RESULTS We recently identified a number of actin-associated factors through proteomic profiling of methylated proteins in migratory neural crest cells. Here, we report the previously undocumented expression pattern of three of these proteins in chick early neural crest development: doublecortin (DCX), tropomyosin-1 (TPM-1), and actin depolymerizing factor (ADF). All three genes are expressed with varying degrees of specificity and intensity in premigratory and migratory neural crest cells, and their resulting proteins exhibit distinct subcellular localization in migratory neural crest cells. Morpholino knock down of ADF reveals it is required for Sox10 gene expression, but minimally important during neural crest migration. CONCLUSIONS Neural crest cells express DCX, TPM-1, and ADF. ADF is necessary during neural crest specification, but largely dispensable for migration.
Collapse
|
27
|
Epigenetic regulation in neural crest development. Dev Biol 2014; 396:159-68. [PMID: 25446277 DOI: 10.1016/j.ydbio.2014.09.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 09/17/2014] [Accepted: 09/25/2014] [Indexed: 12/22/2022]
Abstract
The neural crest is a migratory and multipotent cell population that plays a crucial role in many aspects of embryonic development. In all vertebrate embryos, these cells emerge from the dorsal neural tube then migrate long distances to different regions of the body, where they contribute to formation of many cell types and structures. These include much of the peripheral nervous system, craniofacial skeleton, smooth muscle, and pigmentation of the skin. The best-studied regulatory events guiding neural crest development are mediated by transcription factors and signaling molecules. In recent years, however, growing evidence supports an important role for epigenetic regulation as an additional mechanism for controlling the timing and level of gene expression at different stages of neural crest development. Here, we summarize the process of neural crest formation, with focus on the role of epigenetic regulation in neural crest specification, migration, and differentiation as well as in neural crest related birth defects and diseases.
Collapse
|
28
|
FoxD3 deficiency promotes breast cancer progression by induction of epithelial-mesenchymal transition. Biochem Biophys Res Commun 2014; 446:580-4. [PMID: 24632201 DOI: 10.1016/j.bbrc.2014.03.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/04/2014] [Indexed: 12/11/2022]
Abstract
The transcription factor forkhead box D3 (FOXD3) plays an important role in the development of neural crest and gastric cancer cells. However, the function and mechanisms of FOXD3 in the breast tumorigenesis and progression is still limited. Here, we report that FOXD3 is a tumor suppressor of breast cancer tumorigenicity and aggressiveness. We found that FOXD3 is down-regulated in breast cancer tissues. Patients with low FOXD3 expression have a poor outcome. Depletion of FOXD3 expression promotes breast cancer cell proliferation and invasion in vitro, whereas overexpression of FOXD3 inhibits breast cancer cell proliferation and invasion both in vitro and in vivo. In addition, depletion of FOXD3 is linked to epithelial-mesenchymal transition (EMT)-like phenotype. Our results indicate FOXD3 exhibits tumor suppressive activity and may be useful for breast therapy.
Collapse
|
29
|
Neurotrophin signalling and transcription programmes interactions in the development of somatosensory neurons. Handb Exp Pharmacol 2014; 220:329-53. [PMID: 24668479 DOI: 10.1007/978-3-642-45106-5_13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Somatosensory neurons of the dorsal root ganglia are generated from multipotent neural crest cells by a process of progressive specification and differentiation. Intrinsic transcription programmes active in somatosensory neuron progenitors and early post-mitotic neurons drive the cell-type expression of neurotrophin receptors. In turn, signalling by members of the neurotrophin family controls expression of transcription factors that regulate neuronal sub-type specification. This chapter explores the mechanisms by which this crosstalk between neurotrophin signalling and transcription programmes generates the diverse functional sub-types of somatosensory neurons found in the mature animal.
Collapse
|
30
|
Cebra-Thomas JA, Terrell A, Branyan K, Shah S, Rice R, Gyi L, Yin M, Hu Y, Mangat G, Simonet J, Betters E, Gilbert SF. Late-emigrating trunk neural crest cells in turtle embryos generate an osteogenic ectomesenchyme in the plastron. Dev Dyn 2013; 242:1223-35. [PMID: 23904174 DOI: 10.1002/dvdy.24018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 06/13/2013] [Accepted: 07/03/2013] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The turtle plastron is composed of a keratinized epidermis overlying nine dermal bones. Its developmental origin has been controversial; recent evidence suggests that the plastral bones derive from trunk neural crest cells (NCCs). RESULTS This study extends the observations that there is a turtle-specific, second wave of trunk NCC delamination and migration, after the original NCCs have reached their destination and differentiated. This second wave was confirmed by immunohistochemistry in whole-mounts and serial sections, by injecting DiI (1,1', di-octadecyl-3,3,3',3',-tetramethylindo-carbocyanine perchlorate) into the lumen of the neural tube and tracing labeled cells into the plastron, and by isolating neural tubes from older turtle embryos and observing delaminating NCCs. This later migration gives rise to a plastral ectomesenchyme that expresses NCC markers and can be induced to initiate bone formation. CONCLUSIONS The NCCs of this second migration have properties similar to those of the earlier NCCs, but also express markers characteristic of cranial NCCs. The majority of the cells of the plastron mesenchyme express neural crest markers, and have osteogenic differentiation capabilities that are similar or identical to craniofacial ectomesenchyme. Our evidence supports the contention that turtle plastron bones are derived from a late emigrating population of cells derived from the trunk neural crest.
