1
|
Tu M, Ge B, Li J, Pan Y, Zhao B, Han J, Wu J, Zhang K, Liu G, Hou M, Yue M, Han X, Sun T, An Y. Emerging biological functions of Twist1 in cell differentiation. Dev Dyn 2024. [PMID: 39254141 DOI: 10.1002/dvdy.736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/03/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Twist1 is required for embryonic development and expresses after birth in mesenchymal stem cells derived from mesoderm, where it governs mesenchymal cell development. As a well-known regulator of epithelial-mesenchymal transition or embryonic organogenesis, Twist1 is important in a variety of developmental systems, including mesoderm formation, neurogenesis, myogenesis, cranial neural crest cell migration, and differentiation. In this review, we first highlight the physiological significance of Twist1 in cell differentiation, including osteogenic, chondrogenic, and myogenic differentiation, and then detail its probable molecular processes and signaling pathways. On this premise, we summarize the significance of Twist1 in distinct developmental disorders and diseases to provide a reference for studies on cell differentiation/development-related diseases.
Collapse
Affiliation(s)
- Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Bingqian Ge
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jialin Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Kaifeng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Guangchao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Pei F, Guo T, Zhang M, Ma L, Jing J, Feng J, Ho TV, Wen Q, Chai Y. FGF signaling modulates mechanotransduction/WNT signaling in progenitors during tooth root development. Bone Res 2024; 12:37. [PMID: 38910207 PMCID: PMC11194271 DOI: 10.1038/s41413-024-00345-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/25/2024] Open
Abstract
Stem/progenitor cells differentiate into different cell lineages during organ development and morphogenesis. Signaling pathway networks and mechanotransduction are important factors to guide the lineage commitment of stem/progenitor cells during craniofacial tissue morphogenesis. Here, we used tooth root development as a model to explore the roles of FGF signaling and mechanotransduction as well as their interaction in regulating the progenitor cell fate decision. We show that Fgfr1 is expressed in the mesenchymal progenitor cells and their progeny during tooth root development. Loss of Fgfr1 in Gli1+ progenitors leads to hyperproliferation and differentiation, which causes narrowed periodontal ligament (PDL) space with abnormal cementum/bone formation leading to ankylosis. We further show that aberrant activation of WNT signaling and mechanosensitive channel Piezo2 occurs after loss of FGF signaling in Gli1-CreER;Fgfr1fl/fl mice. Overexpression of Piezo2 leads to increased osteoblastic differentiation and decreased Piezo2 leads to downregulation of WNT signaling. Mechanistically, an FGF/PIEZO2/WNT signaling cascade plays a crucial role in modulating the fate of progenitors during root morphogenesis. Downregulation of WNT signaling rescues tooth ankylosis in Fgfr1 mutant mice. Collectively, our findings uncover the mechanism by which FGF signaling regulates the fate decisions of stem/progenitor cells, and the interactions among signaling pathways and mechanotransduction during tooth root development, providing insights for future tooth root regeneration.
Collapse
Affiliation(s)
- Fei Pei
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Tingwei Guo
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Mingyi Zhang
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Li Ma
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Junjun Jing
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Jifan Feng
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Thach-Vu Ho
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Quan Wen
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA.
| |
Collapse
|
3
|
Jin C, Adachi N, Yoshimoto Y, Sasabuchi A, Kawashima N, Ota MS, Iseki S. Fibroblast growth factor signalling regulates the development of tooth root. J Anat 2024; 244:1067-1077. [PMID: 38258312 PMCID: PMC11095309 DOI: 10.1111/joa.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/03/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Fibroblast growth factor (FGF) signalling plays a crucial role in the morphogenesis of multiple tissues including teeth. While the role of the signal has been studied in tooth crown development, little is known about root development. Of several FGF ligands involved in hard tissue formation, we suggest that FGF18 regulates the development of murine tooth roots. We implanted FGF18-soaked heparin beads into the lower first molar tooth buds at postnatal day 6 (P6), followed by transplantation under the kidney capsule. After 3 weeks, FGF18 significantly facilitated root elongation and periodontal tissue formation compared to the control. In situ hybridisation showed that Fgf18 transcripts were initially localised in the dental pulp along Hertwig's epithelial root sheath at P6 and P10 and subsequently in the dental follicle cells at P14. Fgf receptors were expressed in various dental tissues during these stages. In vitro analysis using the dental pulp stem cells revealed that FGF18 inhibited cell proliferation and decreased expression levels of osteogenic markers, Runx2, Alpl and Sp7. Consistently, after 1 week of kidney capsule transplantation, FGF18 application did not induce the expression of Sp7 and Bsp, but upregulated Periostin in the apical region of dental mesenchyme in the grafted molar. These findings suggest that FGF18 facilitates molar root development by regulating the calcification of periodontal tissues.
Collapse
Affiliation(s)
- Chengxue Jin
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Oral, Plastic and Aesthetic Surgery, Hospital of Stomatology, Jilin University, Changchun, Jilin, China
| | - Noritaka Adachi
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Yoshimoto
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Aino Sasabuchi
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Nobuyuki Kawashima
- Department of Pulp Biology and Endodontics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masato S Ota
- Laboratory of Anatomy, Physiology and Food Biological Science, Department of Food and Nutrition, Faculty of Human Sciences and Design, Japan Women's University, Tokyo, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology and Oral Histology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
4
|
Ou W, Qi Z, Liu N, Zhang J, Mi X, Song Y, Fang Y, Cui B, Hou J, Yuan Z. Elucidating the role of TWIST1 in ulcerative colitis: a comprehensive bioinformatics and machine learning approach. Front Genet 2024; 15:1296570. [PMID: 38510272 PMCID: PMC10952112 DOI: 10.3389/fgene.2024.1296570] [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: 09/19/2023] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
Background: Ulcerative colitis (UC) is a common and progressive inflammatory bowel disease primarily affecting the colon and rectum. Prolonged inflammation can lead to colitis-associated colorectal cancer (CAC). While the exact cause of UC remains unknown, this study aims to investigate the role of the TWIST1 gene in UC. Methods: Second-generation sequencing data from adult UC patients were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified, and characteristic genes were selected using machine learning and Lasso regression. The Receiver Operating Characteristic (ROC) curve assessed TWIST1's potential as a diagnostic factor (AUC score). Enriched pathways were analyzed, including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Variation Analysis (GSVA). Functional mechanisms of marker genes were predicted, considering immune cell infiltration and the competing endogenous RNA (ceRNA) network. Results: We found 530 DEGs, with 341 upregulated and 189 downregulated genes. TWIST1 emerged as one of four potential UC biomarkers via machine learning. TWIST1 expression significantly differed in two datasets, GSE193677 and GSE83687, suggesting its diagnostic potential (AUC = 0.717 in GSE193677, AUC = 0.897 in GSE83687). Enrichment analysis indicated DEGs associated with TWIST1 were involved in processes like leukocyte migration, humoral immune response, and cell chemotaxis. Immune cell infiltration analysis revealed higher rates of M0 macrophages and resting NK cells in the high TWIST1 expression group, while TWIST1 expression correlated positively with M2 macrophages and resting NK cell infiltration. We constructed a ceRNA regulatory network involving 1 mRNA, 7 miRNAs, and 32 long non-coding RNAs (lncRNAs) to explore TWIST1's regulatory mechanism. Conclusion: TWIST1 plays a significant role in UC and has potential as a diagnostic marker. This study sheds light on UC's molecular mechanisms and underscores TWIST1's importance in its progression. Further research is needed to validate these findings in diverse populations and investigate TWIST1 as a therapeutic target in UC.
Collapse
Affiliation(s)
- Wenjie Ou
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhaoxue Qi
- Department of Secretory Metabolism, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ning Liu
- General Surgery of The First Clinical Hospital of Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin, China
| | - Junzi Zhang
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xuguang Mi
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yuan Song
- Department of Gastroenterology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Yanqiu Fang
- Department of Central Laboratory, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Baiying Cui
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junjie Hou
- Department of Comprehensive Oncology, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| | - Zhixin Yuan
- Department of Emergency Surgery, Jilin Provincial People’s Hospital, Changchun, Jilin, China
| |
Collapse
|
5
|
Toriumi K, Onodera Y, Takehara T, Mori T, Hasei J, Shigi K, Iwawaki N, Ozaki T, Akagi M, Nakanishi M, Teramura T. LRRC15 expression indicates high level of stemness regulated by TWIST1 in mesenchymal stem cells. iScience 2023; 26:106946. [PMID: 37534184 PMCID: PMC10391581 DOI: 10.1016/j.isci.2023.106946] [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: 12/25/2022] [Revised: 04/09/2023] [Accepted: 05/19/2023] [Indexed: 08/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are used as a major source for cell therapy, and its application is expanding in various diseases. On the other hand, reliable method to evaluate quality and therapeutic properties of MSC is limited. In this study, we focused on TWIST1 that is a transcription factor regulating stemness of MSCs and found that the transmembrane protein LRRC15 tightly correlated with the expression of TWIST1 and useful to expect TWIST1-regulated stemness of MSCs. The LRRC15-positive MSC populations in human and mouse bone marrow tissues highly expressed stemness-associated transcription factors and therapeutic cytokines, and showed better therapeutic effect in bleomycin-induced pulmonary fibrosis model mice. This study provides evidence for the important role of TWIST1 in the MSC stemness, and for the utility of the LRRC15 protein as a marker to estimate stem cell quality in MSCs before cell transplantation.
Collapse
Affiliation(s)
- Kensuke Toriumi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Yuta Onodera
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshiyuki Takehara
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Tatsufumi Mori
- Life Science Institute, Kindai University, Osaka-sayama, Osaka, Japan
| | - Joe Hasei
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Kanae Shigi
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Natsumi Iwawaki
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Faculty of Medicine, Okayama, Okayama, Japan
| | - Masao Akagi
- Department of Orthopedic Surgery, Kindai University Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | | | - Takeshi Teramura
- Institute of Advanced Clinical Medicine, Kindai University Hospital, Osaka-sayama, Osaka, Japan
| |
Collapse
|
6
|
Koyabu D. Evolution, conservatism and overlooked homologies of the mammalian skull. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220081. [PMID: 37183902 PMCID: PMC10184252 DOI: 10.1098/rstb.2022.0081] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/22/2023] [Indexed: 05/16/2023] Open
Abstract
In the last decade, studies integrating palaeontology, embryology and experimental developmental biology have markedly altered our homological understanding of the mammalian skull. Indeed, new evidence suggests that we should revisit and restructure the conventional anatomical terminology applied to the components of the mammalian skull. Notably, these are classical problems that have remained unresolved since the ninteenth century. In this review, I offer perspectives on the overlooked problems associated with the homology, development, and conservatism of the mammalian skull, aiming to encourage future studies in these areas. I emphasise that ossification patterns, bone fusion, cranial sutures and taxon-specific neomorphic bones in the skull are virtually unexplored, and further studies would improve our homological understanding of the mammalian skull. Lastly, I highlight that overlooked bones may exist in the skull that are not yet known to science and suggest that further search is needed. This article is part of the theme issue 'The mammalian skull: development, structure and function'.
