51
|
Ramanathan A, Srijaya TC, Sukumaran P, Zain RB, Abu Kasim NH. Homeobox genes and tooth development: Understanding the biological pathways and applications in regenerative dental science. Arch Oral Biol 2017; 85:23-39. [PMID: 29031235 DOI: 10.1016/j.archoralbio.2017.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 09/27/2017] [Accepted: 09/30/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Homeobox genes are a group of conserved class of transcription factors that function as key regulators during the embryonic developmental processes. They act as master regulator for developmental genes, which involves coordinated actions of various auto and cross-regulatory mechanisms. In this review, we summarize the expression pattern of homeobox genes in relation to the tooth development and various signaling pathways or molecules contributing to the specific actions of these genes in the regulation of odontogenesis. MATERIALS AND METHODS An electronic search was undertaken using combination of keywords e.g. Homeobox genes, tooth development, dental diseases, stem cells, induced pluripotent stem cells, gene control region was used as search terms in PubMed and Web of Science and relevant full text articles and abstract were retrieved that were written in English. A manual hand search in text books were also carried out. Articles related to homeobox genes in dentistry and tissue engineering and regenerative medicine of odontogenesis were selected. RESULTS The possible perspective of stem cells technology in odontogenesis and subsequent analysis of gene correction pertaining to dental disorders through the possibility of induced pluripotent stem cells technology is also inferred. CONCLUSIONS We demonstrate the promising role of tissue engineering and regenerative medicine on odontogenesis, which can generate a new ray of hope in the field of dental science.
Collapse
Affiliation(s)
- Anand Ramanathan
- Oral Cancer Research and Coordinating Center, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia; Department of Oral & Maxillofacial Clinical Science, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| | | | - Prema Sukumaran
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| | - Rosnah Binti Zain
- Oral Cancer Research and Coordinating Center, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia; Department of Oral & Maxillofacial Clinical Science, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia; Faculty of Dentistry, MAHSA University, Jenjarom, Selangor, Malaysia.
| | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
52
|
Zhang G, Zhang W, Li B, Stringer-Reasor E, Chu C, Sun L, Bae S, Chen D, Wei S, Jiao K, Yang WH, Cui R, Liu R, Wang L. MicroRNA-200c and microRNA- 141 are regulated by a FOXP3-KAT2B axis and associated with tumor metastasis in breast cancer. Breast Cancer Res 2017. [PMID: 28637482 PMCID: PMC5480201 DOI: 10.1186/s13058-017-0858-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Members of the microRNA (miR)-200 family, which are involved in tumor metastasis, have potential as cancer biomarkers, but their regulatory mechanisms remain elusive. Methods We investigated FOXP3-inducible breast cancer cells, Foxp3 heterozygous Scurfy mutant (Foxp3sf/+) female mice, and patients with breast cancer for characterization of the formation and regulation of the miR-200 family in breast cancer cells and circulation. Participants (259), including patients with breast cancer or benign breast tumors, members of breast cancer families, and healthy controls, were assessed for tumor and circulating levels of the miR-200 family. Results First, we identified a FOXP3-KAT2B-miR-200c/141 axis in breast cancer cells. Second, aging Foxp3sf/+ female mice developed spontaneous breast cancers and lung metastases. Levels of miR-200c and miR-141 were lower in Foxp3sf/+ tumor cells than in normal breast epithelial cells, but plasma levels of miR-200c and miR-141 in the Foxp3sf/+ mice increased during tumor progression and metastasis. Third, in patients with breast cancer, the levels of miR-200c and 141 were lower in FOXP3low relative to those with FOXP3high breast cancer cells, especially in late-stage and metastatic cancer cells. The levels of miR-200c and miR-141 were higher in plasma from patients with metastatic breast cancer than in plasma from those with localized breast cancer, with benign breast tumors, with a family history of breast cancer, or from healthy controls. Finally, in Foxp3sf/+ mice, plasma miR-200c and miR-141 appeared to be released from tumor cells. Conclusions miR-200c and miR-141 are regulated by a FOXP3-KAT2B axis in breast cancer cells, and circulating levels of miR-200c and miR-141 are potential biomarkers for early detection of breast cancer metastases. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0858-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guangxin Zhang
- Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China.,Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Wei Zhang
- Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Bingjin Li
- Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China
| | - Erica Stringer-Reasor
- Hematology/Oncology Section, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Chengjing Chu
- Department of Applied Psychology, Humanities and Management Colleges, Guangdong Medical University, Dongguan, 523808, People's Republic of China
| | - Liyan Sun
- Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Sejong Bae
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Dongquan Chen
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Shi Wei
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Kenneth Jiao
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University, Savannah, GA, 31404, USA
| | - Ranji Cui
- Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, 130041, People's Republic of China.
| | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA. .,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
53
|
Yachuan Z, Xuedong Z, Liwei Z. [Expression and function of microRNAs in enamel development]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2017; 35:328-333. [PMID: 28675021 DOI: 10.7518/hxkq.2017.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
microRNAs (miRNAs) are endogenous short, noncoding RNAs that can negatively regulate gene expression post-transcriptionally. miRNAs are involved in multiple developmental events in various tissues and organs, including dental enamel development. Any disruption during enamel development may result in inherited enamel malformations. This article reviews the expression and function of miRNAs in enamel development.
Collapse
Affiliation(s)
- Zhou Yachuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhou Xuedong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zheng Liwei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
54
|
MicroRNAs in ectodermal appendages. Curr Opin Genet Dev 2017; 43:61-66. [DOI: 10.1016/j.gde.2016.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/12/2016] [Accepted: 12/21/2016] [Indexed: 11/22/2022]
|
55
|
Jin Y, Wang C, Cheng S, Zhao Z, Li J. MicroRNA control of tooth formation and eruption. Arch Oral Biol 2017; 73:302-310. [DOI: 10.1016/j.archoralbio.2016.08.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023]
|
56
|
Regulatory roles of microRNAs in human dental tissues. Gene 2017; 596:9-18. [DOI: 10.1016/j.gene.2016.10.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/06/2016] [Accepted: 10/06/2016] [Indexed: 01/04/2023]
|
57
|
Sun Z, Yu W, Sanz Navarro M, Sweat M, Eliason S, Sharp T, Liu H, Seidel K, Zhang L, Moreno M, Lynch T, Holton NE, Rogers L, Neff T, Goodheart MJ, Michon F, Klein OD, Chai Y, Dupuy A, Engelhardt JF, Chen Z, Amendt BA. Sox2 and Lef-1 interact with Pitx2 to regulate incisor development and stem cell renewal. Development 2016; 143:4115-4126. [PMID: 27660324 PMCID: PMC5117215 DOI: 10.1242/dev.138883] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/06/2016] [Indexed: 12/26/2022]
Abstract
Sox2 marks dental epithelial stem cells (DESCs) in both mammals and reptiles, and in this article we demonstrate several Sox2 transcriptional mechanisms that regulate dental stem cell fate and incisor growth. Conditional Sox2 deletion in the oral and dental epithelium results in severe craniofacial defects, including impaired dental stem cell proliferation, arrested incisor development and abnormal molar development. The murine incisor develops initially but is absorbed independently of apoptosis owing to a lack of progenitor cell proliferation and differentiation. Tamoxifen-induced inactivation of Sox2 demonstrates the requirement of Sox2 for maintenance of the DESCs in adult mice. Conditional overexpression of Lef-1 in mice increases DESC proliferation and creates a new labial cervical loop stem cell compartment, which produces rapidly growing long tusk-like incisors, and Lef-1 epithelial overexpression partially rescues the tooth arrest in Sox2 conditional knockout mice. Mechanistically, Pitx2 and Sox2 interact physically and regulate Lef-1, Pitx2 and Sox2 expression during development. Thus, we have uncovered a Pitx2-Sox2-Lef-1 transcriptional mechanism that regulates DESC homeostasis and dental development.