Collapse
|
31
|
Kamel G, Hoyos T, Rochard L, Dougherty M, Kong Y, Tse W, Shubinets V, Grimaldi M, Liao EC. Requirement for frzb and fzd7a in cranial neural crest convergence and extension mechanisms during zebrafish palate and jaw morphogenesis. Dev Biol 2013; 381:423-33. [PMID: 23806211 DOI: 10.1016/j.ydbio.2013.06.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 11/29/2022]
Abstract
Regulation of convergence and extension by wnt-frizzled signaling is a common theme in embryogenesis. This study examines the functional requirements of frzb and fzd7a in convergence and extension mechanisms during craniofacial development. Using a morpholino knockdown approach, we found that frzb and fzd7a are dispensable for directed migration of the bilateral trabeculae, but necessary for the convergence and extension of the palatal elements, where the extension process is mediated by chondrocyte proliferation, morphologic change and intercalation. In contrast, frzb and fzd7a are required for convergence of the mandibular prominences, where knockdown of either frzb or fzd7a resulted in complete loss of lower jaw structures. Further, we found that bapx1 was specifically downregulated in the wnt9a/frzb/fzd7a morphants, while general neural crest markers were unaffected. In addition, expression of wnt9a and frzb was also absent in the edn-/- mutant. Notably, over-expression of bapx1 was sufficient to partially rescue mandibular elements in the wnt9a/frzb/fzd7a morphants, demonstrating genetic epistasis of bapx1 acting downstream of edn1 and wnt9a/frzb/fzd7a in lower jaw development. This study underscores the important role of wnt-frizzled signaling in convergence and extension in palate and craniofacial morphogenesis, distinct regulation of upper vs. lower jaw structures, and integration of wnt-frizzled with endothelin signaling to coordinate shaping of the facial form.
Collapse
Affiliation(s)
- George Kamel
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ignatius MS, Unal Eroglu A, Malireddy S, Gallagher G, Nambiar RM, Henion PD. Distinct functional and temporal requirements for zebrafish Hdac1 during neural crest-derived craniofacial and peripheral neuron development. PLoS One 2013; 8:e63218. [PMID: 23667588 PMCID: PMC3646935 DOI: 10.1371/journal.pone.0063218] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 04/02/2013] [Indexed: 11/19/2022] Open
Abstract
The regulation of gene expression is accomplished by both genetic and epigenetic means and is required for the precise control of the development of the neural crest. In hdac1(b382) mutants, craniofacial cartilage development is defective in two distinct ways. First, fewer hoxb3a, dlx2 and dlx3-expressing posterior branchial arch precursors are specified and many of those that are consequently undergo apoptosis. Second, in contrast, normal numbers of progenitors are present in the anterior mandibular and hyoid arches, but chondrocyte precursors fail to terminally differentiate. In the peripheral nervous system, there is a disruption of enteric, DRG and sympathetic neuron differentiation in hdac1(b382) mutants compared to wildtype embryos. Specifically, enteric and DRG-precursors differentiate into neurons in the anterior gut and trunk respectively, while enteric and DRG neurons are rarely present in the posterior gut and tail. Sympathetic neuron precursors are specified in hdac1(b382) mutants and they undergo generic neuronal differentiation but fail to undergo noradrenergic differentiation. Using the HDAC inhibitor TSA, we isolated enzyme activity and temporal requirements for HDAC function that reproduce hdac1(b382) defects in craniofacial and sympathetic neuron development. Our study reveals distinct functional and temporal requirements for zebrafish hdac1 during neural crest-derived craniofacial and peripheral neuron development.