Collapse
Affiliation(s)
- Daisuke Koyabu
- Research and Development Center for Precision Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
7
|
Zhao X, Erhardt S, Sung K, Wang J. FGF signaling in cranial suture development and related diseases. Front Cell Dev Biol 2023; 11:1112890. [PMID: 37325554 PMCID: PMC10267317 DOI: 10.3389/fcell.2023.1112890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Suture mesenchymal stem cells (SMSCs) are a heterogeneous stem cell population with the ability to self-renew and differentiate into multiple cell lineages. The cranial suture provides a niche for SMSCs to maintain suture patency, allowing for cranial bone repair and regeneration. In addition, the cranial suture functions as an intramembranous bone growth site during craniofacial bone development. Defects in suture development have been implicated in various congenital diseases, such as sutural agenesis and craniosynostosis. However, it remains largely unknown how intricate signaling pathways orchestrate suture and SMSC function in craniofacial bone development, homeostasis, repair and diseases. Studies in patients with syndromic craniosynostosis identified fibroblast growth factor (FGF) signaling as an important signaling pathway that regulates cranial vault development. A series of in vitro and in vivo studies have since revealed the critical roles of FGF signaling in SMSCs, cranial suture and cranial skeleton development, and the pathogenesis of related diseases. Here, we summarize the characteristics of cranial sutures and SMSCs, and the important functions of the FGF signaling pathway in SMSC and cranial suture development as well as diseases caused by suture dysfunction. We also discuss emerging current and future studies of signaling regulation in SMSCs.
Collapse
Affiliation(s)
- Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| | - Kihan Sung
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- MD Anderson Cancer Center and UT Health Graduate School of Biomedical Sciences, The University of Texas, Houston, TX, United States
| |
Collapse
|
8
|
Tooze RS, Calpena E, Weber A, Wilson LC, Twigg SRF, Wilkie AOM. Review of Recurrently Mutated Genes in Craniosynostosis Supports Expansion of Diagnostic Gene Panels. Genes (Basel) 2023; 14:615. [PMID: 36980886 PMCID: PMC10048212 DOI: 10.3390/genes14030615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Craniosynostosis, the premature fusion of the cranial sutures, affects ~1 in 2000 children. Although many patients with a genetically determined cause harbor a variant in one of just seven genes or have a chromosomal abnormality, over 60 genes are known to be recurrently mutated, thus comprising a long tail of rarer diagnoses. Genome sequencing for the diagnosis of rare diseases is increasingly used in clinical settings, but analysis of the data is labor intensive and involves a trade-off between achieving high sensitivity or high precision. PanelApp, a crowd-sourced disease-focused set of gene panels, was designed to enable prioritization of variants in known disease genes for a given pathology, allowing enhanced identification of true-positives. For heterogeneous disorders like craniosynostosis, these panels must be regularly updated to ensure that diagnoses are not being missed. We provide a systematic review of genetic literature on craniosynostosis over the last 5 years, including additional results from resequencing a 42-gene panel in 617 affected individuals. We identify 16 genes (representing a 25% uplift) that should be added to the list of bona fide craniosynostosis disease genes and discuss the insights that these new genes provide into pathophysiological mechanisms of craniosynostosis.
Collapse
Affiliation(s)
- Rebecca S. Tooze
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Eduardo Calpena
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Astrid Weber
- Liverpool Centre for Genomic Medicine, Liverpool Women’s NHS Foundation Trust, Liverpool L8 7SS, UK
| | - Louise C. Wilson
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Stephen R. F. Twigg
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Andrew O. M. Wilkie
- Clinical Genetics Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
9
|
Cooper RL, Nicklin EF, Rasch LJ, Fraser GJ. Teeth outside the mouth: The evolution and development of shark denticles. Evol Dev 2023; 25:54-72. [PMID: 36594351 DOI: 10.1111/ede.12427] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/06/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023]
Abstract
Vertebrate skin appendages are incredibly diverse. This diversity, which includes structures such as scales, feathers, and hair, likely evolved from a shared anatomical placode, suggesting broad conservation of the early development of these organs. Some of the earliest known skin appendages are dentine and enamel-rich tooth-like structures, collectively known as odontodes. These appendages evolved over 450 million years ago. Elasmobranchs (sharks, skates, and rays) have retained these ancient skin appendages in the form of both dermal denticles (scales) and oral teeth. Despite our knowledge of denticle function in adult sharks, our understanding of their development and morphogenesis is less advanced. Even though denticles in sharks appear structurally similar to oral teeth, there has been limited data directly comparing the molecular development of these distinct elements. Here, we chart the development of denticles in the embryonic small-spotted catshark (Scyliorhinus canicula) and characterize the expression of conserved genes known to mediate dental development. We find that shark denticle development shares a vast gene expression signature with developing teeth. However, denticles have restricted regenerative potential, as they lack a sox2+ stem cell niche associated with the maintenance of a dental lamina, an essential requirement for continuous tooth replacement. We compare developing denticles to other skin appendages, including both sensory skin appendages and avian feathers. This reveals that denticles are not only tooth-like in structure, but that they also share an ancient developmental gene set that is likely common to all epidermal appendages.
Collapse
Affiliation(s)
- Rory L Cooper
- Department of Genetics and Evolution, The University of Geneva, Geneva, Switzerland
| | - Ella F Nicklin
- Department of Biology, University of Florida, Gainesville, Florida, USA
| | - Liam J Rasch
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Gareth J Fraser
- Department of Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Ang PS, Matrongolo MJ, Zietowski ML, Nathan SL, Reid RR, Tischfield MA. Cranium growth, patterning and homeostasis. Development 2022; 149:dev201017. [PMID: 36408946 PMCID: PMC9793421 DOI: 10.1242/dev.201017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Craniofacial development requires precise spatiotemporal regulation of multiple signaling pathways that crosstalk to coordinate the growth and patterning of the skull with surrounding tissues. Recent insights into these signaling pathways and previously uncharacterized progenitor cell populations have refined our understanding of skull patterning, bone mineralization and tissue homeostasis. Here, we touch upon classical studies and recent advances with an emphasis on developmental and signaling mechanisms that regulate the osteoblast lineage for the calvaria, which forms the roof of the skull. We highlight studies that illustrate the roles of osteoprogenitor cells and cranial suture-derived stem cells for proper calvarial growth and homeostasis. We also discuss genes and signaling pathways that control suture patency and highlight how perturbing the molecular regulation of these pathways leads to craniosynostosis. Finally, we discuss the recently discovered tissue and signaling interactions that integrate skull and cerebrovascular development, and the potential implications for both cerebrospinal fluid hydrodynamics and brain waste clearance in craniosynostosis.
Collapse
Affiliation(s)
- Phillip S. Ang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Matt J. Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Shelby L. Nathan
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Max A. Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
11
|
Zhou W, Bai Y, Chen J, Li H, Zhang B, Liu H. Revealing the Critical Regulators of Modulated Smooth Muscle Cells in Atherosclerosis in Mice. Front Genet 2022; 13:900358. [PMID: 35677564 PMCID: PMC9168464 DOI: 10.3389/fgene.2022.900358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/15/2022] [Indexed: 01/23/2023] Open
Abstract
Background: There are still residual risks for atherosclerosis (AS)-associated cardiovascular diseases to be resolved. Considering the vital role of phenotypic switching of smooth muscle cells (SMCs) in AS, especially in calcification, targeting SMC phenotypic modulation holds great promise for clinical implications. Methods: To perform an unbiased and systematic analysis of the molecular regulatory mechanism of phenotypic switching of SMCs during AS in mice, we searched and included several publicly available single-cell datasets from the GEO database, resulting in an inclusion of more than 80,000 cells. Algorithms implemented in the Seurat package were used for cell clustering and cell atlas depiction. The pySCENIC and SCENIC packages were used to identify master regulators of interested cell groups. Monocle2 was used to perform pseudotime analysis. clusterProfiler was used for Gene Ontology enrichment analysis. Results: After dimensionality reduction and clustering, reliable annotation was performed. Comparative analysis between cells from normal artery and AS lesions revealed that three clusters emerged as AS progression, designated as mSMC1, mSMC2, and mSMC3. Transcriptional and functional enrichment analysis established a continuous transitional mode of SMCs’ transdifferentiation to mSMCs, which is further supported by pseudotime analysis. A total of 237 regulons were identified with varying activity scores across cell types. A potential core regulatory network was constructed for SMC and mSMC subtypes. In addition, module analysis revealed a coordinate regulatory mode of regulons for a specific cell type. Intriguingly, consistent with gain of ossification-related transcriptional and functional characteristics, a corresponding small set of regulators contributing to osteochondral reprogramming was identified in mSMC3, including Dlx5, Sox9, and Runx2. Conclusion: Gene regulatory network inference indicates a hierarchical organization of regulatory modules that work together in fine-tuning cellular states. The analysis here provides a valuable resource that can provide guidance for subsequent biological experiments.
Collapse
Affiliation(s)
- Wenli Zhou
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yongyi Bai
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianqiao Chen
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huiying Li
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Baohua Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Health Care, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Hongbin Liu,
| |
Collapse
|
12
|
Mo SS, Kim JW, Baik HS, Giap HV, Lee KJ. Age-related osteogenesis on lateral force application to rat incisor – Part III: Periodontal and periosteal bone remodeling. APOS TRENDS IN ORTHODONTICS 2022. [DOI: 10.25259/apos_125_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Objectives:
This study was aimed to compare the histological pattern of bone modeling on either periodontal or periosteal side induced by lateral orthodontic tooth movement in different age groups.
Material and Methods:
A total of 50 male Sprague-Dawley rats (25 rats in the adult group – 52 weeks and 25 rats in the young group – 10 weeks) were utilized in this study. Each age group was classified into the control, 3 days, 7 days, 14 days, and 21 days groups (five rats in each) by the duration of experimental device application. A double-helical spring was produced using 0.014” stainless steel wire to provide 40 g lateral force to the left and right incisors. Hematoxylin-eosin staining, proliferating cell nuclear antigen (PCNA) immunohistochemical staining, fibroblast growth factor receptor 2 (FGFR2) immunohistochemical staining, and Masson trichrome staining were performed; and the slides were subject to histological examination.
Results:
In 7 days, active bone modeling represented by the scalloped surface was observed on the periosteal side of the crestal and middle alveolus at the pressure side in the young group, while similar changes were observed only on the crestal area in the adult group. In the young group, the number of PCNA-positive cells increased significantly on the crestal area and middle alveolus on the 3, 7, and 14 day groups, with subsequent decrease at 21 days. In the adult group, PCNA-positive cells were localized on the crestal area throughout the period. In the young group, FGFR2-positive cells were observed mainly on the crestal and middle alveolus at 3, 7, and 14 days than the control group. In the adult group, these cells appeared on the crestal and middle alveolus in the 3 days group, but mainly on the crestal area at 14 days. In the young group, FGFR2-positive cells were observed on the crestal and middle alveolus on the 3, 7, and 14 days groups more than on the control group. In the adult group, these cells appeared on the crestal and middle alveolus in the 3 days group, but mainly on the crestal area in the 14 days group. In Masson trichrome stain, an increased number of type I collagen fibers were observed after helical spring activation in both age groups. Large resorption lacunae indicating undermining bone resorption were progressively present in both young and adult groups.
Conclusion:
According to these results, orthodontic tooth movement may stimulate cell proliferation and differentiation primarily on the periosteal side according to progressive undermining bone resorption on the periodontal side. This response may lead to prominent bone modeling during tooth movement in the young group, compared to the relatively delayed response in the adult group.