Collapse
Affiliation(s)
- Zhao Sun
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Wenjie Yu
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Maria Sanz Navarro
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland
| | - Mason Sweat
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Steven Eliason
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Thad Sharp
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Huan Liu
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P.R.China
| | - Kerstin Seidel
- Department of Orofacial Sciences and Program in Craniofacial Biology, UCSF, San Francisco, CA 94143-0442, USA
| | - Li Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P.R.China
| | - Myriam Moreno
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Thomas Lynch
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Nathan E Holton
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| | - Laura Rogers
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Traci Neff
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Michael J Goodheart
- Department of Obstetrics and Gynecology, The University of Iowa, Iowa City, IA 52242, USA
| | - Frederic Michon
- Developmental Biology Program, Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, UCSF, San Francisco, CA 94143-0442, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Adam Dupuy
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - John F Engelhardt
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Zhi Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, P.R.China
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, and the Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA 52242, USA
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
58
|
Nascimento MAB, Nonaka CFW, Barboza CAG, Freitas RDA, Pereira Pinto L, Souza LBD. Immunoexpression of BMP-2 and BMP-4 and their receptors, BMPR-IA and BMPR-II, in ameloblastomas and adenomatoid odontogenic tumors. Arch Oral Biol 2016; 73:223-229. [PMID: 27780042 DOI: 10.1016/j.archoralbio.2016.10.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 09/23/2016] [Accepted: 10/17/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The present study evaluated the immunohistochemical expression of BMP-2 and BMP-4 and of their receptors (BMPR-IA and BMPR-II) in solid ameloblastoma (SA), unicystic ameloblastoma (UA) and adenomatoid odontogenic tumor (AOT) in order to obtain a better understanding of their role in the development and biological behavior of these tumors. DESIGN This study analyzed these proteins in 30 cases of SA, 10 cases of UA, and 30 cases of AOT. Immunoexpression was evaluated in the parenchyma and stroma by attributing the following scores: 0, no stained cells; 1, ≤10%; 2, >10% and ≤25%; 3, >25% and ≤50%; 4, >50% and ≤75%.; 5, >75% stained cells. RESULTS In SAs, positive correlations were observed between the stromal and parenchymal expression of BMP-2 (p<0.001) and between the stromal expression of BMP-2 and BMP-4 (p=0.020), as well as between the stromal expression of BMPR-II and BMP-4 (p=0.001) and the stromal and parenchymal expression of BMPR-II (p<0.001). In UAs, correlations were detected between the stromal and parenchymal expression of BMP-4 (p=0.035) and between the stromal expression of BMP-4 and BMPR-IA (p=0.022). In AOTs, analysis of immunoexpression in the parenchyma revealed positive correlations between all proteins. CONCLUSION BMPs and their receptors play an important role in the differentiation and development of ameloblastomas and AOTs, but may not explain the different biological behaviors of these lesions. The positive correlation observed in AOTs might be related to the formation of mineralized material in this tumor.
Collapse
Affiliation(s)
| | | | | | | | - Leão Pereira Pinto
- Postgraduate Program in Oral Pathology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Lélia Batista de Souza
- Postgraduate Program in Oral Pathology, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
59
|
Suppression of MicroRNA 200 Family Expression by Oncogenic KRAS Activation Promotes Cell Survival and Epithelial-Mesenchymal Transition in KRAS-Driven Cancer. Mol Cell Biol 2016; 36:2742-2754. [PMID: 27550813 DOI: 10.1128/mcb.00079-16] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022] Open
Abstract
Oncogenic KRAS contributes to malignant transformation, antiapoptosis, and metastasis in multiple human cancers, such as lung, colon, and pancreatic cancers and melanoma. MicroRNAs (miRNAs) are endogenous 18- to 25-nucleotide noncoding small RNAs that regulate gene expression in a sequence-specific manner via the degradation of target mRNAs or inhibition of protein translation. In the present study, using array-based miRNA profiling in IMR90 and MCF10A cells expressing oncogenic KRAS, we identified that the expression of the microRNA 200 (mir-200) family was suppressed by KRAS activation and that this suppression was mediated by the transcription factors JUN and SP1 in addition to ZEB1. Restoration of mir-200 expression compromised KRAS-induced cellular transformation in vitro and tumor formation in vivo In addition, we found that enforced expression of mir-200 abrogated KRAS-induced resistance to apoptosis by directly targeting the antiapoptotic gene BCL2 Finally, mir-200 was able to antagonize the epithelial-mesenchymal transition (EMT) driven by mutant KRAS. Collectively, our results suggest that repression of endogenous mir-200 expression is one of the important cellular responses to KRAS activation during tumor initiation and progression.
Collapse
|
60
|
Zhang F, Song J, Zhang H, Huang E, Song D, Tollemar V, Wang J, Wang J, Mohammed M, Wei Q, Fan J, Liao J, Zou Y, Liu F, Hu X, Qu X, Chen L, Yu X, Luu HH, Lee MJ, He TC, Ji P. Wnt and BMP Signaling Crosstalk in Regulating Dental Stem Cells: Implications in Dental Tissue Engineering. Genes Dis 2016; 3:263-276. [PMID: 28491933 PMCID: PMC5421560 DOI: 10.1016/j.gendis.2016.09.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tooth is a complex hard tissue organ and consists of multiple cell types that are regulated by important signaling pathways such as Wnt and BMP signaling. Serious injuries and/or loss of tooth or periodontal tissues may significantly impact aesthetic appearance, essential oral functions and the quality of life. Regenerative dentistry holds great promise in treating oral/dental disorders. The past decade has witnessed a rapid expansion of our understanding of the biological features of dental stem cells, along with the signaling mechanisms governing stem cell self-renewal and differentiation. In this review, we first summarize the biological characteristics of seven types of dental stem cells, including dental pulp stem cells, stem cells from apical papilla, stem cells from human exfoliated deciduous teeth, dental follicle precursor cells, periodontal ligament stem cells, alveolar bone-derived mesenchymal stem cells (MSCs), and MSCs from gingiva. We then focus on how these stem cells are regulated by bone morphogenetic protein (BMP) and/or Wnt signaling by examining the interplays between these pathways. Lastly, we analyze the current status of dental tissue engineering strategies that utilize oral/dental stem cells by harnessing the interplays between BMP and Wnt pathways. We also highlight the challenges that must be addressed before the dental stem cells may reach any clinical applications. Thus, we can expect to witness significant progresses to be made in regenerative dentistry in the coming decade.
Collapse
Affiliation(s)
- Fugui Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jinglin Song
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China
| | - Hongmei Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Enyi Huang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Dongzhe Song
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Department of Conservative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Viktor Tollemar
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jing Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jinhua Wang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Maryam Mohammed
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qiang Wei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Junyi Liao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yulong Zou
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Feng Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xue Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xiangyang Qu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Liqun Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China.,Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA.,Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Ping Ji
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing 401147, China
| |
Collapse
|
61
|
Abstract
Transforming growth factor β (TGF-β) and related growth factors are secreted pleiotropic factors that play critical roles in embryogenesis and adult tissue homeostasis by regulating cell proliferation, differentiation, death, and migration. The TGF-β family members signal via heteromeric complexes of type I and type II receptors, which activate members of the Smad family of signal transducers. The main attribute of the TGF-β signaling pathway is context-dependence. Depending on the concentration and type of ligand, target tissue, and developmental stage, TGF-β family members transmit distinct signals. Deregulation of TGF-β signaling contributes to developmental defects and human diseases. More than a decade of studies have revealed the framework by which TGF-βs encode a context-dependent signal, which includes various positive and negative modifiers of the principal elements of the signaling pathway, the receptors, and the Smad proteins. In this review, we first introduce some basic components of the TGF-β signaling pathways and their actions, and then discuss posttranslational modifications and modulatory partners that modify the outcome of the signaling and contribute to its context-dependence, including small noncoding RNAs.