Collapse
Affiliation(s)
- Myron S. Ignatius
- Molecular, Cellular and Developmental Biology Program, Ohio State University, Columbus, Ohio, United States of America
| | - Arife Unal Eroglu
- Molecular, Cellular and Developmental Biology Program, Ohio State University, Columbus, Ohio, United States of America
| | - Smitha Malireddy
- Department of Neuroscience, Ohio State University, Columbus, Ohio, United States of America
| | - Glen Gallagher
- Department of Neuroscience, Ohio State University, Columbus, Ohio, United States of America
| | - Roopa M. Nambiar
- Molecular, Cellular and Developmental Biology Program, Ohio State University, Columbus, Ohio, United States of America
| | - Paul D. Henion
- Department of Neuroscience, Ohio State University, Columbus, Ohio, United States of America
- Molecular, Cellular and Developmental Biology Program, Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
33
|
Induction of RET dependent and independent pro-inflammatory programs in human peripheral blood mononuclear cells from Hirschsprung patients. PLoS One 2013; 8:e59066. [PMID: 23527089 PMCID: PMC3601093 DOI: 10.1371/journal.pone.0059066] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 02/12/2013] [Indexed: 12/22/2022] Open
Abstract
Hirschsprung disease (HSCR) is a rare congenital anomaly characterized by the absence of enteric ganglia in the distal intestinal tract. While classified as a multigenic disorder, the altered function of the RET tyrosine kinase receptor is responsible for the majority of the pathogenesis of HSCR. Recent evidence demonstrate a strong association between RET and the homeostasis of immune system. Here, we utilize a unique cohort of fifty HSCR patients to fully characterize the expression of RET receptor on both innate (monocytes and Natural Killer lymphocytes) and adaptive (B and T lymphocytes) human peripheral blood mononuclear cells (PBMCs) and to explore the role of RET signaling in the immune system. We show that the increased expression of RET receptor on immune cell subsets from HSCR individuals correlates with the presence of loss-of-function RET mutations. Moreover, we demonstrate that the engagement of RET on PBMCs induces the modulation of several inflammatory genes. In particular, RET stimulation with glial-cell line derived neurotrophic factor family (GDNF) and glycosyl-phosphatidylinositol membrane anchored co-receptor α1 (GFRα1) trigger the up-modulation of genes encoding either for chemokines (CCL20, CCL2, CCL3, CCL4, CCL7, CXCL1) and cytokines (IL-1β, IL-6 and IL-8) and the down-regulation of chemokine/cytokine receptors (CCR2 and IL8-Rα). Although at different levels, the modulation of these “RET-dependent genes” occurs in both healthy donors and HSCR patients. We also describe another set of genes that, independently from RET stimulation, are differently regulated in healthy donors versus HSCR patients. Among these “RET-independent genes”, there are CSF-1R, IL1-R1, IL1-R2 and TGFβ-1, whose levels of transcripts were lower in HSCR patients compared to healthy donors, thus suggesting aberrancies of inflammatory responses at mucosal level. Overall our results demonstrate that immune system actively participates in the physiopathology of HSCR disease by modulating inflammatory programs that are either dependent or independent from RET signaling.
Collapse
|
34
|
Wiszniak S, Lumb R, Kabbara S, Scherer M, Schwarz Q. Li-gazing at the crest: modulation of the neural crest by the ubiquitin pathway. Int J Biochem Cell Biol 2013; 45:1087-91. [PMID: 23458963 DOI: 10.1016/j.biocel.2013.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 02/08/2013] [Accepted: 02/22/2013] [Indexed: 10/27/2022]
Abstract
Neural crest cells are a transient population of stem cells that give rise to a diverse range of cell types during embryonic development. Through gain-of-function and loss-of-function studies in several model organisms many key signalling pathways and cell-type specific transcription factors essential for neural crest cell development have been identified. However, the role of post-translational regulation remains largely unexplored. Here we review this cell type with a foray into the known and potential roles of the ubiquitination pathway in key signalling events during neural crest cell development.
Collapse
Affiliation(s)
- Sophie Wiszniak
- Centre for Cancer Biology, SA Pathology, Adelaide 5000, Australia
| | | | | | | | | |
Collapse
|
35
|
Bohnsack BL, Kahana A. Thyroid hormone and retinoic acid interact to regulate zebrafish craniofacial neural crest development. Dev Biol 2013; 373:300-9. [PMID: 23165295 PMCID: PMC3534885 DOI: 10.1016/j.ydbio.2012.11.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 11/07/2012] [Accepted: 11/08/2012] [Indexed: 01/17/2023]
Abstract
Craniofacial and ocular morphogenesis require proper regulation of cranial neural crest migration, proliferation, survival and differentiation. Although alterations in maternal thyroid hormone (TH) are associated with congenital craniofacial anomalies, the role of TH on the neural crest has not been previously described. Using zebrafish, we demonstrate that pharmacologic and genetic alterations in TH signaling disrupt cranial neural crest migration, proliferation, and survival, leading to craniofacial, extraocular muscle, and ocular developmental abnormalities. In the rostral cranial neural crest that gives rise to the periocular mesenchyme and the frontonasal process, retinoic acid (RA) rescued migratory defects induced by decreased TH signaling. In the caudal cranial neural crest, TH and RA had reciprocal effects on anterior and posterior pharyngeal arch development. The interactions between TH and RA signaling were partially mediated by the retinoid X receptor. We conclude that TH regulates both rostral and caudal cranial neural crest. Further, coordinated interactions of TH and RA are required for proper craniofacial and ocular development.