Collapse
Affiliation(s)
- Sung-Seo Mo
- Department of Orthodontics, Division of Dentistry, College of Medicine, The Catholic University, Seoul, Korea,
| | - Jin-Wook Kim
- Department of Orthodontics, College of Dentistry, Institute of Craniofacial Deformity, Seoul, Korea,
| | - Hyoung-Seon Baik
- Department of Orthodontics, College of Dentistry, Institute of Craniofacial Deformity, Seoul, Korea,
| | - Hai-Van Giap
- Department of Orthodontics, College of Dentistry, Institute of Craniofacial Deformity, Seoul, Korea,
| | - Kee-Joon Lee
- Department of Orthodontics, College of Dentistry, Institute of Craniofacial Deformity, Seoul, Korea,
| |
Collapse
|
13
|
Biosse Duplan M, Dambroise E, Estibals V, Veziers J, Guicheux J, Legeai-Mallet L. An Fgfr3-activating mutation in immature murine osteoblasts affects the appendicular and craniofacial skeleton. Dis Model Mech 2021; 14:dmm048272. [PMID: 33737326 PMCID: PMC8084574 DOI: 10.1242/dmm.048272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Achondroplasia (ACH), the most common form of dwarfism, is caused by a missense mutation in the gene coding for fibroblast growth factor receptor 3 (FGFR3). The resulting increase in FGFR3 signaling perturbs the proliferation and differentiation of chondrocytes (CCs), alters the process of endochondral ossification and thus reduces bone elongation. Increased FGFR3 signaling in osteoblasts (OBs) might also contribute to bone anomalies in ACH. In the present study of a mouse model of ACH, we sought to determine whether FGFR3 overactivation in OBs leads to bone modifications. The model carries an Fgfr3-activating mutation (Fgfr3Y367C/+) that accurately mimics ACH; we targeted the mutation to either immature OBs and hypertrophic CCs or to mature OBs by using the Osx-cre and collagen 1α1 (2.3 kb Col1a1)-cre mouse strains, respectively. We observed that Fgfr3 activation in immature OBs and hypertrophic CCs (Osx-Fgfr3) not only perturbed the hypertrophic cells of the growth plate (thus affecting long bone growth) but also led to osteopenia and low cortical thickness in long bones in adult (3-month-old) mice but not growing (3-week-old) mice. Importantly, craniofacial membranous bone defects were present in the adult mice. In contrast, activation of Fgfr3 in mature OBs (Col1-Fgfr3) had very limited effects on skeletal shape, size and micro-architecture. In vitro, we observed that Fgfr3 activation in immature OBs was associated with low mineralization activity. In conclusion, immature OBs appear to be affected by Fgfr3 overactivation, which might contribute to the bone modifications observed in ACH independently of CCs.
Collapse
Affiliation(s)
- Martin Biosse Duplan
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
- Service de Médecine Bucco-Dentaire, Hôpital Bretonneau, AP-HP, Paris 75018, France
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Valentin Estibals
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| | - Joelle Veziers
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Jérome Guicheux
- Inserm, UMR 1229, RMeS – Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, F-44042, France
- SC3M, SFR Santé F. Bonamy, FED 4203, UMS Inserm 016, CNRS 3556, Nantes F-44042, France
- CHU Nantes, PHU4 OTONN, Nantes, F-44093, France
| | - Laurence Legeai-Mallet
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, Imagine Institute, Paris 75015, France
- Université de Paris, Paris 75006, France
| |
Collapse
|
14
|
White HE, Goswami A, Tucker AS. The Intertwined Evolution and Development of Sutures and Cranial Morphology. Front Cell Dev Biol 2021; 9:653579. [PMID: 33842480 PMCID: PMC8033035 DOI: 10.3389/fcell.2021.653579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022] Open
Abstract
Phenotypic variation across mammals is extensive and reflects their ecological diversification into a remarkable range of habitats on every continent and in every ocean. The skull performs many functions to enable each species to thrive within its unique ecological niche, from prey acquisition, feeding, sensory capture (supporting vision and hearing) to brain protection. Diversity of skull function is reflected by its complex and highly variable morphology. Cranial morphology can be quantified using geometric morphometric techniques to offer invaluable insights into evolutionary patterns, ecomorphology, development, taxonomy, and phylogenetics. Therefore, the skull is one of the best suited skeletal elements for developmental and evolutionary analyses. In contrast, less attention is dedicated to the fibrous sutural joints separating the cranial bones. Throughout postnatal craniofacial development, sutures function as sites of bone growth, accommodating expansion of a growing brain. As growth frontiers, cranial sutures are actively responsible for the size and shape of the cranial bones, with overall skull shape being altered by changes to both the level and time period of activity of a given cranial suture. In keeping with this, pathological premature closure of sutures postnatally causes profound misshaping of the skull (craniosynostosis). Beyond this crucial role, sutures also function postnatally to provide locomotive shock absorption, allow joint mobility during feeding, and, in later postnatal stages, suture fusion acts to protect the developed brain. All these sutural functions have a clear impact on overall cranial function, development and morphology, and highlight the importance that patterns of suture development have in shaping the diversity of cranial morphology across taxa. Here we focus on the mammalian cranial system and review the intrinsic relationship between suture development and morphology and cranial shape from an evolutionary developmental biology perspective, with a view to understanding the influence of sutures on evolutionary diversity. Future work integrating suture development into a comparative evolutionary framework will be instrumental to understanding how developmental mechanisms shaping sutures ultimately influence evolutionary diversity.
Collapse
Affiliation(s)
- Heather E White
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Anjali Goswami
- Department of Life Sciences, Natural History Museum, London, United Kingdom.,Division of Biosciences, University College London, London, United Kingdom
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
15
|
DeSisto J, O'Rourke R, Jones HE, Pawlikowski B, Malek AD, Bonney S, Guimiot F, Jones KL, Siegenthaler JA. Single-Cell Transcriptomic Analyses of the Developing Meninges Reveal Meningeal Fibroblast Diversity and Function. Dev Cell 2021; 54:43-59.e4. [PMID: 32634398 DOI: 10.1016/j.devcel.2020.06.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 03/18/2020] [Accepted: 06/01/2020] [Indexed: 01/18/2023]
Abstract
The meninges are a multilayered structure composed of fibroblasts, blood and lymphatic vessels, and immune cells. Meningeal fibroblasts secrete a variety of factors that control CNS development, yet strikingly little is known about their heterogeneity or development. Using single-cell sequencing, we report distinct transcriptional signatures for fibroblasts in the embryonic dura, arachnoid, and pia. We define new markers for meningeal layers and show conservation in human meninges. We find that embryonic meningeal fibroblasts are transcriptionally distinct between brain regions and identify a regionally localized pial subpopulation marked by the expression of μ-crystallin. Developmental analysis reveals a progressive, ventral-to-dorsal maturation of telencephalic meninges. Our studies have generated an unparalleled view of meningeal fibroblasts, providing molecular profiles of embryonic meningeal fibroblasts by layer and yielding insights into the mechanisms of meninges development and function.
Collapse
Affiliation(s)
- John DeSisto
- Department of Pediatrics Section of Hematology, Oncology, Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rebecca O'Rourke
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hannah E Jones
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bradley Pawlikowski
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alexandra D Malek
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie Bonney
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fabien Guimiot
- INSERM UMR 1141, Hôpital Robert Debré, 75019 Paris, France
| | - Kenneth L Jones
- Department of Pediatrics Section of Hematology, Oncology, Bone Marrow Transplant, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Julie A Siegenthaler
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
16
|
Economou AD, Monk NAM, Green JBA. Perturbation analysis of a multi-morphogen Turing reaction-diffusion stripe patterning system reveals key regulatory interactions. Development 2020; 147:dev190553. [PMID: 33033117 PMCID: PMC7648603 DOI: 10.1242/dev.190553] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/11/2020] [Indexed: 01/23/2023]
Abstract
Periodic patterning is widespread in development and can be modelled by reaction-diffusion (RD) processes. However, minimal two-component RD descriptions are vastly simpler than the multi-molecular events that actually occur and are often hard to relate to real interactions measured experimentally. Addressing these issues, we investigated the periodic striped patterning of the rugae (transverse ridges) in the mammalian oral palate, focusing on multiple previously implicated pathways: FGF, Hh, Wnt and BMP. For each, we experimentally identified spatial patterns of activity and distinct responses of the system to inhibition. Through numerical and analytical approaches, we were able to constrain substantially the number of network structures consistent with the data. Determination of the dynamics of pattern appearance further revealed its initiation by 'activators' FGF and Wnt, and 'inhibitor' Hh, whereas BMP and mesenchyme-specific-FGF signalling were incorporated once stripes were formed. This further limited the number of possible networks. Experimental constraint thus limited the number of possible minimal networks to 154, just 0.004% of the number of possible diffusion-driven instability networks. Together, these studies articulate the principles of multi-morphogen RD patterning and demonstrate the utility of perturbation analysis for constraining RD systems.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Andrew D Economou
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London, SE1 9RT, UK
| | - Nicholas A M Monk
- School of Mathematics and Statistics, University of Sheffield, Sheffield, S3 7RH, UK
| | - Jeremy B A Green
- Department of Craniofacial Development & Stem Cell Biology, King's College London, London, SE1 9RT, UK
| |
Collapse
|
17
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 379] [Impact Index Per Article: 94.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
18
|
Dias MS, Samson T, Rizk EB, Governale LS, Richtsmeier JT. Identifying the Misshapen Head: Craniosynostosis and Related Disorders. Pediatrics 2020; 146:peds.2020-015511. [PMID: 32868470 DOI: 10.1542/peds.2020-015511] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pediatric care providers, pediatricians, pediatric subspecialty physicians, and other health care providers should be able to recognize children with abnormal head shapes that occur as a result of both synostotic and deformational processes. The purpose of this clinical report is to review the characteristic head shape changes, as well as secondary craniofacial characteristics, that occur in the setting of the various primary craniosynostoses and deformations. As an introduction, the physiology and genetics of skull growth as well as the pathophysiology underlying craniosynostosis are reviewed. This is followed by a description of each type of primary craniosynostosis (metopic, unicoronal, bicoronal, sagittal, lambdoid, and frontosphenoidal) and their resultant head shape changes, with an emphasis on differentiating conditions that require surgical correction from those (bathrocephaly, deformational plagiocephaly/brachycephaly, and neonatal intensive care unit-associated skill deformation, known as NICUcephaly) that do not. The report ends with a brief discussion of microcephaly as it relates to craniosynostosis as well as fontanelle closure. The intent is to improve pediatric care providers' recognition and timely referral for craniosynostosis and their differentiation of synostotic from deformational and other nonoperative head shape changes.
Collapse
Affiliation(s)
- Mark S Dias
- Section of Pediatric Neurosurgery, Department of Neurosurgery and
| | - Thomas Samson
- Division of Plastic Surgery, Department of Surgery, College of Medicine and
| | - Elias B Rizk
- Section of Pediatric Neurosurgery, Department of Neurosurgery and
| | - Lance S Governale
- Lillian S. Wells Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, Florida
| | - Joan T Richtsmeier
- Department of Anthropology, College of the Liberal Arts and Huck Institutes of the Life Sciences, Pennsylvania State University, State College, Pennsylvania; and
| | | |
Collapse
|
19
|
Kim K, Gil M, Dayem AA, Choi S, Kang GH, Yang GM, Cho S, Jeong Y, Kim SJ, Seok J, Kwak HJ, Kumar Saha S, Kim A, Cho SG. Improved Isolation and Culture of Urine-Derived Stem Cells (USCs) and Enhanced Production of Immune Cells from the USC-Derived Induced Pluripotent Stem Cells. J Clin Med 2020; 9:E827. [PMID: 32197458 PMCID: PMC7141314 DOI: 10.3390/jcm9030827] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
The availability of autologous adult stem cells is one of the essential prerequisites for human stem cell therapy. Urine-derived stem cells (USCs) are considered as desirable cell sources for cell therapy because donor-specific USCs are easily and non-invasively obtained from urine. Efficient isolation, expansion, and differentiation methods of USCs are necessary to increase their availability. Here, we developed a method for efficient isolation and expansion of USCs using Matrigel, and the rho-associated protein kinase (ROCK) inhibitor, Y-27632. The prepared USCs showed significantly enhanced migration, colony forming capacity, and differentiation into osteogenic or chondrogenic lineage. The USCs were successfully reprogramed into induced pluripotent stem cells (USC-iPSCs) and further differentiated into kidney organoid and hematopoietic progenitor cells (HPCs). Using flavonoid molecules, the isolation efficiency of USCs and the production of HPCs from the USC-iPSCs was increased. Taken together, we present an improved isolation method of USCs utilizing Matrigel, a ROCK inhibitor and flavonoids, and enhanced differentiation of USC-iPSC to HPC by flavonoids. These novel findings could significantly enhance the use of USCs and USC-iPSCs for stem cell research and further application in regenerative stem cell-based therapies.