Collapse
Affiliation(s)
- Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94143
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
62
|
Hong L, Sharp T, Khorsand B, Fischer C, Eliason S, Salem A, Akkouch A, Brogden K, Amendt BA. MicroRNA-200c Represses IL-6, IL-8, and CCL-5 Expression and Enhances Osteogenic Differentiation. PLoS One 2016; 11:e0160915. [PMID: 27529418 PMCID: PMC4987006 DOI: 10.1371/journal.pone.0160915] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 07/27/2016] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRs) regulate inflammation and BMP antagonists, thus they have potential uses as therapeutic reagents. However, the molecular function of miR-200c in modulating proinflammatory and bone metabolic mediators and osteogenic differentiation is not known. After miR-200c was transduced into a human embryonic palatal mesenchyme (HEPM) (a cell line of preosteoblasts), using lentiviral vectors, the resulting miR-200c overexpression increased osteogenic differentiation biomarkers, including osteocalcin (OCN) transcripts and calcium content. miR-200c expression also down-regulated interleukin (IL)-6, IL-8, and chemokine (C-C motif) ligand (CCL)-5 under lipopolysaccharide (LPS) stimulation and increased osteoprotegerin (OPG) in these cells. miR-200c directly regulates the expression of IL-6, IL-8 and CCL-5 transcripts by binding to their 3'UTRs. A plasmid-based miR-200c inhibitor effectively reduces their binding activities. Additionally, miR-200c delivered using polyethylenimine (PEI) nanoparticles effectively inhibits IL-6, IL-8 and CCL-5 in primary human periodontal ligament fibroblasts and increases the biomarkers of osteogenic differentiation in human bone marrow mesenchymal stem cells (MSCs), including calcium content, ALP, and Runx2. These data demonstrate that miR-200c represses IL-6, IL-8 and CCL-5 and improves osteogenic differentiation. miR-200c may potentially be used as an effective means to prevent periodontitis-associated bone loss by arresting inflammation and osteoclastogenesis and enhancing bone regeneration.
Collapse
Affiliation(s)
- Liu Hong
- Dows Institute for Dental Research, College of Dentistry, the University of Iowa, Iowa City, IA, United States of America
- Center for Craniofacial Anomalies Research, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
| | - Thad Sharp
- Department of Anatomy and Cell Biology, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
| | - Behnoush Khorsand
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, the University of Iowa, Iowa City, IA, United States of America
| | - Carol Fischer
- Dows Institute for Dental Research, College of Dentistry, the University of Iowa, Iowa City, IA, United States of America
| | - Steven Eliason
- Center for Craniofacial Anomalies Research, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
- Department of Anatomy and Cell Biology, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
| | - Ali Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, the University of Iowa, Iowa City, IA, United States of America
| | - Adil Akkouch
- Dows Institute for Dental Research, College of Dentistry, the University of Iowa, Iowa City, IA, United States of America
| | - Kim Brogden
- Dows Institute for Dental Research, College of Dentistry, the University of Iowa, Iowa City, IA, United States of America
| | - Brad A. Amendt
- Dows Institute for Dental Research, College of Dentistry, the University of Iowa, Iowa City, IA, United States of America
- Center for Craniofacial Anomalies Research, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
- Department of Anatomy and Cell Biology, Carver College of Medicine, the University of Iowa, Iowa City, IA, United States of America
| |
Collapse
|
63
|
The EMT-activator ZEB1 induces bone metastasis associated genes including BMP-inhibitors. Oncotarget 2016; 6:14399-412. [PMID: 25973542 PMCID: PMC4546475 DOI: 10.18632/oncotarget.3882] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 04/21/2015] [Indexed: 01/05/2023] Open
Abstract
Tumor cell invasion, dissemination and metastasis is triggered by an aberrant activation of epithelial-to-mesenchymal transition (EMT), often mediated by the transcription factor ZEB1. Disseminating tumor cells must acquire specific features that allow them to colonize at different organ sites. Here we identify a set of genes that is highly expressed in breast cancer bone metastasis and activated by ZEB1. This gene set includes various secreted factors, e.g. the BMP-inhibitor FST, that are described to reorganize the bone microenvironment. By inactivating BMP-signaling, BMP-inhibitors are well-known to induce osteolysis in development and disease. We here demonstrate that the expression of ZEB1 and BMP-inhibitors is correlated with bone metastasis, but not with brain or lung metastasis of breast cancer patients. In addition, we show that this correlated expression pattern is causally linked, as ZEB1 induces the expression of the BMP-inhibitors NOG, FST and CHRDL1 both by directly increasing their gene transcription, as well as by indirectly suppressing their reduction via miR-200 family members. Consequently, ZEB1 stimulates BMP-inhibitor mediated osteoclast differentiation. These findings suggest that ZEB1 is not only driving EMT, but also contributes to the formation of osteolytic bone metastases in breast cancer.
Collapse
|
64
|
miR-200c: a versatile watchdog in cancer progression, EMT, and drug resistance. J Mol Med (Berl) 2016; 94:629-44. [PMID: 27094812 DOI: 10.1007/s00109-016-1420-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 04/05/2016] [Accepted: 04/11/2016] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are 20-22-nucleotide small endogenous non-coding RNAs which regulate gene expression at post-transcriptional level. In the last two decades, identification of almost 2600 miRNAs in human and their potential to be modulated opened a new avenue to target almost all hallmarks of cancer. miRNAs have been classified as tumor suppressors or oncogenes depending on the phenotype they induce, the targets they modulate, and the tissue where they function. miR-200c, an illustrious tumor suppressor, is one of the highly studied miRNAs in terms of development, stemness, proliferation, epithelial-mesenchymal transition (EMT), therapy resistance, and metastasis. In this review, we first focus on the regulation of miR-200c expression and its role in regulating EMT in a ZEB1/E-cadherin axis-dependent and ZEB1/E-cadherin axis-independent manner. We then describe the role of miR-200c in therapy resistance in terms of multidrug resistance, chemoresistance, targeted therapy resistance, and radiotherapy resistance in various cancer types. We highlight the importance of miR-200c at the intersection of EMT and chemoresistance. Furthermore, we show how miR-200c coordinates several important signaling cascades such as TGF-β signaling, PI3K/Akt signaling, Notch signaling, VEGF signaling, and NF-κB signaling. Finally, we discuss miR-200c as a potential prognostic/diagnostic biomarker in several diseases, but mainly focusing on cancer and its potential application in future therapeutics.
Collapse
|
65
|
An integrated miRNA functional screening and target validation method for organ morphogenesis. Sci Rep 2016; 6:23215. [PMID: 26980315 PMCID: PMC4793243 DOI: 10.1038/srep23215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 02/26/2016] [Indexed: 12/20/2022] Open
Abstract
The relative ease of identifying microRNAs and their increasing recognition as important regulators of organogenesis motivate the development of methods to efficiently assess microRNA function during organ morphogenesis. In this context, embryonic organ explants provide a reliable and reproducible system that recapitulates some of the important early morphogenetic processes during organ development. Here we present a method to target microRNA function in explanted mouse embryonic organs. Our method combines the use of peptide-based nanoparticles to transfect specific microRNA inhibitors or activators into embryonic organ explants, with a microRNA pulldown assay that allows direct identification of microRNA targets. This method provides effective assessment of microRNA function during organ morphogenesis, allows prioritization of multiple microRNAs in parallel for subsequent genetic approaches, and can be applied to a variety of embryonic organs.