Collapse
Affiliation(s)
- Brenda L. Bohnsack
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor MI
| | - Alon Kahana
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor MI
| |
Collapse
|
36
|
Chen L, Tweddle DA. p53, SKP2, and DKK3 as MYCN Target Genes and Their Potential Therapeutic Significance. Front Oncol 2012; 2:173. [PMID: 23226679 PMCID: PMC3508619 DOI: 10.3389/fonc.2012.00173] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 11/01/2012] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma is the most common extra-cranial solid tumor of childhood. Despite significant advances, it currently still remains one of the most difficult childhood cancers to cure, with less than 40% of patients with high-risk disease being long-term survivors. MYCN is a proto-oncogene implicated to be directly involved in neuroblastoma development. Amplification of MYCN is associated with rapid tumor progression and poor prognosis. Novel therapeutic strategies which can improve the survival rates whilst reducing the toxicity in these patients are therefore required. Here we discuss genes regulated by MYCN in neuroblastoma, with particular reference to p53, SKP2, and DKK3 and strategies that may be employed to target them.
Collapse
Affiliation(s)
- Lindi Chen
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University Newcastle, UK
| | | |
Collapse
|
37
|
Lee PC, Taylor-Jaffe KM, Nordin KM, Prasad MS, Lander RM, LaBonne C. SUMOylated SoxE factors recruit Grg4 and function as transcriptional repressors in the neural crest. ACTA ACUST UNITED AC 2012; 198:799-813. [PMID: 22927467 PMCID: PMC3432773 DOI: 10.1083/jcb.201204161] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SUMOylation of SoxE alters its recruitment of transcriptional coregulatory factors, displacing the binding of coactivators and promoting the recruitment of the corepressor Grg4. A growing number of transcriptional regulatory proteins are known to be modified by the small ubiquitin-like protein, SUMO. Posttranslational modification by SUMO may be one means by which transcriptional regulatory factors that play context-dependent roles in multiple processes can be regulated such that they direct the appropriate cellular and developmental outcomes. In early vertebrate embryos, SUMOylation of SoxE transcription factors profoundly affects their function, inhibiting their neural crest–inducing activity and promoting ear formation. In this paper, we provide mechanistic insight into how SUMO modification modulates SoxE function. We show that SUMOylation dramatically altered recruitment of transcriptional coregulator factors by SoxE proteins, displacing coactivators CREB-binding protein/p300 while promoting the recruitment of a corepressor, Grg4. These data demonstrate that SoxE proteins can function as transcriptional repressors in a SUMO-dependent manner. They further suggest a novel multivalent mechanism for SUMO-mediated recruitment of transcriptional coregulatory factors.
Collapse
Affiliation(s)
- Pei-Chih Lee
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | | | | | | | | | | |
Collapse
|
38
|
Becker SF, Langhe R, Huang C, Wedlich D, Kashef J. Giving the right tug for migration: Cadherins in tissue movements. Arch Biochem Biophys 2012; 524:30-42. [DOI: 10.1016/j.abb.2012.02.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/16/2012] [Accepted: 02/17/2012] [Indexed: 01/01/2023]
|
39
|
Schmidt J, Piekarski N, Olsson L. Cranial muscles in amphibians: development, novelties and the role of cranial neural crest cells. J Anat 2012; 222:134-46. [PMID: 22780231 DOI: 10.1111/j.1469-7580.2012.01541.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Our research on the evolution of the vertebrate head focuses on understanding the developmental origins of morphological novelties. Using a broad comparative approach in amphibians, and comparisons with the well-studied quail-chicken system, we investigate how evolutionarily conserved or variable different aspects of head development are. Here we review research on the often overlooked development of cranial muscles, and on its dependence on cranial cartilage development. In general, cranial muscle cell migration and the spatiotemporal pattern of cranial muscle formation appears to be very conserved among the few species of vertebrates that have been studied. However, fate-mapping of somites in the Mexican axolotl revealed differences in the specific formation of hypobranchial muscles (tongue muscles) in comparison to the chicken. The proper development of cranial muscles has been shown to be strongly dependent on the mostly neural crest-derived cartilage elements in the larval head of amphibians. For example, a morpholino-based knock-down of the transcription factor FoxN3 in Xenopus laevis has drastic indirect effects on cranial muscle patterning, although the direct function of the gene is mostly connected to neural crest development. Furthermore, extirpation of single migratory streams of cranial neural crest cells in combination with fate-mapping in a frog shows that individual cranial muscles and their neural crest-derived connective tissue attachments originate from the same visceral arch, even when the muscles attach to skeletal components that are derived from a different arch. The same pattern has also been found in the chicken embryo, the only other species that has been thoroughly investigated, and thus might be a conserved pattern in vertebrates that reflects the fundamental nature of a mechanism that keeps the segmental order of the head in place despite drastic changes in adult anatomy. There is a need for detailed comparative fate-mapping of pre-otic paraxial mesoderm in amphibians, to determine developmental causes underlying the complicated changes in cranial muscle development and architecture within amphibians, and in particular how the novel mouth apparatus in frog tadpoles evolved. This will also form a foundation for further research into the molecular mechanisms that regulate rostral head morphogenesis. Our empirical studies are discussed within a theoretical framework concerned with the evolutionary origin and developmental basis of novel anatomical structures in general. We argue that a common developmental origin is not a fool-proof guide to homology, and that a view that sees only structures without homologs as novel is too restricted, because novelties must be produced by changes in the same framework of developmental processes. At the level of developmental processes and mechanisms, novel structures are therefore likely to have homologs, and we need to develop a hierarchical concept of novelty that takes this into account.