Collapse
Affiliation(s)
- Kyeongseok Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Minchan Gil
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Ahmed Abdal Dayem
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Sangbaek Choi
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Geun-Ho Kang
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Gwang-Mo Yang
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Sungha Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Yeojin Jeong
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Se Jong Kim
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Jaekwon Seok
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Hee Jeong Kwak
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Subbroto Kumar Saha
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| | - Aram Kim
- Department of Urology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05029, Korea;
| | - Ssang-Goo Cho
- Department of Stem Cell & Regenerative Biotechnology and Incurable Disease Animal Model and Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea; (K.K.); (M.G.); (A.A.D.); (S.C.); (G.-H.K.); (G.-M.Y.); (S.C.); (Y.J.); (S.J.K.); (J.S.); (H.J.K.); (S.K.S.)
| |
Collapse
|
20
|
Catala M, Khonsari RH, Paternoster G, Arnaud É. [Development and growth of the vault of the skull]. Neurochirurgie 2019; 65:210-215. [PMID: 31586575 DOI: 10.1016/j.neuchi.2019.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 02/03/2023]
Abstract
The vault of the skull is a region of the neurocranium formed by a process of membranous ossification. It consists of several bones: frontal bone, parietal bone, squamous part of the temporal bone, lamina ascendens of the sphenoid, and interparietal bone. The embryological origin of the bones of the skull vault is still the subject of controversy. This can be explained by the different animal models used for these purposes, but also by the various techniques applied to this problem. At all events, it seems that the cells of the neural crest generate some of the bones of the vault and that the others are derived from the mesoderm. This uncertainty should lead readers to be extremely cautious before using the presumptive maps published in the literature. Several tissues interact with osteo-progenitor cells: neural tube, surface ectoderm and dura mater. Analysis of genes in which mutations lead to abnormalities of the skull vault has partly revealed the molecular interactions. These are very complex and are the field of very numerous experimental investigations. In the relatively near future, we can hope to discover some of the molecular networks leading to the formation of these bony structures.
Collapse
Affiliation(s)
- M Catala
- UMR biologie du développement (Sorbonne université, CNRS, Inserm, IBPS), Sorbonne université (site Pierre-et-Marie-Curie), 9, quai Saint-Bernard, bâtiment C, 75252 Paris cedex 05, France.
| | - R H Khonsari
- Service de chirurgie maxillo-faciale et plastique, centre de référence maladies rares MAFACE, filière maladies rares CRANIOST, université Sorbonne Paris Cité, université Paris Descartes, hôpital Necker-Enfants-Malades, Assistance publique-Hôpitaux de Paris, Paris, France
| | - G Paternoster
- Service de neurochirurgie pédiatrique, hôpital Necker-Enfants-Malades, Assistance publique-Hôpitaux de Paris, Paris, France
| | - É Arnaud
- 34, avenue d'Eylau, Paris, France
| |
Collapse
|
21
|
Yilmaz E, Mihci E, Nur B, Alper ÖM, Taçoy Ş. Recent Advances in Craniosynostosis. Pediatr Neurol 2019; 99:7-15. [PMID: 31421914 DOI: 10.1016/j.pediatrneurol.2019.01.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 12/25/2018] [Accepted: 01/24/2019] [Indexed: 12/27/2022]
Abstract
Craniosynostosis is a pathologic craniofacial disorder and is defined as the premature fusion of one or more cranial (calvarial) sutures. Cranial sutures are fibrous joints consisting of nonossified mesenchymal cells that play an important role in the development of healthy craniofacial skeletons. Early fusion of these sutures results in incomplete brain development that may lead to complications of several severe medical conditions including seizures, brain damage, mental delay, complex deformities, strabismus, and visual and breathing problems. As a congenital disease, craniosynostosis has a heterogeneous origin that can be affected by genetic and epigenetic alterations, teratogens, and environmental factors and make the syndrome highly complex. To date, approximately 200 syndromes have been linked to craniosynostosis. In addition to being part of a syndrome, craniosynostosis can be nonsyndromic, formed without any additional anomalies. More than 50 nuclear genes that relate to craniosynostosis have been identified. Besides genetic factors, epigenetic factors like microRNAs and mechanical forces also play important roles in suture fusion. As craniosynostosis is a multifactorial disorder, evaluating the craniosynostosis syndrome requires and depends on all the information obtained from clinical findings, genetic analysis, epigenetic or environmental factors, or gene modulators. In this review, we will focus on embryologic and genetic studies, as well as epigenetic and environmental studies. We will discuss published studies and correlate the findings with unknown aspects of craniofacial disorders.
Collapse
Affiliation(s)
- Elanur Yilmaz
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Ercan Mihci
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Banu Nur
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| | - Özgül M Alper
- Department of Medical Biology and Genetics, Akdeniz University Medical School, Antalya, Turkey.
| | - Şükran Taçoy
- Department of Pediatric Genetics, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
22
|
Luo W, Yi Y, Jing D, Zhang S, Men Y, Ge WP, Zhao H. Investigation of Postnatal Craniofacial Bone Development with Tissue Clearing-Based Three-Dimensional Imaging. Stem Cells Dev 2019; 28:1310-1321. [PMID: 31392933 PMCID: PMC6767869 DOI: 10.1089/scd.2019.0104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
Traditional two-dimensional histological sections and microcomputed tomography remain to be the major tools for studying craniofacial bones despite the complicated spatial organization of craniofacial organs. Recently, our laboratory developed the Poly(Ethylene Glycol) Associated Solvent System (PEGASOS) tissue clearing method, which can efficiently render hard tissues, including bones and teeth fully transparent without losing endogenous fluorescent signals. Complete tissue transparency enables us to acquire three-dimensional (3D) images of craniofacial bone vasculature, osteogenesis utilizing various labeling strategies, thus to investigate the spatial relationship among different tissues during postnatal craniofacial development. We found out that during the early stage of postnatal development, craniofacial osteogenesis occurs throughout the entire craniofacial bones, including the periosteum, dura, bone marrow, and suture. After 3-4 weeks, craniofacial osteogenesis is gradually restricted to the suture region and remaining bone marrow space. Similarly, craniofacial bone vasculature gradually restricts to the suture region. Osteogenesis is spatially associated with vasculature during the entire postnatal development. Importantly, we demonstrated that in adult calvarial bones, Gli1+ mesenchymal stem cells were also spatially associated with the vasculature. These findings indicate that craniofacial bones share similar osteogenesis mechanism as the long bone despite their distinct osteogenic mechanisms. In addition, the PEGASOS tissue clearing method-based 3D imaging technique is a useful new tool for craniofacial research.
Collapse
Affiliation(s)
- Wenjing Luo
- Department of Biomedical Sciences, College of Dentistry, Texas A&M University, Dallas, Texas
| | - Yating Yi
- Department of Restorative Sciences, College of Dentistry, Texas A&M University, Dallas, Texas
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Dian Jing
- Department of Restorative Sciences, College of Dentistry, Texas A&M University, Dallas, Texas
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Yi Men
- Department of Restorative Sciences, College of Dentistry, Texas A&M University, Dallas, Texas
| | - Woo-Ping Ge
- Children's Research Institute, UT Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, Texas
| | - Hu Zhao
- Department of Restorative Sciences, College of Dentistry, Texas A&M University, Dallas, Texas
| |
Collapse
|
23
|
Lee C, Richtsmeier JT, Kraft RH. A coupled reaction-diffusion-strain model predicts cranial vault formation in development and disease. Biomech Model Mechanobiol 2019; 18:1197-1211. [PMID: 31006064 PMCID: PMC6625897 DOI: 10.1007/s10237-019-01139-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/18/2019] [Indexed: 01/16/2023]
Abstract
How cells utilize instructions provided by genes and integrate mechanical forces generated by tissue growth to produce morphology is a fundamental question of biology. Dermal bones of the vertebrate cranial vault are formed through the direct differentiation of mesenchymal cells on the neural surface into osteoblasts through intramembranous ossification. Here we join a self-organizing Turing mechanism, computational biomechanics, and experimental data to produce a 3D representative model of the growing cerebral surface, cranial vault bones, and sutures. We show how changes in single parameters regulating signaling during osteoblast differentiation and bone formation may explain cranial vault shape variation in craniofacial disorders. A key result is that toggling a parameter in our model results in closure of a cranial vault suture, an event that occurred during evolution of the cranial vault and that occurs in craniofacial disorders. Our approach provides an initial and important step toward integrating biomechanics into the genotype phenotype map to explain the production of variation in head morphology by developmental mechanisms.
Collapse
Affiliation(s)
- Chanyoung Lee
- Department of Mechanical Engineering, Pennsylvania State University, 341 Leonhard Building, University Park, PA, 16802, USA
| | - Joan T Richtsmeier
- Department of Anthropology, Pennsylvania State University, 409 Carpenter Building, University Park, PA, 16802, USA
| | - Reuben H Kraft
- Department of Mechanical and Nuclear Engineering, Pennsylvania State University, 320 Leonhard Building, University Park, PA, 16802, USA.
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA.
- Institute for Cyberscience, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
24
|
Tuzon CT, Rigueur D, Merrill AE. Nuclear Fibroblast Growth Factor Receptor Signaling in Skeletal Development and Disease. Curr Osteoporos Rep 2019; 17:138-146. [PMID: 30982184 PMCID: PMC8221190 DOI: 10.1007/s11914-019-00512-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Fibroblast growth factor receptor (FGFR) signaling regulates proliferation and differentiation during development and homeostasis. While membrane-bound FGFRs play a central role in these processes, the function of nuclear FGFRs is also critical. Here, we highlight mechanisms for nuclear FGFR translocation and the effects of nuclear FGFRs on skeletal development and disease. RECENT FINDINGS Full-length FGFRs, internalized by endocytosis, enter the nucleus through β-importin-dependent mechanisms that recognize the nuclear localization signal within FGFs. Alternatively, soluble FGFR intracellular fragments undergo nuclear translocation following their proteolytic release from the membrane. FGFRs enter the nucleus during the cellular transition between proliferation and differentiation. Once nuclear, FGFRs interact with chromatin remodelers to alter the epigenetic state and transcription of their target genes. Dysregulation of nuclear FGFR is linked to the etiology of congenital skeletal disorders and neoplastic transformation. Revealing the activities of nuclear FGFR will advance our understanding of 20 congenital skeletal disorders caused by FGFR mutations, as well as FGFR-related cancers.