Collapse
|
66
|
Li A, Li Y, Song T, Wang F, Liu D, Fan Z, Cheng S, Zhang C, Wang J, He J, Wang S. Identification of differential microRNA expression during tooth morphogenesis in the heterodont dentition of miniature pigs, SusScrofa. BMC DEVELOPMENTAL BIOLOGY 2015; 15:51. [PMID: 26715101 PMCID: PMC4696248 DOI: 10.1186/s12861-015-0099-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/18/2015] [Indexed: 01/14/2023]
Abstract
Background It has been found that microRNAs (miRNAs) play important roles in the regulation of tooth development, and most likely increase the complexity of the genetic network, thus lead to greater complexity of teeth. But there has been no research about the key microRNAs associated with tooth morphogenesis based on miRNAs expression profiles. Compared to mice, the pig model has plentiful types of teeth, which is similar with the human dental pattern. Therefore, we used miniature pigs as large-animal models to investigate differentially expressed miRNAs expression during tooth morphogenesis in the early developmental stages of tooth germ. Results A custom-designed miRNA microarray with 742 miRNA gene probes was used to analyze the expression profiles of four types of teeth at three stages of tooth development. Of the 591 detectable miRNA transcripts, 212 miRNAs were continuously expressed in all types of tooth germ, but the numbers of miRNA transcript among the four different types of teeth at each embryonic stage were statistically significant differences (p < 0.01). The hierarchical clustering and principal component analysis results suggest that the miRNA expression was globally altered by types and temporal changes. By clustering analysis, we predicted 11 unique miRNA sequences that belong to mir-103 and mir-107, mir-133a and mir-133b, and mir-127 isomiR families. The results of real-time reverse-transcriptase PCR and in situ hybridization experiments revealed that five representative miRNAs may play important roles during different developmental stages of the incisor, canine, biscuspid, and molar, respectively. Conclusions The present study indicated that these five miRNAs, including ssc-miR-103 and ssc-miR-107, ssc-miR-133a and ssc-miR-133b, and ssc-miR-127, may play key regulatory roles in different types of teeth during different stages and thus may play critical roles in tooth morphogenesis during early development in miniature pigs. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0099-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ang Li
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China. .,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi Wu Lu No.98, Xi'an, 710004, China.
| | - Ye Li
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China. .,Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi Wu Lu No.98, Xi'an, 710004, China.
| | - Tieli Song
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China.
| | - Fu Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China.
| | - Dayong Liu
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China.
| | - Zhipeng Fan
- Laboratory of Molecular Signaling and Stem Cells Therapy, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, China.
| | - San Cheng
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, You An Men Wai Xi TouTiao No.10, Beijing, 100069, China.
| | - Chunmei Zhang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China.
| | - Jinsong Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China. .,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, You An Men Wai Xi TouTiao No.10, Beijing, 100069, China.
| | - Junqi He
- Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, You An Men Wai Xi TouTiao No.10, Beijing, 100069, China.
| | - Songlin Wang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Tian Tan Xi Li No.4, Beijing, 100050, China. .,Department of Biochemistry and Molecular Biology, Capital Medical University School of Basic Medical Sciences, You An Men Wai Xi TouTiao No.10, Beijing, 100069, China.
| |
Collapse
|
67
|
Bloomquist RF, Parnell NF, Phillips KA, Fowler TE, Yu TY, Sharpe PT, Streelman JT. Coevolutionary patterning of teeth and taste buds. Proc Natl Acad Sci U S A 2015; 112:E5954-62. [PMID: 26483492 PMCID: PMC4640805 DOI: 10.1073/pnas.1514298112] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Teeth and taste buds are iteratively patterned structures that line the oro-pharynx of vertebrates. Biologists do not fully understand how teeth and taste buds develop from undifferentiated epithelium or how variation in organ density is regulated. These organs are typically studied independently because of their separate anatomical location in mammals: teeth on the jaw margin and taste buds on the tongue. However, in many aquatic animals like bony fishes, teeth and taste buds are colocalized one next to the other. Using genetic mapping in cichlid fishes, we identified shared loci controlling a positive correlation between tooth and taste bud densities. Genome intervals contained candidate genes expressed in tooth and taste bud fields. sfrp5 and bmper, notable for roles in Wingless (Wnt) and bone morphogenetic protein (BMP) signaling, were differentially expressed across cichlid species with divergent tooth and taste bud density, and were expressed in the development of both organs in mice. Synexpression analysis and chemical manipulation of Wnt, BMP, and Hedgehog (Hh) pathways suggest that a common cichlid oral lamina is competent to form teeth or taste buds. Wnt signaling couples tooth and taste bud density and BMP and Hh mediate distinct organ identity. Synthesizing data from fish and mouse, we suggest that the Wnt-BMP-Hh regulatory hierarchy that configures teeth and taste buds on mammalian jaws and tongues may be an evolutionary remnant inherited from ancestors wherein these organs were copatterned from common epithelium.
Collapse
Affiliation(s)
- Ryan F Bloomquist
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332; College of Dental Medicine, Georgia Regents University, Augusta, GA 30912;
| | - Nicholas F Parnell
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kristine A Phillips
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Teresa E Fowler
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Tian Y Yu
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London WC2R 2LS, United Kingdom
| | - Paul T Sharpe
- Department of Craniofacial Development and Stem Cell Biology, Dental Institute, King's College London, London WC2R 2LS, United Kingdom
| | - J Todd Streelman
- School of Biology and Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332;
| |
Collapse
|
68
|
Tavares ALP, Artinger KB, Clouthier DE. Regulating Craniofacial Development at the 3' End: MicroRNAs and Their Function in Facial Morphogenesis. Curr Top Dev Biol 2015; 115:335-75. [PMID: 26589932 DOI: 10.1016/bs.ctdb.2015.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Defects in craniofacial development represent a majority of observed human birth defects, occurring at a rate as high as 1:800 live births. These defects often occur due to changes in neural crest cell (NCC) patterning and development and can affect non-NCC-derived structures due to interactions between NCCs and the surrounding cell types. Proper craniofacial development requires an intricate array of gene expression networks that are tightly controlled spatiotemporally by a number of regulatory mechanisms. One of these mechanisms involves the action of microRNAs (miRNAs), a class of noncoding RNAs that repress gene expression by binding to miRNA recognition sequences typically located in the 3' UTR of target mRNAs. Recent evidence illustrates that miRNAs are crucial for vertebrate facial morphogenesis, with changes in miRNA expression leading to facial birth defects, including some in complex human syndromes such as 22q11 (DiGeorge Syndrome). In this review, we highlight the current understanding of miRNA biogenesis, the roles of miRNAs in overall craniofacial development, the impact that loss of miRNAs has on normal development and the requirement for miRNAs in the development of specific craniofacial structures, including teeth. From these studies, it is clear that miRNAs are essential for normal facial development and morphogenesis, and a potential key in establishing new paradigms for repair and regeneration of facial defects.
Collapse
Affiliation(s)
- Andre L P Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristin B Artinger
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
69
|
Yu T, Volponi AA, Babb R, An Z, Sharpe PT. Stem Cells in Tooth Development, Growth, Repair, and Regeneration. Curr Top Dev Biol 2015; 115:187-212. [PMID: 26589926 DOI: 10.1016/bs.ctdb.2015.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human teeth contain stem cells in all their mesenchymal-derived tissues, which include the pulp, periodontal ligament, and developing roots, in addition to the support tissues such as the alveolar bone. The precise roles of these cells remain poorly understood and most likely involve tissue repair mechanisms but their relative ease of harvesting makes teeth a valuable potential source of mesenchymal stem cells (MSCs) for therapeutic use. These dental MSC populations all appear to have the same developmental origins, being derived from cranial neural crest cells, a population of embryonic stem cells with multipotential properties. In rodents, the incisor teeth grow continuously throughout life, a feature that requires populations of continuously active mesenchymal and epithelial stem cells. The discrete locations of these stem cells in the incisor have rendered them amenable for study and much is being learnt about the general properties of these stem cells for the incisor as a model system. The incisor MSCs appear to be a heterogeneous population consisting of cells from different neural crest-derived tissues. The epithelial stem cells can be traced directly back in development to a Sox10(+) population present at the time of tooth initiation. In this review, we describe the basic biology of dental stem cells, their functions, and potential clinical uses.
Collapse
Affiliation(s)
- Tian Yu
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Ana Angelova Volponi
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Rebecca Babb
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Zhengwen An
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Paul T Sharpe
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom.
| |
Collapse
|
70
|
Lozano-Velasco E, Vallejo D, Esteban FJ, Doherty C, Hernández-Torres F, Franco D, Aránega AE. A Pitx2-MicroRNA Pathway Modulates Cell Proliferation in Myoblasts and Skeletal-Muscle Satellite Cells and Promotes Their Commitment to a Myogenic Cell Fate. Mol Cell Biol 2015; 35:2892-909. [PMID: 26055324 PMCID: PMC4525317 DOI: 10.1128/mcb.00536-15] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 01/21/2023] Open
Abstract
The acquisition of a proliferating-cell status from a quiescent state as well as the shift between proliferation and differentiation are key developmental steps in skeletal-muscle stem cells (satellite cells) to provide proper muscle regeneration. However, how satellite cell proliferation is regulated is not fully understood. Here, we report that the c-isoform of the transcription factor Pitx2 increases cell proliferation in myoblasts by downregulating microRNA 15b (miR-15b), miR-23b, miR-106b, and miR-503. This Pitx2c-microRNA (miRNA) pathway also regulates cell proliferation in early-activated satellite cells, enhancing Myf5(+) satellite cells and thereby promoting their commitment to a myogenic cell fate. This study reveals unknown functions of several miRNAs in myoblast and satellite cell behavior and thus may have future applications in regenerative medicine.