Collapse
Affiliation(s)
- Jennifer Schmidt
- Institut für Spezielle Zoologie und Evolutionsbiologie mit Phyletischem Museum, Friedrich-Schiller-Universität Jena, Jena, Germany
| | | | | |
Collapse
|
40
|
Cantilena S, Pastorino F, Pezzolo A, Chayka O, Pistoia V, Ponzoni M, Sala A. Frizzled receptor 6 marks rare, highly tumourigenic stem-like cells in mouse and human neuroblastomas. Oncotarget 2012; 2:976-83. [PMID: 22249030 PMCID: PMC3282103 DOI: 10.18632/oncotarget.410] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Wnt signalling is an important component of vertebrate development, required for specification of the neural crest. Ten Wnt receptors [Frizzled receptor 1-10 (Fzd1-10)] have been identified so far, some of which are expressed in the developing nervous system and the neural crest. Here we show that expression of one such receptors, Fzd6, predicts poor survival in neuroblastoma patients and marks rare, HIF1/2 α-positive cells in tumour hypoxic areas. Fzd6 positive neuroblastoma cells form neurospheres with high efficiency, are resistant to doxorubicin killing and express high levels of mesenchymal markers such as Twist1 and Notch1. Expression of Fzd6 is required for the expression of genes of the non-canonical Wnt pathway and the spheres forming activity. When transplanted into immunodeficient mice, neuroblastoma cells expressing the Fzd6 marker grow more aggressively than their Fzd6 negative counterparts. We conclude that Fzd6 is a new surface marker of aggressive neuroblastoma cells with stem cell-like features.
Collapse
Affiliation(s)
- Sandra Cantilena
- Molecular Haematology and Cancer Biology Unit, UCL Institute of Child Health, 30 Guilford st London WC1N 1EH London UK
| | | | | | | | | | | | | |
Collapse
|
41
|
Das S, Chang C. Regulation of early xenopus embryogenesis by Smad ubiquitination regulatory factor 2. Dev Dyn 2012; 241:1260-73. [PMID: 22674516 DOI: 10.1002/dvdy.23811] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Smad ubiquitination regulatory factor (Smurf) 1 and 2 are E3 ubiquitin ligases originally identified as inhibitors of transforming growth factor beta signaling and are shown to modulate multiple cellular activities. The roles of Smurfs in vertebrate embryogenesis, however, are not completely understood. RESULTS Here we investigate the function of Smurf2 during early Xenopus development. We show that distinctly from Smurf1, overexpression of Smurf2 in presumptive mesoderm interfered with mesoderm induction and caused axial defects, whereas knockdown of Smurf2 with antisense morpholino oligonucleotides resulted in expansion of the mesoderm. These results imply that Smurf2 may modulate nodal-mediated mesodermal induction. Consistently, ventral expression of Smurf2 induced a partial secondary axis with head structures. In the ectoderm, Smurf2 resembled Smurf1 in controlling neural and epidermal marker expression and influencing head formation. Smurf1, but not Smurf2, additionally affected neural tube closure. Interestingly, both Smurfs could enhance as well as repress neural crest markers, implying that they modulate their targets dynamically during neural plate border specification. CONCLUSION Our data demonstrate that Smurf1 and Smurf2 have overlapping and distinct functionalities during early frog embryogenesis; collectively, they regulate ectodermal and mesodermal induction and patterning to ensure normal development of Xenopus embryos.