Collapse
Affiliation(s)
- Creighton T Tuzon
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Diana Rigueur
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA
| | - Amy E Merrill
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
25
|
Abstract
Deviations from the precisely coordinated programme of human head development can lead to craniofacial and orofacial malformations often including a variety of dental abnormalities too. Although the aetiology is still unknown in many cases, during the last decades different intracellular signalling pathways have been genetically linked to specific disorders. Among these pathways, the RAS/extracellular signal-regulated kinase (ERK) signalling cascade is the focus of this review since it encompasses a large group of genes that when mutated cause some of the most common and severe developmental anomalies in humans. We present the components of the RAS/ERK pathway implicated in craniofacial and orodental disorders through a series of human and animal studies. We attempt to unravel the specific molecular targets downstream of ERK that act on particular cell types and regulate key steps in the associated developmental processes. Finally we point to ambiguities in our current knowledge that need to be clarified before RAS/ERK-targeting therapeutic approaches can be implemented.
Collapse
|
26
|
Aizawa R, Yamada A, Seki T, Tanaka J, Nagahama R, Ikehata M, Kato T, Sakashita A, Ogata H, Chikazu D, Maki K, Mishima K, Yamamoto M, Kamijo R. Cdc42 regulates cranial suture morphogenesis and ossification. Biochem Biophys Res Commun 2019; 512:145-149. [PMID: 30853186 DOI: 10.1016/j.bbrc.2019.02.106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022]
Abstract
Cdc42 (cell division cycle 42) is ubiquitously expressed small GTPases belonging to the Rho family of proteins. Previously, we generated limb bud mesenchyme-specific Cdc42 inactivated mice (Cdc42 conditional knockout mice; Cdc42 fl/fl; Prx1-Cre), which showed short limbs and cranial bone deformities, though the mechanism related to the cranium phenotype was unclear. In the present study, we investigated the role of Cdc42 in cranial bone development. Our results showed that loss of Cdc42 caused a defect of intramembranous ossification in cranial bone tissues which is related to decreased expressions of cranial suture morphogenesis genes, including Indian hedgehog (Ihh) and bone morphogenetic proteins (BMPs). These findings demonstrate that Cdc42 plays a crucial role in cranial osteogenesis, and is controlled by Ihh- and BMP-mediated signaling during cranium development.
Collapse
Affiliation(s)
- Ryo Aizawa
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan; Department of Periodontology, School of Dentistry, Showa University, Ohta, Tokyo, Japan
| | - Atsushi Yamada
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan.
| | - Tatsuaki Seki
- Department of Periodontology, School of Dentistry, Showa University, Ohta, Tokyo, Japan
| | - Junichi Tanaka
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan
| | - Ryo Nagahama
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan; Department of Orthodontics, School of Dentistry, Showa University, Ohta, Tokyo, Japan
| | - Mikiko Ikehata
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan; Department of Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, Japan
| | - Tadashi Kato
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan; Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Akiko Sakashita
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Hiroaki Ogata
- Department of Internal Medicine, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Daichi Chikazu
- Department of Oral and Maxillofacial Surgery, Tokyo Medical University, Tokyo, Japan
| | - Koutaro Maki
- Department of Orthodontics, School of Dentistry, Showa University, Ohta, Tokyo, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan
| | - Matsuo Yamamoto
- Department of Periodontology, School of Dentistry, Showa University, Ohta, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, School of Dentistry, Showa University, Shinagawa, Tokyo, Japan
| |
Collapse
|
27
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Pierre J Marie
- UMR-1132 Inserm (Institut national de la Santé et de la Recherche Médicale) and University Paris Diderot, Sorbonne Paris Cité, Hôpital Lariboisière, Paris, France
| |
Collapse
|
28
|
Ferguson J, Atit RP. A tale of two cities: The genetic mechanisms governing calvarial bone development. Genesis 2019; 57:e23248. [PMID: 30155972 PMCID: PMC7433025 DOI: 10.1002/dvg.23248] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 12/25/2022]
Abstract
The skull bones must grow in a coordinated, three-dimensional manner to coalesce and form the head and face. Mammalian skull bones have a dual embryonic origin from cranial neural crest cells (CNCC) and paraxial mesoderm (PM) and ossify through intramembranous ossification. The calvarial bones, the bones of the cranium which cover the brain, are derived from the supraorbital arch (SOA) region mesenchyme. The SOA is the site of frontal and parietal bone morphogenesis and primary center of ossification. The objective of this review is to frame our current in vivo understanding of the morphogenesis of the calvarial bones and the gene networks regulating calvarial bone initiation in the SOA mesenchyme.
Collapse
Affiliation(s)
- James Ferguson
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106
- Department of Genetics, Case Western Reserve University, Cleveland OH 44106
- Department of Dermatology, Case Western Reserve University, Cleveland OH 44106
| | - Radhika P. Atit
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106
- Department of Genetics, Case Western Reserve University, Cleveland OH 44106
- Department of Dermatology, Case Western Reserve University, Cleveland OH 44106
| |
Collapse
|
29
|
Camp E, Pribadi C, Anderson PJ, Zannettino AC, Gronthos S. miRNA-376c-3p Mediates TWIST-1 Inhibition of Bone Marrow-Derived Stromal Cell Osteogenesis and Can Reduce Aberrant Bone Formation of TWIST-1 Haploinsufficient Calvarial Cells. Stem Cells Dev 2018; 27:1621-1633. [DOI: 10.1089/scd.2018.0083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Esther Camp
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Clara Pribadi
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Peter J. Anderson
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
- Australian Craniofacial Unit, Faculty of Health and Medical Sciences, Adelaide Medical School and Dentistry, Women's and Children's Hospital, The University of Adelaide, Adelaide, Australia
| | - Andrew C.W. Zannettino
- South Australian Health and Medical Research Institute, Adelaide, Australia
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
30
|
Kumar S, Franz-Odendaal TA. Analysis of the FGFR spatiotemporal expression pattern within the chicken scleral ossicle system. Gene Expr Patterns 2018; 30:7-13. [DOI: 10.1016/j.gep.2018.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 12/26/2022]
|
31
|
Fgfr1 conditional-knockout in neural crest cells induces heterotopic chondrogenesis and osteogenesis in mouse frontal bones. Med Mol Morphol 2018; 52:156-163. [PMID: 30499042 DOI: 10.1007/s00795-018-0213-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Abstract
Most facial bones, including frontal bones, are derived from neural crest cells through intramembranous ossification. Fibroblast growth factor receptor 1 (Fgfr1) plays a pivotal role in craniofacial bone development, and loss of Fgfr1 leads to cleft palate and facial cleft defects in newborn mice. However, the potential role of the Fgfr1 gene in neural crest cell-mediated craniofacial development remains unclear. To investigate the role of Fgfr1 in neural crest cells, we analyzed Wnt1-Cre;Fgfr1flox/flox mice. Our results show that specific knockout of Fgfr1 in neural crest cells induced heterotopic chondrogenesis and osteogenesis at the interface of the anterior portions of frontal bones. We observed that heterotopic bone formation continued through postnatal day 28, whereas heterotopic chondrogenesis lasted only through the embryonic period. In summary, our results indicate that loss of Fgfr1 in neural crest cells leads to heterotopic chondrogenesis and osteogenesis.
Collapse
|
32
|
Teng CS, Ting MC, Farmer DT, Brockop M, Maxson RE, Crump JG. Altered bone growth dynamics prefigure craniosynostosis in a zebrafish model of Saethre-Chotzen syndrome. eLife 2018; 7:37024. [PMID: 30375332 PMCID: PMC6207424 DOI: 10.7554/elife.37024] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/14/2018] [Indexed: 01/09/2023] Open
Abstract
Cranial sutures separate the skull bones and house stem cells for bone growth and repair. In Saethre-Chotzen syndrome, mutations in TCF12 or TWIST1 ablate a specific suture, the coronal. This suture forms at a neural-crest/mesoderm interface in mammals and a mesoderm/mesoderm interface in zebrafish. Despite this difference, we show that combinatorial loss of TCF12 and TWIST1 homologs in zebrafish also results in specific loss of the coronal suture. Sequential bone staining reveals an initial, directional acceleration of bone production in the mutant skull, with subsequent localized stalling of bone growth prefiguring coronal suture loss. Mouse genetics further reveal requirements for Twist1 and Tcf12 in both the frontal and parietal bones for suture patency, and to maintain putative progenitors in the coronal region. These findings reveal conservation of coronal suture formation despite evolutionary shifts in embryonic origins, and suggest that the coronal suture might be especially susceptible to imbalances in progenitor maintenance and osteoblast differentiation. Some of the most common birth defects involve improper development of the head and face. One such birth defect is called craniosynostosis. Normally, an infant’s skull bones are not fully fused together. Instead, they are held together by soft tissue that allows the baby’s skull to more easily pass through the birth canal. This tissue also houses specialized cells called stem cells that allow the brain and skull to grow with the child. But in craniosynostosis these stem cells behave abnormally, which fuses the skull bones together and prevents the skull and brain from growing properly during childhood. One form of craniosynostosis called Saethre-Chotzen syndrome is caused by mutations in one of two genes that ensure the proper separation of two bones in the roof of the skull. Mice with mutations in the mouse versions of these genes develop the same problem and are used to study this condition. Mouse studies have looked mostly at what happens after birth. Studies looking at what happens in embryos with these mutations could help scientists learn more. One way to do so would be to genetically engineer zebrafish with the equivalent mutations. This is because zebrafish embryos are transparent and grow outside their mother’s body, making it easier for scientists to watch them develop. Now, Teng et al. have grown zebrafish with mutations in the zebrafish versions of the genes that cause Saethre-Chotzen syndrome. In the experiments, imaging tools were used to observe the live fish as they developed. This showed that the stem cells in their skulls become abnormal much earlier than previous studies had suggested. Teng et al. also showed that similar stem cells are responsible for growth of the skull in zebrafish and mice. Babies with craniosynostosis often need multiple, risky surgeries to separate their skull bones and allow their brain and head to grow. Unfortunately, these bones often fuse again because they have abnormal stem cells. Teng et al. provide new information on what goes wrong in these stem cells. Hopefully, this new information will help scientists to one day correct the defective stem cells in babies with craniosynostosis, thus reducing the number of surgeries needed to correct the problem.
Collapse
Affiliation(s)
- Camilla S Teng
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States.,Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - Man-Chun Ting
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - D'Juan T Farmer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States
| | - Mia Brockop
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - Robert E Maxson
- Department of Biochemistry, Keck School of Medicine, University of Southern California, Los Angeles, United States
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, United States
| |
Collapse
|
33
|
Quarto N, Shailendra S, Meyer NP, Menon S, Renda A, Longaker MT. Twist1-Haploinsufficiency Selectively Enhances the Osteoskeletal Capacity of Mesoderm-Derived Parietal Bone Through Downregulation of Fgf23. Front Physiol 2018; 9:1426. [PMID: 30374308 PMCID: PMC6196243 DOI: 10.3389/fphys.2018.01426] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/19/2018] [Indexed: 02/05/2023] Open
Abstract
Craniofacial development is a program exquisitely orchestrated by tissue contributions and regulation of genes expression. The basic helix–loop–helix (bHLH) transcription factor Twist1 expressed in the skeletal mesenchyme is a key regulator of craniofacial development playing an important role during osteoskeletogenesis. This study investigates the postnatal impact of Twist1 haploinsufficiency on the osteoskeletal ability and regeneration on two calvarial bones arising from tissues of different embryonic origin: the neural crest-derived frontal and the mesoderm-derived parietal bones. We show that Twist1 haplonsufficiency as well Twist1-sh-mediated silencing selectively enhanced osteogenic and tissue regeneration ability of mesoderm-derived bones. Transcriptomic profiling, gain-and loss-of-function experiments revealed that Twist1 haplonsufficiency triggers its selective activity on mesoderm-derived bone through a sharp downregulation of the bone-derived hormone Fgf23 that is upregulated exclusively in wild-type parietal bone.