Collapse
Affiliation(s)
- Estefanía Lozano-Velasco
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Daniel Vallejo
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Francisco J Esteban
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Chris Doherty
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Francisco Hernández-Torres
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Diego Franco
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Amelia Eva Aránega
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaén, Jaén, Spain
| |
Collapse
|
71
|
Sangani R, Periyasamy-Thandavan S, Kolhe R, Bhattacharyya MH, Chutkan N, Hunter M, Isales C, Hamrick M, Hill WD, Fulzele S. MicroRNAs-141 and 200a regulate the SVCT2 transporter in bone marrow stromal cells. Mol Cell Endocrinol 2015; 410:19-26. [PMID: 25617715 PMCID: PMC4824062 DOI: 10.1016/j.mce.2015.01.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/07/2015] [Accepted: 01/07/2015] [Indexed: 12/16/2022]
Abstract
Vitamin C is a micro-nutrient which plays an important role in bone marrow stromal cell (BMSCs) differentiation to osteogenesis. This vitamin is transported into the BMSCs through the sodium dependent vitamin C transporter 2 (SVCT2). We previously reported that knockdown of the SVCT2 transporter decreases osteogenic differentiation. However, our understanding of the post-transcriptional regulatory mechanism of the SVCT2 transporter remains poor. MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate the messenger RNAs of protein-coding genes. In this study, we aimed to investigate the impact of miR-141 and miR-200a on SVCT2 expression. We found that mouse BMSCs expressed miR-141 and miR-200a and repressed SVCT2 expression at the functional level by targeting the 3'-untranslated region of mRNA. We also found that miR-141 and miR-200a decreased osteogenic differentiation. Furthermore, miRNA inhibitors increased SVCT2 and osteogenic gene expression in BMSCs. Taken together, these results indicate that both miRNAs are novel regulators of the SVCT2 transporter and play an important role in the osteogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Rajnikumar Sangani
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Ravindra Kolhe
- Department of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Maryka H Bhattacharyya
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Monte Hunter
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Carlos Isales
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Mark Hamrick
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - William D Hill
- Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
72
|
MicroRNA 665 Regulates Dentinogenesis through MicroRNA-Mediated Silencing and Epigenetic Mechanisms. Mol Cell Biol 2015; 35:3116-30. [PMID: 26124283 DOI: 10.1128/mcb.00093-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/18/2015] [Indexed: 12/26/2022] Open
Abstract
Studies of proteins involved in microRNA (miRNA) processing, maturation, and silencing have indicated the importance of miRNAs in skeletogenesis, but the specific miRNAs involved in this process are incompletely defined. Here, we identified miRNA 665 (miR-665) as a potential repressor of odontoblast maturation. Studies with cultured cell lines and primary embryonic cells showed that miR-665 represses the expression of early and late odontoblast marker genes and stage-specific proteases involved in dentin maturation. Notably, miR-665 directly targeted Dlx3 mRNA and decreased Dlx3 expression. Furthermore, RNA-induced silencing complex (RISC) immunoprecipitation and biotin-labeled miR-665 pulldown studies identified Kat6a as another potential target of miR-665. KAT6A interacted physically and functionally with RUNX2, activating tissue-specific promoter activity and prompting odontoblast differentiation. Overexpression of miR-665 reduced the recruitment of KAT6A to Dspp and Dmp1 promoters and prevented KAT6A-induced chromatin remodeling, repressing gene transcription. Taken together, our results provide novel molecular evidence that miR-665 functions in an miRNA-epigenetic regulatory network to control dentinogenesis.
Collapse
|
73
|
Qi S, Wu Q, Ma J, Li J, Chen F, Xu Y, Pan Q, Wang R. Effects of neurotrophin receptor-mediated MAGE homology on proliferation and odontoblastic differentiation of mouse dental pulp cells. Cell Prolif 2015; 48:221-30. [PMID: 25736627 DOI: 10.1111/cpr.12171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/30/2014] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES This study aimed to investigate effects of neurotrophin receptor-mediated melanoma antigen-encoding gene homology (NRAGE) on proliferation and odontoblastic differentiation of mouse dental pulp cells (mDPCs). MATERIALS AND METHODS Mouse dental pulp cells were infected with recombinant lentivirus to stably knockdown expression of NRAGE, and biological effects of NRAGE on the cells were detected. Proliferation and odontoblastic differentiation of mDPCs were observed. Simultaneously, mRNA and protein levels of NRAGE and nuclear factor-κB (NF-κB) protein expression were detected. Immunofluorescence assay was used to detect expression and location of NRAGE and NF-κB. RESULTS NRAGE mRNA and protein levels reduced significantly after mDPC odontoblastic induction. Knockdown of NRAGE inhibited the proliferation of mDPCs. However, knockdown of NRAGE enhanced their odontoblastic differentiation with up-regulated ALPase activity. It also promoted mineral nodule formation as well as mRNA and protein expressions of ALP, DSPP and DMP1. Protein levels of NF-κB/p50 significantly increased, whereas NF-κB/p105 protein expression decreased in the mDPC/shNRG group. Immunofluorescence revealed that relocation of NF-κB was similar to that of NRAGE during odontoblastic induction, in which NF-κB translocated from the cytoplasm to the nucleus. CONCLUSION NRAGE is a potent regulator of proliferation and odontoblastic differentiation of mDPCs, which might be via the NF-κB signalling pathway.
Collapse
Affiliation(s)
- S Qi
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Ellis NA, Glazer AM, Donde NN, Cleves PA, Agoglia RM, Miller CT. Distinct developmental genetic mechanisms underlie convergently evolved tooth gain in sticklebacks. Development 2015; 142:2442-51. [PMID: 26062935 DOI: 10.1242/dev.124248] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022]
Abstract
Teeth are a classic model system of organogenesis, as repeated and reciprocal epithelial and mesenchymal interactions pattern placode formation and outgrowth. Less is known about the developmental and genetic bases of tooth formation and replacement in polyphyodonts, which are vertebrates with continual tooth replacement. Here, we leverage natural variation in the threespine stickleback fish Gasterosteus aculeatus to investigate the genetic basis of tooth development and replacement. We find that two derived freshwater stickleback populations have both convergently evolved more ventral pharyngeal teeth through heritable genetic changes. In both populations, evolved tooth gain manifests late in development. Using pulse-chase vital dye labeling to mark newly forming teeth in adult fish, we find that both high-toothed freshwater populations have accelerated tooth replacement rates relative to low-toothed ancestral marine fish. Despite the similar evolved phenotype of more teeth and an accelerated adult replacement rate, the timing of tooth number divergence and the spatial patterns of newly formed adult teeth are different in the two populations, suggesting distinct developmental mechanisms. Using genome-wide linkage mapping in marine-freshwater F2 genetic crosses, we find that the genetic basis of evolved tooth gain in the two freshwater populations is largely distinct. Together, our results support a model whereby increased tooth number and an accelerated tooth replacement rate have evolved convergently in two independently derived freshwater stickleback populations using largely distinct developmental and genetic mechanisms.