Collapse
Affiliation(s)
- Shaonli Das
- Department of Cell Biology, University of Alabama, Birmingham, Alabama, USA
| | | |
Collapse
|
42
|
Liu HX, Komatsu Y, Mishina Y, Mistretta CM. Neural crest contribution to lingual mesenchyme, epithelium and developing taste papillae and taste buds. Dev Biol 2012; 368:294-303. [PMID: 22659543 DOI: 10.1016/j.ydbio.2012.05.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/21/2012] [Accepted: 05/23/2012] [Indexed: 01/06/2023]
Abstract
The epithelium of mammalian tongue hosts most of the taste buds that transduce gustatory stimuli into neural signals. In the field of taste biology, taste bud cells have been described as arising from "local epithelium", in distinction from many other receptor organs that are derived from neurogenic ectoderm including neural crest (NC). In fact, contribution of NC to both epithelium and mesenchyme in the developing tongue is not fully understood. In the present study we used two independent, well-characterized mouse lines, Wnt1-Cre and P0-Cre that express Cre recombinase in a NC-specific manner, in combination with two Cre reporter mouse lines, R26R and ZEG, and demonstrate a contribution of NC-derived cells to both tongue mesenchyme and epithelium including taste papillae and taste buds. In tongue mesenchyme, distribution of NC-derived cells is in close association with taste papillae. In tongue epithelium, labeled cells are observed in an initial scattered distribution and progress to a clustered pattern between papillae, and within papillae and early taste buds. This provides evidence for a contribution of NC to lingual epithelium. Together with previous reports for the origin of taste bud cells from local epithelium in postnatal mouse, we propose that NC cells migrate into and reside in the epithelium of the tongue primordium at an early embryonic stage, acquire epithelial cell phenotypes, and undergo cell proliferation and differentiation that is involved in the development of taste papillae and taste buds. Our findings lead to a new concept about derivation of taste bud cells that include a NC origin.
Collapse
Affiliation(s)
- Hong-Xiang Liu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | |
Collapse
|
43
|
Fishwick KJ, Kim E, Bronner ME. ILF-3 is a regulator of the neural plate border marker Zic1 in chick embryos. Dev Dyn 2012; 241:1325-32. [PMID: 22639388 DOI: 10.1002/dvdy.23809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2012] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The neural crest is a multipotent cell type unique to the vertebrate lineage and capable of differentiating into a large number of varied cell types, including ganglia of the peripheral nervous system, cartilage, and glia. An early step in neural crest specification occurs at the neural plate border, a region defined by the overlap of transcription factors of the Zic, Msx, and Pax families. RESULTS Here we identify a novel chick gene with close homology to double-stranded RNA-binding protein Interleukin enhancer binding factor 3 (ILF-3) in other species. Our results show that chick Ilf-3 is required for proper expression of the transcription factor, Zic-1, at the neural plate border. CONCLUSION We have identified a novel chick gene and show it has a role in the correct specification of Zic-1 at the neural plate border.
Collapse
Affiliation(s)
- K J Fishwick
- Division of Biology, California Institute of Technology, Pasadena, California, USA
| | | | | |
Collapse
|
44
|
McNulty MS, Bedell VM, Greenwood TM, Craig TA, Ekker SC, Kumar R. Expression of sclerostin in the developing zebrafish (Danio rerio) brain and skeleton. Gene Expr Patterns 2012; 12:228-35. [PMID: 22575304 DOI: 10.1016/j.gep.2012.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/28/2012] [Accepted: 04/27/2012] [Indexed: 11/15/2022]
Abstract
Sclerostin is a highly conserved, secreted, cystine-knot protein which regulates osteoblast function. Humans with mutations in the sclerostin gene (SOST), manifest increased axial and appendicular skeletal bone density with attendant complications. In adult bone, sclerostin is expressed in osteocytes and osteoblasts. Danio rerio sclerostin-like protein is closely related to sea bass sclerostin, and is related to chicken and mammalian sclerostins. Little is known about the expression of sclerostin in early developing skeletal or extra-skeletal tissues. We assessed sclerostin (sost) gene expression in developing zebrafish (D. rerio) embryos with whole mount is situ hybridization methods. The earliest expression of sost mRNA was noted during 12h post-fertilization (hpf). At 15 hpf, sost mRNA was detected in the developing nervous system and in Kupffer's vesicle. At 18, 20 and 22 hpf, expression in rhombic lip precursors was seen. By 24 hpf, expression in the upper and lower rhombic lip and developing spinal cord was noted. Expression in the rhombic lip and spinal cord persisted through 28 hpf and then diminished in intensity through 44 hpf. At 28 hpf, sost expression was noted in developing pharyngeal cartilage; expression in pharyngeal cartilage increased with time. By 48 hpf, sost mRNA was clearly detected in the developing pharyngeal arch cartilage. Sost mRNA was abundantly expressed in the pharyngeal arch cartilage, and in developing pectoral fins, 72, 96 and 120 hpf. Our study is the first detailed analysis of sost gene expression in early metazoan development.