Collapse
Affiliation(s)
- Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States.,Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Siny Shailendra
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Nathaniel P Meyer
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Siddharth Menon
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| | - Andrea Renda
- Dipartimento di Scienze Biomediche Avanzate, Universita' degli Studi di Napoli Federico II, Naples, Italy
| | - Michael T Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
34
|
Kawane T, Qin X, Jiang Q, Miyazaki T, Komori H, Yoshida CA, Matsuura-Kawata VKDS, Sakane C, Matsuo Y, Nagai K, Maeno T, Date Y, Nishimura R, Komori T. Runx2 is required for the proliferation of osteoblast progenitors and induces proliferation by regulating Fgfr2 and Fgfr3. Sci Rep 2018; 8:13551. [PMID: 30202094 PMCID: PMC6131145 DOI: 10.1038/s41598-018-31853-0] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/28/2018] [Indexed: 01/18/2023] Open
Abstract
Runx2 and Sp7 are essential transcription factors for osteoblast differentiation. However, the molecular mechanisms responsible for the proliferation of osteoblast progenitors remain unclear. The early onset of Runx2 expression caused limb defects through the Fgfr1–3 regulation by Runx2. To investigate the physiological role of Runx2 in the regulation of Fgfr1–3, we compared osteoblast progenitors in Sp7−/− and Runx2−/− mice. Osteoblast progenitors accumulated and actively proliferated in calvariae and mandibles of Sp7−/− but not of Runx2−/− mice, and the number of osteoblast progenitors and their proliferation were dependent on the gene dosage of Runx2 in Sp7−/− background. The expression of Fgfr2 and Fgfr3, which were responsible for the proliferation of osteoblast progenitors, was severely reduced in Runx2−/− but not in Sp7−/− calvariae. Runx2 directly regulated Fgfr2 and Fgfr3, increased the proliferation of osteoblast progenitors, and augmented the FGF2-induced proliferation. The proliferation of Sp7−/− osteoblast progenitors was enhanced and strongly augmented by FGF2, and Runx2 knockdown reduced the FGF2-induced proliferation. Fgfr inhibitor AZD4547 abrogated all of the enhanced proliferation. These results indicate that Runx2 is required for the proliferation of osteoblast progenitors and induces proliferation, at least partly, by regulating Fgfr2 and Fgfr3 expression.
Collapse
Affiliation(s)
- Tetsuya Kawane
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Xin Qin
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Qing Jiang
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Toshihiro Miyazaki
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Hisato Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Carolina Andrea Yoshida
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | | | - Chiharu Sakane
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Yuki Matsuo
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Kazuhiro Nagai
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, 852-8501, Japan
| | - Takafumi Maeno
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, 545-8585, Japan
| | - Yuki Date
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.,Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, 565-0871, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan. .,Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8588, Japan.
| |
Collapse
|
35
|
Cesario JM, Landin Malt A, Chung JU, Khairallah MP, Dasgupta K, Asam K, Deacon LJ, Choi V, Almaidhan AA, Darwiche NA, Kim J, Johnson RL, Jeong J. Anti-osteogenic function of a LIM-homeodomain transcription factor LMX1B is essential to early patterning of the calvaria. Dev Biol 2018; 443:103-116. [PMID: 29852132 DOI: 10.1016/j.ydbio.2018.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/15/2018] [Accepted: 05/26/2018] [Indexed: 12/22/2022]
Abstract
The calvaria (upper part of the skull) is made of plates of bone and fibrous joints (sutures and fontanelles), and the proper balance and organization of these components are crucial to normal development of the calvaria. In a mouse embryo, the calvaria develops from a layer of head mesenchyme that surrounds the brain from shortly after mid-gestation. The mesenchyme just above the eye (supra-orbital mesenchyme, SOM) generates ossification centers for the bones, which then grow toward the apex gradually. In contrast, the mesenchyme apical to SOM (early migrating mesenchyme, EMM), including the area at the vertex, does not generate an ossification center. As a result, the dorsal midline of the head is occupied by sutures and fontanelles at birth. To date, the molecular basis for this regional difference in developmental programs is unknown. The current study provides vital insights into the genetic regulation of calvarial patterning. First, we showed that osteogenic signals were active in both EMM and SOM during normal development, which suggested the presence of an anti-osteogenic factor in EMM to counter the effect of these signals. Subsequently, we identified Lmx1b as an anti-osteogenic gene that was expressed in EMM but not in SOM. Furthermore, head mesenchyme-specific deletion of Lmx1b resulted in heterotopic ossification from EMM at the vertex, and craniosynostosis affecting multiple sutures. Conversely, forced expression of Lmx1b in SOM was sufficient to inhibit osteogenic specification. Therefore, we conclude that Lmx1b plays a key role as an anti-osteogenic factor in patterning the head mesenchyme into areas with different osteogenic competence. In turn, this patterning event is crucial to generating the proper organization of the bones and soft tissue joints of the calvaria.
Collapse
Affiliation(s)
- Jeffry M Cesario
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - André Landin Malt
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Jong Uk Chung
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Michael P Khairallah
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Krishnakali Dasgupta
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Kesava Asam
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Lindsay J Deacon
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Veronica Choi
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Asma A Almaidhan
- Department of Orthodontics, New York University College of Dentistry, New York, NY, United States
| | - Nadine A Darwiche
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Jimin Kim
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States
| | - Randy L Johnson
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, United States.
| |
Collapse
|
36
|
Camp E, Anderson PJ, Zannettino ACW, Glackin CA, Gronthos S. Tyrosine kinase receptor c‐ros‐oncogene 1 inhibition alleviates aberrant bone formation of TWIST‐1 haploinsufficient calvarial cells from Saethre–Chotzen syndrome patients. J Cell Physiol 2018; 233:7320-7332. [DOI: 10.1002/jcp.26563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/23/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Esther Camp
- Mesenchymal Stem Cell LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Peter J. Anderson
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Australian Craniofacial UnitWomen's and Children's HospitalNorth AdelaideSouth AustraliaAustralia
| | - Andrew C. W. Zannettino
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Myeloma Research LaboratoryAdelaide Medical School, Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Carlotta A. Glackin
- Molecular Medicine and NeurosciencesCity of Hope National Medical Center and Beckman Research InstituteDuarteCalifornia
| | - Stan Gronthos
- Mesenchymal Stem Cell LaboratoryAdelaide Medical SchoolFaculty of Health and Medical SciencesUniversity of AdelaideAdelaideSouth AustraliaAustralia
- Cancer ThemeSouth Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| |
Collapse
|
37
|
Ledwon JK, Turin SY, Gosain AK, Topczewska JM. The expression of fgfr3 in the zebrafish head. Gene Expr Patterns 2018; 29:32-38. [PMID: 29630949 DOI: 10.1016/j.gep.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/08/2018] [Accepted: 04/03/2018] [Indexed: 12/29/2022]
Abstract
Fibroblast growth factor (FGF) signaling is essential for many developmental processes and plays a pivotal role in skeletal homeostasis, regeneration and wound healing. FGF signals through one of five tyrosine kinase receptors: Fgfr1a, -1b, -2, -3, -4. To characterize the expression of zebrafish fgfr3 from the larval stage to adulthood, we used RNAscope in situ hybridization on paraffin sections of the zebrafish head. Our study revealed spatial and temporal distribution of fgfr3 transcript in chondrocytes of the head cartilages, osteoblasts involved in bone formation, ventricular zone of the brain, undifferentiated mesenchymal cells of the skin, and lens epithelium of the eye. In general, the expression pattern of zebrafish fgfr3 is similar to the expression observed in higher vertebrates.
Collapse
Affiliation(s)
- Joanna K Ledwon
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Sergey Y Turin
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Arun K Gosain
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA
| | - Jolanta M Topczewska
- Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Division of Plastic and Reconstructive Surgery, Stanley Manne Children's Research Institute, Chicago, IL, USA.
| |
Collapse
|
38
|
Tang H, Massi D, Hemmings BA, Mandalà M, Hu Z, Wicki A, Xue G. AKT-ions with a TWIST between EMT and MET. Oncotarget 2018; 7:62767-62777. [PMID: 27623213 PMCID: PMC5308764 DOI: 10.18632/oncotarget.11232] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 07/28/2016] [Indexed: 02/07/2023] Open
Abstract
The transcription factor Twist is an important regulator of cranial suture during embryogenesis. Closure of the neural tube is achieved via Twist-triggered cellular transition from an epithelial to mesenchymal phenotype, a process known as epithelial-mesenchymal transition (EMT), characterized by a remarkable increase in cell motility. In the absence of Twist activity, EMT and associated phenotypic changes in cell morphology and motility can also be induced, albeit moderately, by other transcription factor families, including Snail and Zeb. Aberrant EMT triggered by Twist in human mammary tumour cells was first reported to drive metastasis to the lung in a metastatic breast cancer model. Subsequent analysis of many types of carcinoma demonstrated overexpression of these unique EMT transcription factors, which statistically correlated with worse outcome, indicating their potential as biomarkers in the clinic. However, the mechanisms underlying their activation remain unclear. Interestingly, increasing evidence indicates they are selectively activated by distinct intracellular kinases, thereby acting as downstream effectors facilitating transduction of cytoplasmic signals into nucleus and reprogramming EMT and mesenchymal-epithelial transition (MET) transcription to control cell plasticity. Understanding these relationships and emerging data indicating differential phosphorylation of Twist leads to complex and even paradoxical functionalities, will be vital to unlocking their potential in clinical settings.
Collapse
Affiliation(s)
- Huifang Tang
- Department of Pharmacology, Zhejiang University School of Basic Medical Sciences, Hangzhou, China
| | - Daniela Massi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Brian A Hemmings
- Department of Mechanisms of Cancer, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Mario Mandalà
- Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Zhengqiang Hu
- Department of Pharmacology, Zhejiang University School of Basic Medical Sciences, Hangzhou, China
| | - Andreas Wicki
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Gongda Xue
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
39
|
Järvinen E, Shimomura-Kuroki J, Balic A, Jussila M, Thesleff I. Mesenchymal Wnt/β-catenin signaling limits tooth number. Development 2018; 145:dev.158048. [PMID: 29437780 DOI: 10.1242/dev.158048] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/21/2018] [Indexed: 12/29/2022]
Abstract
Tooth agenesis is one of the predominant developmental anomalies in humans, usually affecting the permanent dentition generated by sequential tooth formation and, in most cases, caused by mutations perturbing epithelial Wnt/β-catenin signaling. In addition, loss-of-function mutations in the Wnt feedback inhibitor AXIN2 lead to human tooth agenesis. We have investigated the functions of Wnt/β-catenin signaling during sequential formation of molar teeth using mouse models. Continuous initiation of new teeth, which is observed after genetic activation of Wnt/β-catenin signaling in the oral epithelium, was accompanied by enhanced expression of Wnt antagonists and a downregulation of Wnt/β-catenin signaling in the dental mesenchyme. Genetic and pharmacological activation of mesenchymal Wnt/β-catenin signaling negatively regulated sequential tooth formation, an effect partly mediated by Bmp4. Runx2, a gene whose loss-of-function mutations result in sequential formation of supernumerary teeth in the human cleidocranial dysplasia syndrome, suppressed the expression of Wnt inhibitors Axin2 and Drapc1 in dental mesenchyme. Our data indicate that increased mesenchymal Wnt signaling inhibits the sequential formation of teeth, and suggest that Axin2/Runx2 antagonistic interactions modulate the level of mesenchymal Wnt/β-catenin signaling, underlying the contrasting dental phenotypes caused by human AXIN2 and RUNX2 mutations.