Collapse
Affiliation(s)
- Nicholas A Ellis
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Andrew M Glazer
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Nikunj N Donde
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Phillip A Cleves
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Rachel M Agoglia
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| | - Craig T Miller
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
75
|
Bioactive nanofibers enable the identification of thrombospondin 2 as a key player in enamel regeneration. Biomaterials 2015; 61:216-28. [PMID: 26004236 DOI: 10.1016/j.biomaterials.2015.05.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/14/2015] [Accepted: 05/18/2015] [Indexed: 12/19/2022]
Abstract
Tissue regeneration and development involves highly synchronized signals both between cells and with the extracellular environment. Biomaterials can be tuned to mimic specific biological signals and control cell response(s). As a result, these materials can be used as tools to elucidate cell signaling pathways and candidate molecules involved with cellular processes. In this work, we explore enamel-forming cells, ameloblasts, which have a limited regenerative capacity. By exposing undifferentiated cells to a self-assembling matrix bearing RGDS epitopes, we elicited a regenerative signal at will that subsequently led to the identification of thrombospondin 2 (TSP2), an extracellular matrix protein that has not been previously recognized as a key player in enamel development and regeneration. Targeted disruption of the thrombospondin 2 gene (Thbs2) resulted in enamel formation with a disordered architecture that was highly susceptible to wear compared to their wild-type counterparts. To test the regenerative capacity, we injected the bioactive matrix into the enamel organ and discovered that the enamel organic epithelial cells in TSP-null mice failed to polarize on the surface of the artificial matrix, greatly reducing integrin β1 and Notch1 expression levels, which represent signaling pathways known to be associated with TSP2. These results suggest TSP2 plays an important role in regulating cell-matrix interactions during enamel formation. Exploiting the signaling pathways activated by biomaterials can provide insight into native signaling mechanisms crucial for tooth development and cell-based strategies for enamel regeneration.
Collapse
|
76
|
Brazil DP, Church RH, Surae S, Godson C, Martin F. BMP signalling: agony and antagony in the family. Trends Cell Biol 2015; 25:249-64. [DOI: 10.1016/j.tcb.2014.12.004] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 01/14/2023]
|
77
|
Olive V, Minella AC, He L. Outside the coding genome, mammalian microRNAs confer structural and functional complexity. Sci Signal 2015; 8:re2. [PMID: 25783159 PMCID: PMC4425368 DOI: 10.1126/scisignal.2005813] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) comprise a class of small, regulatory noncoding RNAs (ncRNAs) with pivotal roles in posttranscriptional gene regulation. Since their initial discovery in 1993, numerous miRNAs have been identified in mammalian genomes, many of which play important roles in diverse cellular processes in development and disease. These small ncRNAs regulate the expression of many protein-coding genes posttranscriptionally, thus adding a substantial complexity to the molecular networks underlying physiological development and disease. In part, this complexity arises from the distinct gene structures, the extensive genomic redundancy, and the complex regulation of the expression and biogenesis of miRNAs. These characteristics contribute to the functional robustness and versatility of miRNAs and provide important clues to the functional significance of these small ncRNAs. The unique structure and function of miRNAs will continue to inspire many to explore the vast noncoding genome and to elucidate the molecular basis for the functional complexity of mammalian genomes.
Collapse
Affiliation(s)
- Virginie Olive
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94705, USA
| | - Alex C Minella
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI 53226, USA
| | - Lin He
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA 94705, USA.
| |
Collapse
|
78
|
Babajko S, de La Dure-Molla M, Jedeon K, Berdal A. MSX2 in ameloblast cell fate and activity. Front Physiol 2015; 5:510. [PMID: 25601840 PMCID: PMC4283505 DOI: 10.3389/fphys.2014.00510] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/08/2014] [Indexed: 11/29/2022] Open
Abstract
While many effectors have been identified in enamel matrix and cells via genetic studies, physiological networks underlying their expression levels and thus the natural spectrum of enamel thickness and degree of mineralization are now just emerging. Several transcription factors are candidates for enamel gene expression regulation and thus the control of enamel quality. Some of these factors, such as MSX2, are mainly confined to the dental epithelium. MSX2 homeoprotein controls several stages of the ameloblast life cycle. This chapter introduces MSX2 and its target genes in the ameloblast and provides an overview of knowledge regarding its effects in vivo in transgenic mouse models. Currently available in vitro data on the role of MSX2 as a transcription factor and its links to other players in ameloblast gene regulation are considered. MSX2 modulations are relevant to the interplay between developmental, hormonal and environmental pathways and in vivo investigations, notably in the rodent incisor, have provided insight into dental physiology. Indeed, in vivo models are particularly promising for investigating enamel formation and MSX2 function in ameloblast cell fate. MSX2 may be central to the temporal-spatial restriction of enamel protein production by the dental epithelium and thus regulation of enamel quality (thickness and mineralization level) under physiological and pathological conditions. Studies on MSX2 show that amelogenesis is not an isolated process but is part of the more general physiology of coordinated dental-bone complex growth.
Collapse
Affiliation(s)
- Sylvie Babajko
- Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, UMRS 1138 Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Descartes Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Diderot Paris, France
| | - Muriel de La Dure-Molla
- Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, UMRS 1138 Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Descartes Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Diderot Paris, France ; Centre de Référence des Maladies Rares de la Face et de la Cavité Buccale MAFACE, Hôpital Rothschild Paris, France
| | - Katia Jedeon
- Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, UMRS 1138 Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Descartes Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Diderot Paris, France
| | - Ariane Berdal
- Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale, UMRS 1138 Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Descartes Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie-Paris Paris, France ; Laboratory of Molecular Oral Pathophysiology, Centre de Recherche des Cordeliers, Université Paris-Diderot Paris, France ; Centre de Référence des Maladies Rares de la Face et de la Cavité Buccale MAFACE, Hôpital Rothschild Paris, France
| |
Collapse
|
79
|
Gao S, Moreno M, Eliason S, Cao H, Li X, Yu W, Bidlack FB, Margolis HC, Baldini A, Amendt BA. TBX1 protein interactions and microRNA-96-5p regulation controls cell proliferation during craniofacial and dental development: implications for 22q11.2 deletion syndrome. Hum Mol Genet 2015; 24:2330-48. [PMID: 25556186 DOI: 10.1093/hmg/ddu750] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
T-box transcription factor TBX1 is the major candidate gene for 22q11.2 deletion syndrome (22q11.2DS, DiGeorge syndrome/Velo-cardio-facial syndrome), whose phenotypes include craniofacial malformations such as dental defects and cleft palate. In this study, Tbx1 was conditionally deleted or over-expressed in the oral and dental epithelium to establish its role in odontogenesis and craniofacial developmental. Tbx1 lineage tracing experiments demonstrated a specific region of Tbx1-positive cells in the labial cervical loop (LaCL, stem cell niche). We found that Tbx1 conditional knockout (Tbx1(cKO)) mice featured microdontia, which coincides with decreased stem cell proliferation in the LaCL of Tbx1(cKO) mice. In contrast, Tbx1 over-expression increased dental epithelial progenitor cells in the LaCL. Furthermore, microRNA-96 (miR-96) repressed Tbx1 expression and Tbx1 repressed miR-96 expression, suggesting that miR-96 and Tbx1 work in a regulatory loop to maintain the correct levels of Tbx1. Cleft palate was observed in both conditional knockout and over-expression mice, consistent with the craniofacial/tooth defects associated with TBX1 deletion and the gene duplication that leads to 22q11.2DS. The biochemical analyses of TBX1 human mutations demonstrate functional differences in their transcriptional regulation of miR-96 and co-regulation of PITX2 activity. TBX1 interacts with PITX2 to negatively regulate PITX2 transcriptional activity and the TBX1 N-terminus is required for its repressive activity. Overall, our results indicate that Tbx1 regulates the proliferation of dental progenitor cells and craniofacial development through miR-96-5p and PITX2. Together, these data suggest a new molecular mechanism controlling pathogenesis of dental anomalies in human 22q11.2DS.