Collapse
Affiliation(s)
- Melissa S McNulty
- Division of Nephrology and Hypertension, Mayo Clinic, 200 1st St., Southwest, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
45
|
Monsonego-Ornan E, Kosonovsky J, Bar A, Roth L, Fraggi-Rankis V, Simsa S, Kohl A, Sela-Donenfeld D. Matrix metalloproteinase 9/gelatinase B is required for neural crest cell migration. Dev Biol 2012; 364:162-77. [DOI: 10.1016/j.ydbio.2012.01.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 01/30/2012] [Accepted: 01/31/2012] [Indexed: 11/27/2022]
|
46
|
Tu CT, Yang TC, Huang HY, Tsai HJ. Zebrafish arl6ip1 is required for neural crest development during embryogenesis. PLoS One 2012; 7:e32899. [PMID: 22427906 PMCID: PMC3298456 DOI: 10.1371/journal.pone.0032899] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 02/06/2012] [Indexed: 12/02/2022] Open
Abstract
Background Although the embryonic expression pattern of ADP ribosylation factor-like 6 interacting protein 1 (Arl6ip1) has been reported, its function in neural crest development is unclear. Methods/Principal Findings We found that knockdown of Arl6ip1 caused defective embryonic neural crest derivatives that were particularly severe in craniofacial cartilages. Expressions of the ectodermal patterning factors msxb, dlx3b, and pax3 were normal, but the expressions of the neural crest specifier genes foxd3, snai1b, and sox10 were greatly reduced. These findings suggest that arl6ip1 is essential for specification of neural crest derivatives, but not neural crest induction. Furthermore, we revealed that the streams of crestin- and sox10-expressing neural crest cells, which migrate ventrally from neural tube into trunk, were disrupted in arl6ip1 morphants. This migration defect was not only in the trunk neural crest, but also in the enteric tract where the vagal-derived neural crest cells failed to populate the enteric nervous system. We found that this migration defect was induced by dampened Shh signaling, which may have resulted from defective cilia. These data further suggested that arl6ip1 is required for neural crest migration. Finally, by double-staining of TUNEL and crestin, we confirmed that the loss of neural crest cells could not be attributed to apoptosis. Conclusions/Significance Therefore, we concluded that arl6ip1 is required for neural crest migration and sublineage specification.
Collapse
Affiliation(s)
| | | | | | - Huai-Jen Tsai
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
47
|
Bohnsack BL, Kasprick DS, Kish PE, Goldman D, Kahana A. A zebrafish model of axenfeld-rieger syndrome reveals that pitx2 regulation by retinoic acid is essential for ocular and craniofacial development. Invest Ophthalmol Vis Sci 2012; 53:7-22. [PMID: 22125274 PMCID: PMC3292384 DOI: 10.1167/iovs.11-8494] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 10/09/2011] [Accepted: 11/10/2011] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The homeobox transcription factor PITX2 is a known regulator of mammalian ocular development, and human PITX2 mutations are associated with Axenfeld-Rieger syndrome (ARS). However, the treatment of patients with ARS remains mostly supportive and palliative. METHODS The authors used molecular genetic, pharmacologic, and embryologic techniques to study the biology of ARS in a zebrafish model that uses transgenes to mark neural crest and muscle cells in the head. RESULTS The authors demonstrated in vivo that pitx2 is a key downstream target of retinoic acid (RA) in craniofacial development, and this pathway is required for coordinating neural crest, mesoderm, and ocular development. pitx2a knockdown using morpholino oligonucleotides disrupts jaw and pharyngeal arch formation and recapitulates ocular characteristics of ARS, including corneal and iris stroma maldevelopment. These phenotypes could be rescued with human PITX2A mRNA, demonstrating the specificity of the knockdown and evolutionary conservation of pitx2a function. Expression of the ARS dominant negative human PITX2A K50E allele also caused ARS-like phenotypes. Similarly, inhibition of RA synthesis in the developing eye (genetic or pharmacologic) disrupted craniofacial and ocular development, and human PITX2A mRNA partially rescued these defects. CONCLUSIONS RA regulation of pitx2 is essential for coordinating interactions among neural crest, mesoderm, and developing eye. The marked evolutionary conservation of Pitx2 function in eye and craniofacial development makes zebrafish a potentially powerful model of ARS, amenable to in vivo experimentation and development of potential therapies.