Collapse
Affiliation(s)
- Elina Järvinen
- Institute of Biotechnology, University of Helsinki, Helsinki 007100, Finland.,Merck Oy, Espoo 02150, Finland
| | - Junko Shimomura-Kuroki
- Institute of Biotechnology, University of Helsinki, Helsinki 007100, Finland.,Department of Pediatric Dentistry, The Nippon Dental University, School of Life Dentistry at Niigata, Niigata 951-8580, Japan
| | - Anamaria Balic
- Institute of Biotechnology, University of Helsinki, Helsinki 007100, Finland
| | - Maria Jussila
- Institute of Biotechnology, University of Helsinki, Helsinki 007100, Finland
| | - Irma Thesleff
- Institute of Biotechnology, University of Helsinki, Helsinki 007100, Finland
| |
Collapse
|
40
|
Veistinen LK, Mustonen T, Hasan MR, Takatalo M, Kobayashi Y, Kesper DA, Vortkamp A, Rice DP. Regulation of Calvarial Osteogenesis by Concomitant De-repression of GLI3 and Activation of IHH Targets. Front Physiol 2017; 8:1036. [PMID: 29311969 PMCID: PMC5742257 DOI: 10.3389/fphys.2017.01036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/29/2017] [Indexed: 12/24/2022] Open
Abstract
Loss-of-function mutations in GLI3 and IHH cause craniosynostosis and reduced osteogenesis, respectively. In this study, we show that Ihh ligand, the receptor Ptch1 and Gli transcription factors are differentially expressed in embryonic mouse calvaria osteogenic condensations. We show that in both Ihh-/- and Gli3Xt-J/Xt-J embryonic mice, the normal gene expression architecture is lost and this results in disorganized calvarial bone development. RUNX2 is a master regulatory transcription factor controlling osteogenesis. In the absence of Gli3, RUNX2 isoform II and IHH are upregulated, and RUNX2 isoform I downregulated. This is consistent with the expanded and aberrant osteogenesis observed in Gli3Xt-J/Xt-J mice, and consistent with Runx2-I expression by relatively immature osteoprogenitors. Ihh-/- mice exhibited small calvarial bones and HH target genes, Ptch1 and Gli1, were absent. This indicates that IHH is the functional HH ligand, and that it is not compensated by another HH ligand. To decipher the roles and potential interaction of Gli3 and Ihh, we generated Ihh-/-;Gli3Xt-J/Xt-J compound mutant mice. Even in the absence of Ihh, Gli3 deletion was sufficient to induce aberrant precocious ossification across the developing suture, indicating that the craniosynostosis phenotype of Gli3Xt-J/Xt-J mice is not dependent on IHH ligand. Also, we found that Ihh was not required for Runx2 expression as the expression of RUNX2 target genes was unaffected by deletion of Ihh. To test whether RUNX2 has a role upstream of IHH, we performed RUNX2 siRNA knock down experiments in WT calvarial osteoblasts and explants and found that Ihh expression is suppressed. Our results show that IHH is the functional HH ligand in the embryonic mouse calvaria osteogenic condensations, where it regulates the progression of osteoblastic differentiation. As GLI3 represses the expression of Runx2-II and Ihh, and also elevates the Runx2-I expression, and as IHH may be regulated by RUNX2 these results raise the possibility of a regulatory feedback circuit to control calvarial osteogenesis and suture patency. Taken together, RUNX2-controlled osteoblastic cell fate is regulated by IHH through concomitant inhibition of GLI3-repressor formation and activation of downstream targets.
Collapse
Affiliation(s)
- Lotta K Veistinen
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Tuija Mustonen
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Minerva Research Institute, Helsinki, Finland
| | - Md Rakibul Hasan
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Yukiho Kobayashi
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Orthodontics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Dörthe A Kesper
- Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - Andrea Vortkamp
- Center of Medical Biotechnology, University of Duisburg-Essen, Essen, Germany
| | - David P Rice
- Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Orthodontics, Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
41
|
Freitas ECLBD, Nascimento SRD, de Mello MP, Gil-da-Silva-Lopes VL. Q289P Mutation in FGFR2 Gene Causes Saethre-Chotzen Syndrome: Some Considerations About Familial Heterogeneity. Cleft Palate Craniofac J 2017; 43:142-7. [PMID: 16526917 DOI: 10.1597/04-155.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
ObjectiveTo describe the first report on a three-generation family presenting typical features of Saethre-Chotzen syndrome, in which the Q289P mutation in the FGFR2 gene was detected.DesignDysmorphological evaluation was performed by a clinical geneticist. Direct sequencing of the polymerase chain reaction-amplified coding region of TWIST and screening for the P250R mutation in the FGFR3 gene were performed. Exons IIIa and IIIc of FGFR2 were sequenced also. The mutation was confirmed by both restriction-enzyme digestion and allelic-specific polymerase chain reaction.ResultsNeither TWIST gene analysis nor analysis of the P250R mutation on gene FGFR3 showed mutation within the coding sequence. A nucleotide change from CAG to CCG in exon IIIa of the FGFR2 gene that caused a Q289P mutation was detected, although exon IIIc in the propositus was normal. These same results were detected in his mother, but no other members of the kindred presented clinical features consistent with Saethre-Chotzen syndrome.ConclusionsThis mutation was previously reported in individuals with Crouzon and Jackson-Weiss syndromes. The FGFR2 mutation in the family with Saethre-Chotzen syndrome herein reported reinforces the idea of an interaction among TWIST and FGFR genes during development. Absence of the Q289P mutation in some affected individuals in this family is discussed.
Collapse
|
42
|
Yang J, Liu Z, Liu H, He J, Yang J, Lin P, Wang Q, Du J, Ma W, Yin Z, Davis E, Orlowski RZ, Hou J, Yi Q. C-reactive protein promotes bone destruction in human myeloma through the CD32-p38 MAPK-Twist axis. Sci Signal 2017; 10:10/509/eaan6282. [PMID: 29233917 DOI: 10.1126/scisignal.aan6282] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bone destruction is a hallmark of myeloma and affects 80% of patients. Myeloma cells promote bone destruction by activating osteoclasts. In investigating the underlying mechanism, we found that C-reactive protein (CRP), a protein secreted in increased amounts by hepatocytes in response to myeloma-derived cytokines, activated myeloma cells to promote osteoclastogenesis and bone destruction in vivo. In mice bearing human bone grafts and injected with multiple myeloma cells, CRP bound to surface CD32 (also known as FcγRII) on myeloma cells, which activated a pathway mediated by the kinase p38 MAPK and the transcription factor Twist that enhanced the cells' secretion of osteolytic cytokines. Furthermore, analysis of clinical samples from newly diagnosed myeloma patients revealed a positive correlation between the amount of serum CRP and the number of osteolytic bone lesions. These findings establish a mechanism by which myeloma cells are activated to promote bone destruction and suggest that CRP may be targeted to prevent or treat myeloma-associated bone disease in patients.
Collapse
Affiliation(s)
- Jing Yang
- Guangzhou Key Laboratory of Translational Medicine on Malignant Tumor Treatment, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China. .,Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhiqiang Liu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Physiology and Pathology, Tianjin Medical University, Tianjin 300070, China
| | - Huan Liu
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jin He
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianling Yang
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pei Lin
- Department of Hematopathology, Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Qiang Wang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Juan Du
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, Shanghai 200085, China
| | - Wencai Ma
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Eric Davis
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and the Center for Cancer Immunology Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Hou
- Department of Hematology, The Myeloma and Lymphoma Center, Changzheng Hospital, The Second Military Medical University, Shanghai 200085, China
| | - Qing Yi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Nguyen D, Yamada R, Yoshimitsu N, Oguri A, Kojima T, Takahashi N. Involvement of the Mab21l1 gene in calvarial osteogenesis. Differentiation 2017; 98:70-78. [PMID: 29156428 DOI: 10.1016/j.diff.2017.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 10/19/2017] [Accepted: 11/08/2017] [Indexed: 01/09/2023]
Abstract
The Mab-21 gene family is crucial for animal development. A deficiency in the Mab-21 genes associates with several defects, including skeletal malformation in mice and humans. In this study, we observed that mice lacking Mab21l1 displayed an unclosed fontanelle, suggesting impaired calvarial bone development. Cells isolated from the calvaria of these mice showed a greater osteoblast differentiation potential as evidenced by the abundance of mineralized bone nodules and higher expression levels of osteogenic markers than wild-type cells. Mab21l1-/- osteoblasts also expressed higher levels of adipocyte genes and interferon-regulated genes at early stages of osteogenesis. Rankl/Opg expression levels were also higher in Mab21l1-/- osteoblasts than in wild-type cells. These data suggest that Mab21l1 is involved in either the regulation of mesenchymal cell proliferation and differentiation or the balance between bone formation and resorption. An alteration in these regulatory machineries, therefore, may lead to insufficient bone formation, causing the bone phenotype in Mab21l1-/- mice.
Collapse
Affiliation(s)
- Dan Nguyen
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ryuichi Yamada
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan; RNA Company Limited, 7-25-7, Nishikamata, Ota-ku, Tokyo 114-8661, Japan
| | - Nodoka Yoshimitsu
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Akira Oguri
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takuya Kojima
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan; RNA Company Limited, 7-25-7, Nishikamata, Ota-ku, Tokyo 114-8661, Japan
| | - Naoki Takahashi
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
44
|
Abstract
Crouzon syndrome is an autosomal-dominant congenital disease due to a mutation in the fibroblast growth factor receptor 2 protein. The purpose of this study is to evaluate wound-healing potential of Crouzon osteoblasts and adipose-derived stem cells (ADSCs) in a murine model. Parietal skull defects were created in Crouzon and mature wild-type (WT) CD-1 mice. One group of WT and Crouzon mice were left untreated. Another group was transplanted with both WT and Crouzon adipose-derived stem cells. Additional groups compared the use of a fibrin glue scaffold and periosteum removal. Skulls were harvested from each group and evaluated histologically at 8-week and/or 16-week periods. Mean areas of defect were quantified and compared via ANOVA F-test. The average area of defect after 8 and 16 weeks in untreated Crouzon mice was 15.37 ± 1.08 cm and 16.69 ± 1.51 cm, respectively. The average area of the defect in untreated WT mice after 8 and 16 weeks averaged 14.17 ± 1.88 cm and 14.96 ± 2.26 cm, respectively. WT mice with autologous ADSCs yielded an average area of 15.35 ± 1.34 cm after 16 weeks while Crouzon mice with WT ADSCs healed to an average size of 12.98 ± 1.89 cm. Crouzon ADSCs transplanted into WT mice yielded an average area of 15.47 ± 1.29 cm while autologous Crouzon ADSCs yielded an area of 14.22 ± 3.32 cm. ANOVA F-test yielded P = .415. The fibroblast growth factor receptor 2 mutation in Crouzon syndrome does not promote reossification of critical-sized defects in mature WT and Crouzon mice. Furthermore, Crouzon ADSCs do not possess osteogenic advantage over WT ADSCs.