Collapse
Affiliation(s)
- Shan Gao
- Texas A&M University Health Science Center, Houston, TX, USA
| | - Myriam Moreno
- Department of Anatomy and Cell Biology, Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA
| | - Steven Eliason
- Department of Anatomy and Cell Biology, Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA
| | - Huojun Cao
- Texas A&M University Health Science Center, Houston, TX, USA
| | - Xiao Li
- Department of Anatomy and Cell Biology, Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA
| | - Wenjie Yu
- Department of Anatomy and Cell Biology, Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA
| | | | - Henry C Margolis
- Center for Biomineralization, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA, USA and
| | - Antonio Baldini
- Department of Molecular Medicine and Medical Biotechnology, University Federico II and the Institute of Genetics and Biophysics CNR, Naples, Italy
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA,
| |
Collapse
|
80
|
Mitsiadis TA, Filatova A, Papaccio G, Goldberg M, About I, Papagerakis P. Distribution of the amelogenin protein in developing, injured and carious human teeth. Front Physiol 2014; 5:477. [PMID: 25540624 PMCID: PMC4261713 DOI: 10.3389/fphys.2014.00477] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/22/2014] [Indexed: 12/02/2022] Open
Abstract
Amelogenin is the major enamel matrix protein with key roles in amelogenesis. Although for many decades amelogenin was considered to be exclusively expressed by ameloblasts, more recent studies have shown that amelogenin is also expressed in other dental and no-dental cells. However, amelogenin expression in human tissues remains unclear. Here, we show that amelogenin protein is not only expressed during human embryonic development but also in pathological conditions such as carious lesions and injuries after dental cavity preparation. In developing embryonic teeth, amelogenin stage-specific expression is found in all dental epithelia cell populations but with different intensities. In the different layers of enamel matrix, waves of positive vs. negative immunostaining for amelogenin are detected suggesting that the secretion of amelogenin protein is orchestrated by a biological clock. Amelogenin is also expressed transiently in differentiating odontoblasts during predentin formation, but was absent in mature functional odontoblasts. In intact adult teeth, amelogenin was not present in dental pulp, odontoblasts, and dentin. However, in injured and carious adult human teeth amelogenin is strongly re-expressed in newly differentiated odontoblasts and is distributed in the dentinal tubuli under the lesion site. In an in vitro culture system, amelogenin is expressed preferentially in human dental pulp cells that start differentiating into odontoblast-like cells and form mineralization nodules. These data suggest that amelogenin plays important roles not only during cytodifferentiation, but also during tooth repair processes in humans.
Collapse
Affiliation(s)
- Thimios A Mitsiadis
- Orofacial Development and Regeneration Unit, Faculty of Medicine, Institute of Oral Biology, ZZM, University of Zurich Zurich, Switzerland
| | - Anna Filatova
- Orofacial Development and Regeneration Unit, Faculty of Medicine, Institute of Oral Biology, ZZM, University of Zurich Zurich, Switzerland
| | - Gianpaolo Papaccio
- Dipartimento di Medicina Sperimentale, Sezione di Biotecnologie, Istologia Medica e Biologia Molecolare, Seconda Università Degli Studi di Napoli Napoli, Italy
| | - Michel Goldberg
- INSERM UMR-S 1124, Biomédicale des Saints Pères, University Paris Descartes Paris, France
| | - Imad About
- CNRS, Institut des Sciences du Mouvement UMR 7287, Aix-Marseille Université Marseille, France
| | - Petros Papagerakis
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan Ann Arbor, USA ; Center for Organogenesis, School of Medicine, University of Michigan Ann Arbor, USA ; Center for Computational Medicine and Bioinformatics, School of Medicine, University of Michigan Ann Arbor, USA
| |
Collapse
|
81
|
Simmons O, Snider P, Wang J, Schwartz RJ, Chen Y, Conway SJ. Persistent Noggin arrests cardiomyocyte morphogenesis and results in early in utero lethality. Dev Dyn 2014; 244:457-67. [PMID: 25428115 DOI: 10.1002/dvdy.24233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 11/13/2014] [Accepted: 11/16/2014] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Multiple bone morphogenetic protein (BMP) genes are expressed in the developing heart from the initiation to late-differentiation stages, and play pivotal roles in cardiovascular development. In this study, we investigated the requirement of BMP activity in heart development by transgenic over-expression of extracellular BMP antagonist Noggin. RESULTS Using Nkx2.5-Cre to drive lineage-restricted Noggin within cardiomyocyte progenitors, we show persistent Noggin arrests cardiac development at the linear heart stage. This is coupled with a significantly reduced cell proliferation rate, subsequent cardiomyocyte programmed cell death and reduction of downstream intracellular pSMAD1/5/8 expression. Noggin mutants exhibit reduced heartbeat which likely results in subsequent fully penetrant in utero lethality. Significantly, confocal and electron micrographic examination revealed considerably fewer contractile elements, as well as a lack of maturation of actin-myosin microfilaments. Molecular analysis demonstrated that ectopic Noggin-expressing regions in the early heart's pacemaker region, failed to express the potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4 (Hcn4), resulting in an overall decrease in Hcn4 levels. CONCLUSIONS Combined, our results reveal a novel role for BMP signaling in the progression of heart development from the tubular heart stage to the looped stage by means of regulation of proliferation and promotion of maturation of the in utero heart's contractile apparatus and pacemaker.
Collapse
Affiliation(s)
- Olga Simmons
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | | | | | | | | | | |
Collapse
|
82
|
Differentiation of mouse iPS cells into ameloblast-like cells in cultures using medium conditioned by epithelial cell rests of Malassez and gelatin-coated dishes. Med Mol Morphol 2014; 48:138-45. [PMID: 25319805 DOI: 10.1007/s00795-014-0088-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/24/2014] [Indexed: 01/29/2023]
Abstract
Induced pluripotent stem (iPS) cells are generated from adult cells and are potentially of great value in regenerative medicine. Recently, it was shown that iPS cells can differentiate into ameloblast-like cells in cultures using feeder cells. In the present study, we sought to induce differentiation of ameloblast-like cells from iPS cells under feeder-free conditions using medium conditioned by cultured epithelial cell rests of Malassez (ERM) cells and gelatin-coated dishes. Two culture conditions were compared: co-cultures of iPS cells and ERM cells; and, culture of iPS cells in ERM cell-conditioned medium. Differentiation of ameloblast-like cells in the cultures was assessed using real-time RT-PCR assays of expression of the marker genes keratin 14, amelogenin, and ameloblastin and by immunocytochemical staining for amelogenin. We found greater evidence of ameloblast-like cell differentiation in the cultures using the conditioned medium. In the latter, the level of amelogenin expression increased daily and was significantly higher than controls on the 7th, 10th, and 14th days. Expression of ameloblastin also increased daily and was significantly higher than controls on the 14th day. The present study demonstrates that mouse iPS cells can be induced to differentiate into ameloblast-like cells in feeder-free cell cultures using ERM cell-conditioned medium and gelatin-coated dishes.
Collapse
|
83
|
Sharp T, Wang J, Li X, Cao H, Gao S, Moreno M, Amendt BA. A pituitary homeobox 2 (Pitx2):microRNA-200a-3p:β-catenin pathway converts mesenchymal cells to amelogenin-expressing dental epithelial cells. J Biol Chem 2014; 289:27327-27341. [PMID: 25122764 PMCID: PMC4175363 DOI: 10.1074/jbc.m114.575654] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 08/12/2014] [Indexed: 12/21/2022] Open
Abstract
Pitx2, Wnt/β-catenin signaling, and microRNAs (miRs) play a critical role in the regulation of dental stem cells during embryonic development. In this report, we have identified a Pitx2:β-catenin regulatory pathway involved in epithelial cell differentiation and conversion of mesenchymal cells to amelogenin expressing epithelial cells via miR-200a. Pitx2 and β-catenin are expressed in the labial incisor cervical loop or epithelial stem cell niche, with decreased expression in the differentiating ameloblast cells of the mouse lower incisor. Bioinformatics analyses reveal that miR-200a-3p expression is activated in the pre-ameloblast cells to enhance epithelial cell differentiation. We demonstrate that Pitx2 activates miR-200a-3p expression and miR-200a-3p reciprocally represses Pitx2 and β-catenin expression. Pitx2 and β-catenin interact to synergistically activate gene expression during odontogenesis and miR-200a-3p attenuates their expression and directs differentiation. To understand how this mechanism controls cell differentiation and cell fate, oral epithelial and odontoblast mesenchymal cells were reprogrammed by a two-step induction method using Pitx2 and miR-200a-3p. Conversion to amelogenin expressing dental epithelial cells involved an up-regulation of the stem cell marker Sox2 and proliferation genes and decreased expression of mesenchymal markers. E-cadherin expression was increased as well as ameloblast specific factors. The combination of Pitx2, a regulator of dental stem cells and miR-200a converts mesenchymal cells to a fully differentiated dental epithelial cell type. This pathway and reprogramming can be used to reprogram mesenchymal or oral epithelial cells to dental epithelial (ameloblast) cells, which can be used in tissue repair and regeneration studies.