Collapse
Affiliation(s)
- Brenda L Bohnsack
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | |
Collapse
|
48
|
Archbold HC, Yang YX, Chen L, Cadigan KM. How do they do Wnt they do?: regulation of transcription by the Wnt/β-catenin pathway. Acta Physiol (Oxf) 2012; 204:74-109. [PMID: 21624092 DOI: 10.1111/j.1748-1716.2011.02293.x] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Wnt/β-catenin signalling is known to play many roles in metazoan development and tissue homeostasis. Misregulation of the pathway has also been linked to many human diseases. In this review, specific aspects of the pathway's involvement in these processes are discussed, with an emphasis on how Wnt/β-catenin signalling regulates gene expression in a cell and temporally specific manner. The T-cell factor (TCF) family of transcription factors, which mediate a large portion of Wnt/β-catenin signalling, will be discussed in detail. Invertebrates contain a single TCF gene that contains two DNA-binding domains, the high mobility group (HMG) domain and the C-clamp, which increases the specificity of DNA binding. In vertebrates, the situation is more complex, with four TCF genes producing many isoforms that contain the HMG domain, but only some of which possess a C-clamp. Vertebrate TCFs have been reported to act in concert with many other transcription factors, which may explain how they obtain sufficient specificity for specific DNA sequences, as well as how they achieve a wide diversity of transcriptional outputs in different cells.
Collapse
Affiliation(s)
- H C Archbold
- Program in Cell and Molecular Biology, University of Michigan, Ann Arbor, 48109-1048, USA
| | | | | | | |
Collapse
|
49
|
Gouti M, Briscoe J, Gavalas A. Anterior Hox genes interact with components of the neural crest specification network to induce neural crest fates. Stem Cells 2011; 29:858-70. [PMID: 21433221 PMCID: PMC3184476 DOI: 10.1002/stem.630] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 03/02/2011] [Indexed: 12/29/2022]
Abstract
Hox genes play a central role in neural crest (NC) patterning particularly in the cranial region of the body. Despite evidence that simultaneous loss of Hoxa1 and Hoxb1 function resulted in NC specification defects, the role of Hox genes in NC specification has remained unclear due to extended genetic redundancy among Hox genes. To circumvent this problem, we expressed anterior Hox genes in the trunk neural tube of the developing chick embryo. This demonstrated that anterior Hox genes play a central role in NC cell specification by rapidly inducing the key transcription factors Snail2 and Msx1/2 and a neural progenitor to NC cell fate switch characterized by cell adhesion changes and an epithelial-to-mesenchymal transition (EMT). Cells delaminated from dorsal and medial neural tube levels and generated ectopic neurons, glia progenitors, and melanocytes. The mobilization of the NC genetic cascade was dependent upon bone morphogenetic protein signaling and optimal levels of Notch signaling. Therefore, anterior Hox patterning genes participate in NC specification and EMT by interacting with NC-inducing signaling pathways and regulating the expression of key genes involved in these processes.
Collapse
Affiliation(s)
- Mina Gouti
- Developmental Biology Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA)Athens, Greece
| | - James Briscoe
- Division of Developmental Neurobiology, MRC National Institute for Medical Research (NIMR)The Ridgeway, Mill Hill, London, United Kingdom
| | - Anthony Gavalas
- Developmental Biology Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA)Athens, Greece
| |
Collapse
|
50
|
Katiyar P, Aplin AE. FOXD3 regulates migration properties and Rnd3 expression in melanoma cells. Mol Cancer Res 2011; 9:545-52. [PMID: 21478267 DOI: 10.1158/1541-7786.mcr-10-0454] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Forkhead transcription factor, Foxd3, plays a critical role during development by controlling the lineage specification of neural crest cells. Notably, Foxd3 is highly expressed during the wave of neural crest cell migration that forms peripheral neurons and glial cells but is downregulated prior to migration of cells that give rise to the melanocytic lineage. Melanoma is the deadliest form of skin cancer and is derived from melanocytes. Recently, we showed that FOXD3 expression is elevated following the targeted inhibition of the B-RAF-MEK (MAP/ERK kinase)-ERK (extracellular signal-regulated kinase)1/2 pathway in mutant B-RAF melanoma cells. Because melanoma cells are highly migratory and invasive in a B-RAF-dependent manner, we explored the role of FOXD3 in these processes. In this study, we show that ectopic FOXD3 expression inhibits the migration, invasion, and spheroid outgrowth of mutant B-RAF melanoma cells. Upregulation of FOXD3 expression following inhibition of B-RAF and MEK correlates with the downregulation of Rnd3, a Rho GTPase and inhibitor of RhoA-ROCK signaling. Indeed, expression of FOXD3 alone was sufficient to downregulate Rnd3 expression at the mRNA and protein levels. Mechanistically, FOXD3 was found to be recruited to the Rnd3 promoter. Inhibition of ROCK partially restored migration in FOXD3-expressing cells. These data show that FOXD3 expression downregulates migration and invasion in melanoma cells and Rnd3, a target known to be involved in these properties.
Collapse
Affiliation(s)
- Pragati Katiyar
- Department of Cancer Biology, Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA 19107, USA
| | | |
Collapse
|