Collapse
|
45
|
Murphy MP, Quarto N, Longaker MT, Wan DC. * Calvarial Defects: Cell-Based Reconstructive Strategies in the Murine Model. Tissue Eng Part C Methods 2017; 23:971-981. [PMID: 28825366 DOI: 10.1089/ten.tec.2017.0230] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Calvarial defects pose a continued clinical dilemma for reconstruction. Advancements within the fields of stem cell biology and tissue engineering have enabled researchers to develop reconstructive strategies using animal models. We review the utility of various animal models and focus on the mouse, which has aided investigators in understanding cranial development and calvarial bone healing. The murine model has also been used to study regenerative approaches to critical-sized calvarial defects, and we discuss the application of stem cells such as bone marrow-derived mesenchymal stromal cells, adipose-derived stromal cells, muscle-derived stem cells, and pluripotent stem cells to address deficient bone in this animal. Finally, we highlight strategies to manipulate stem cells using various growth factors and inhibitors and ultimately how these factors may prove crucial in future advancements within calvarial reconstruction using native skeletal stem cells.
Collapse
Affiliation(s)
- Matthew P Murphy
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Natalina Quarto
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California.,2 Lorry I. Lokey Stem Cell Research Building, Stanford Stem Cell Biology and Regenerative Medicine Institute, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University , Stanford, California
| |
Collapse
|
46
|
Schock EN, Brugmann SA. Discovery, Diagnosis, and Etiology of Craniofacial Ciliopathies. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028258. [PMID: 28213462 DOI: 10.1101/cshperspect.a028258] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Seventy-five percent of congenital disorders present with some form of craniofacial malformation. The frequency and severity of these malformations makes understanding the etiological basis crucial for diagnosis and treatment. A significant link between craniofacial malformations and primary cilia arose several years ago with the determination that ∼30% of ciliopathies could be primarily defined by their craniofacial phenotype. The link between the cilium and the face has proven significant, as several new "craniofacial ciliopathies" have recently been diagnosed. Herein, we reevaluate public disease databases, report several new craniofacial ciliopathies, and propose several "predicted" craniofacial ciliopathies. Furthermore, we discuss why the craniofacial complex is so sensitive to ciliopathic dysfunction, addressing tissue-specific functions of the cilium as well as its role in signal transduction relevant to craniofacial development. As a whole, these analyses suggest a characteristic facial phenotype associated with craniofacial ciliopathies that can perhaps be used for rapid discovery and diagnosis of similar disorders in the future.
Collapse
Affiliation(s)
- Elizabeth N Schock
- Division of Plastic Surgery, Department of Surgery, and Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Samantha A Brugmann
- Division of Plastic Surgery, Department of Surgery, and Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| |
Collapse
|
47
|
Percival CJ, Marangoni P, Tapaltsyan V, Klein O, Hallgrímsson B. The Interaction of Genetic Background and Mutational Effects in Regulation of Mouse Craniofacial Shape. G3 (BETHESDA, MD.) 2017; 7:1439-1450. [PMID: 28280213 PMCID: PMC5427488 DOI: 10.1534/g3.117.040659] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/03/2017] [Indexed: 11/18/2022]
Abstract
Inbred genetic background significantly influences the expression of phenotypes associated with known genetic perturbations and can underlie variation in disease severity between individuals with the same mutation. However, the effect of epistatic interactions on the development of complex traits, such as craniofacial morphology, is poorly understood. Here, we investigated the effect of three inbred backgrounds (129X1/SvJ, C57BL/6J, and FVB/NJ) on the expression of craniofacial dysmorphology in mice (Mus musculus) with loss of function in three members of the Sprouty family of growth factor negative regulators (Spry1, Spry2, or Spry4) in order to explore the impact of epistatic interactions on skull morphology. We found that the interaction of inbred background and the Sprouty genotype explains as much craniofacial shape variation as the Sprouty genotype alone. The most severely affected genotypes display a relatively short and wide skull, a rounded cranial vault, and a more highly angled inferior profile. Our results suggest that the FVB background is more resilient to Sprouty loss of function than either C57 or 129, and that Spry4 loss is generally less severe than loss of Spry1 or Spry2 While the specific modifier genes responsible for these significant background effects remain unknown, our results highlight the value of intercrossing mice of multiple inbred backgrounds to identify the genes and developmental interactions that modulate the severity of craniofacial dysmorphology. Our quantitative results represent an important first step toward elucidating genetic interactions underlying variation in robustness to known genetic perturbations in mice.
Collapse
Affiliation(s)
- Christopher J Percival
- Alberta Children's Hospital Institute for Child and Maternal Health, University of Calgary, Alberta T2N 4N1, Canada
- The McCaig Bone and Joint Institute, University of Calgary, Alberta T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Alberta T2N 4N1, Canada
| | - Pauline Marangoni
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
| | - Vagan Tapaltsyan
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
- Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, California 94143
| | - Ophir Klein
- Department of Orofacial Sciences, University of California, San Francisco, California 94143
- Program in Craniofacial Biology, University of California, San Francisco, California 94143
- Department of Pediatrics, University of California, San Francisco, California 94143
- Institute for Human Genetics, University of California, San Francisco, California 94143
| | - Benedikt Hallgrímsson
- Alberta Children's Hospital Institute for Child and Maternal Health, University of Calgary, Alberta T2N 4N1, Canada
- The McCaig Bone and Joint Institute, University of Calgary, Alberta T2N 4Z6, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
48
|
Cleary MA, Narcisi R, Albiero A, Jenner F, de Kroon LMG, Koevoet WJLM, Brama PAJ, van Osch GJVM. Dynamic Regulation of TWIST1 Expression During Chondrogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev 2017; 26:751-761. [PMID: 28300491 DOI: 10.1089/scd.2016.0308] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (BMSCs) are clinically promising to repair damaged articular cartilage. This study investigated TWIST1, an important transcriptional regulator in mesenchymal lineages, in BMSC chondrogenesis. We hypothesized that downregulation of TWIST1 expression is required for in vitro chondrogenic differentiation. Indeed, significant downregulation of TWIST1 was observed in murine skeletal progenitor cells during limb development (N = 3 embryos), and during chondrogenic differentiation of culture-expanded human articular chondrocytes (N = 3 donors) and isolated adult human BMSCs (N = 7 donors), consistent with an inhibitory effect of TWIST1 expression on chondrogenic differentiation. Silencing of TWIST1 expression in BMSCs by siRNA, however, did not improve chondrogenic differentiation potential. Interestingly, additional investigation revealed that downregulation of TWIST1 in chondrogenic BMSCs is preceded by an initial upregulation. Similar upregulation is observed in non-chondrogenic BMSCs (N = 5 donors); however, non-chondrogenic cells fail to downregulate TWIST1 expression thereafter, preventing their chondrogenic differentiation. This study describes for the first time endogenous TWIST1 expression during in vitro chondrogenic differentiation of human BMSCs, demonstrating dynamic regulation of TWIST1 expression whereby upregulation and then downregulation of TWIST1 expression are required for chondrogenic differentiation of BMSCs. Elucidation of the molecular regulation of, and by, TWIST1 will provide targets for optimization of BMSC chondrogenic differentiation culture.
Collapse
Affiliation(s)
- Mairéad A Cleary
- 1 School of Veterinary Medicine, Veterinary Clinical Sciences, University College Dublin , Dublin, Ireland .,2 Department of Orthopaedics, Erasmus MC, University Medical Center , Rotterdam, the Netherlands
| | - Roberto Narcisi
- 2 Department of Orthopaedics, Erasmus MC, University Medical Center , Rotterdam, the Netherlands
| | - Anna Albiero
- 2 Department of Orthopaedics, Erasmus MC, University Medical Center , Rotterdam, the Netherlands
| | - Florien Jenner
- 3 University of Veterinary Medicine Vienna , Equine Hospital, Vienna, Austria
| | - Laurie M G de Kroon
- 2 Department of Orthopaedics, Erasmus MC, University Medical Center , Rotterdam, the Netherlands .,4 Department of Rheumatology, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Wendy J L M Koevoet
- 5 Department of Otorhinolaryngology, Erasmus MC, University Medical Center , Rotterdam, the Netherlands
| | - Pieter A J Brama
- 1 School of Veterinary Medicine, Veterinary Clinical Sciences, University College Dublin , Dublin, Ireland
| | - Gerjo J V M van Osch
- 2 Department of Orthopaedics, Erasmus MC, University Medical Center , Rotterdam, the Netherlands .,5 Department of Otorhinolaryngology, Erasmus MC, University Medical Center , Rotterdam, the Netherlands
| |
Collapse
|
49
|
Yi S, Yu M, Yang S, Miron RJ, Zhang Y. Tcf12, A Member of Basic Helix-Loop-Helix Transcription Factors, Mediates Bone Marrow Mesenchymal Stem Cell Osteogenic Differentiation In Vitro and In Vivo. Stem Cells 2017; 35:386-397. [PMID: 27574032 DOI: 10.1002/stem.2491] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 07/05/2016] [Accepted: 07/29/2016] [Indexed: 01/12/2023]
Abstract
Several basic Helix-Loop-Helix transcription factors have recently been identified to regulate mesenchymal stem cell (MSC) differentiation. In the present study, Tcf12 was investigated for its involvement in the osteoblastic cell commitment of MSCs. Tcf12 was found highly expressed in undifferentiated MSCs whereas its expression decreased following osteogenic culture differentiation. Interestingly, Tcf12 endogenous silencing using shRNA lentivirus significantly promoted the differentiation ability of MSCs evaluated by alkaline phosphatase staining, alizarin red staining and expression of osteoblast-specific markers by real-time PCR. Conversely, overexpression of Tcf12 in MSCs suppressed osteoblast differentiation. It was further found that silencing of Tcf12 activated bone morphogenetic protein (BMP) signaling and extracellular signal-regulated kinase (Erk)1/2 signaling pathway activity and upregulated the expression of phospho-SMAD1 and phospho-Erk1/2. A BMP inhibitor (LDN-193189) and Erk1/2 signaling pathway inhibitor (U0126) reduced these findings in the Tcf12 silencing group. Following these in vitro results, a poly-L-lactic acid/Hydroxyappatite scaffold carrying Tcf12 silencing lentivirus was utilized to investigate the repair of bone defects in vivo. The use of Tcf12 silencing lentivirus significantly promoted new bone formation in 3-mm mouse calvarial defects as assessed by micro-CT and histological examination whereas overexpression of Tcf12 inhibited new bone formation. Collectively, these data indicate that Tcf12 is a transcription factor highly expressed in the nuclei of stem cells and its downregulation plays an essential role in osteoblast differentiation partially via BMP and Erk1/2 signaling pathways. Stem Cells 2017;35:386-397.
Collapse
Affiliation(s)
- Siqi Yi
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
- Medical Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Miao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Shuang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
| | - Richard J Miron
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, Florida, USA
- Department of Preventive, Restorative and Pediatric Dentistry, School of Dental Medicine, University of Bern, Switzerland
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, People's Republic of China
- Medical Research Institute, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
50
|
Embryonic Explant Culture: Studying Effects of Regulatory Molecules on Gene Expression in Craniofacial Tissues. Methods Mol Biol 2017; 1537:367-380. [PMID: 27924605 DOI: 10.1007/978-1-4939-6685-1_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
The ex vivo culture of embryonic tissue explants permits the continuous monitoring of growth and morphogenesis at specific embryonic stages. The functions of soluble regulatory molecules can be analyzed by introducing them into culture medium or locally with beads to the tissue. Gene expression in the manipulated tissue explants can be analyzed using in situ hybridization, quantitative PCR, and reporter constructs combined to organ culture to examine the functions of the signaling molecules.
Collapse
|