Collapse
Affiliation(s)
- Thad Sharp
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Jianbo Wang
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Xiao Li
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Huojun Cao
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Shan Gao
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas 77030, and
| | - Myriam Moreno
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242,; Craniofacial Anomalies Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242.
| |
Collapse
|
84
|
Wong TS, Gao W, Chan JYW. Interactions between E-cadherin and microRNA deregulation in head and neck cancers: the potential interplay. BIOMED RESEARCH INTERNATIONAL 2014; 2014:126038. [PMID: 25161999 PMCID: PMC4138976 DOI: 10.1155/2014/126038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/07/2014] [Accepted: 07/11/2014] [Indexed: 12/27/2022]
Abstract
E-cadherin expression in the head and neck epithelium is essential for the morphogenesis and homeostasis of epithelial tissues. The cadherin-mediated cell-cell contacts are required for the anchorage-dependent growth of epithelial cells. Further, survival and proliferation require physical tethering created by proper cell-cell adhesion. Otherwise, the squamous epithelial cells will undergo programmed cell death. Head and neck cancers can escape from anoikis and enter into the epithelial-mesenchymal transition stages via the modulation of E-cadherin expression with epigenetic mechanisms. At epigenetic level, gene expression control is not dependent on the DNA sequence. In the context of E-cadherin regulation in head and neck cancers, 2 major mechanisms including de novo promoter hypermethylation and microRNA dysregulation are most extensively studied. Both of them control E-cadherin expression at transcription level and subsequently hinder the overall E-cadherin protein level in the head and neck cancer cells. Increasing evidence suggested that microRNA mediated E-cadherin expression in the head and neck cancers by directly/indirectly targeting the transcription suppressors of E-cadherin, ZEB1 and ZEB2.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wei Gao
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jimmy Yu-Wai Chan
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
85
|
Guan X, Bidlack FB, Stokes N, Bartlett JD. E-cadherin can replace N-cadherin during secretory-stage enamel development. PLoS One 2014; 9:e102153. [PMID: 25014356 PMCID: PMC4094553 DOI: 10.1371/journal.pone.0102153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 06/16/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND N-cadherin is a cell-cell adhesion molecule and deletion of N-cadherin in mice is embryonic lethal. During the secretory stage of enamel development, E-cadherin is down-regulated and N-cadherin is specifically up-regulated in ameloblasts when groups of ameloblasts slide by one another to form the rodent decussating enamel rod pattern. Since N-cadherin promotes cell migration, we asked if N-cadherin is essential for ameloblast cell movement during enamel development. METHODOLOGY/PRINCIPAL FINDINGS The enamel organ, including its ameloblasts, is an epithelial tissue and for this study a mouse strain with N-cadherin ablated from epithelium was generated. Enamel from wild-type (WT) and N-cadherin conditional knockout (cKO) mice was analyzed. μCT and scanning electron microscopy showed that thickness, surface structure, and prism pattern of the cKO enamel looked identical to WT. No significant difference in hardness was observed between WT and cKO enamel. Interestingly, immunohistochemistry revealed the WT and N-cadherin cKO secretory stage ameloblasts expressed approximately equal amounts of total cadherins. Strikingly, E-cadherin was not normally down-regulated during the secretory stage in the cKO mice suggesting that E-cadherin can compensate for the loss of N-cadherin. Previously it was demonstrated that bone morphogenetic protein-2 (BMP2) induces E- and N-cadherin expression in human calvaria osteoblasts and we show that the N-cadherin cKO enamel organ expressed significantly more BMP2 and significantly less of the BMP antagonist Noggin than did WT enamel organ. CONCLUSIONS/SIGNIFICANCE The E- to N-cadherin switch at the secretory stage is not essential for enamel development or for forming the decussating enamel rod pattern. E-cadherin can substitute for N-cadherin during these developmental processes. Bmp2 expression may compensate for the loss of N-cadherin by inducing or maintaining E-cadherin expression when E-cadherin is normally down-regulated. Notably, this is the first demonstration of a natural endogenous increase in E-cadherin expression due to N-cadherin ablation in a healthy developing tissue.
Collapse
Affiliation(s)
- Xiaomu Guan
- Department of Mineralized Tissue Biology and Harvard School of Dental Medicine, The Forsyth Institute, Cambridge, Massachusetts, United States of America
| | - Felicitas B. Bidlack
- Department of Mineralized Tissue Biology and Harvard School of Dental Medicine, The Forsyth Institute, Cambridge, Massachusetts, United States of America
| | - Nicole Stokes
- Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, New York, United States of America
| | - John D. Bartlett
- Department of Mineralized Tissue Biology and Harvard School of Dental Medicine, The Forsyth Institute, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
86
|
Trümbach D, Prakash N. The conserved miR-8/miR-200 microRNA family and their role in invertebrate and vertebrate neurogenesis. Cell Tissue Res 2014; 359:161-77. [PMID: 24875007 DOI: 10.1007/s00441-014-1911-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/05/2014] [Indexed: 01/24/2023]
Abstract
Since their discovery in the early 1990s, microRNAs have emerged as key components of the post-transcriptional regulation of gene expression. MicroRNAs occur in the plant and animal kingdoms, with the numbers of microRNAs encoded in the genome increasing together with the evolutionary expansion of the phyla. By base-pairing with complementary sequences usually located within the 3' untranslated region, microRNAs target mRNAs for degradation, destabilization and/or translational inhibition. Because one microRNA can have many, if not hundreds, of target mRNAs and because one mRNA can, in turn, be targeted by many microRNAs, these small single-stranded RNAs can exert extensive pleiotropic functions during the development, adulthood and ageing of an organism. Specific functions of an increasing number of microRNAs have been described for the invertebrate and vertebrate nervous systems. Among these, the miR-8/miR-200 microRNA family has recently emerged as an important regulator of neurogenesis and gliogenesis and of adult neural homeostasis in the central nervous system of fruit flies, zebrafish and rodents. This highly conserved microRNA family consists of a single ortholog in the fruit fly (miR-8) and five members in vertebrates (miR-200a, miR-200b, miR-200c, miR-141 and miR-429). Here, we review our current knowledge about the functions of the miR-8/miR-200 microRNA family during invertebrate and vertebrate neural development and adult homeostasis and, in particular, about their role in the regulation of neural stem/progenitor cell proliferation, cell cycle exit, transition to a neural precursor/neuroblast state, neuronal differentiation and cell survival and during glial cell growth and differentiation into mature oligodendrocytes.
Collapse
Affiliation(s)
- Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | | |
Collapse
|
87
|
Kim E, Lee M, Li L, Yoon K, Kim K, Jung H. Failure of Tooth Formation Mediated by miR-135a Overexpression via BMP Signaling. J Dent Res 2014; 93:571-5. [DOI: 10.1177/0022034514529303] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 03/04/2014] [Indexed: 12/30/2022] Open
Abstract
MicroRNAs (miRNAs) are known to regulate a variety of gene functions in many tissues and organs, but their expression and function in tooth development are not well-understood. A specific miRNA, miR-135a, was determined to be highly expressed at the bud stage. Interestingly, after the cap stage, miR-135a was expressed in the epithelium and mesenchyme but not in the inner enamel epithelium. To identify the relationship between miR-135a and its putative target genes, Bmpr-Ia and Bmpr-Ib, in early tooth development, miR-135a was ectopically overexpressed with a lentivirus. This overexpression resulted in the repression of Bmpr-Ia and Bmpr-Ib. Furthermore, miR-135a inhibited both Bmpr-Ia and Bmpr-Ib transcription. BMP2 proteins were expressed ectopically in tooth germs during the cap stage to determine the relationship between miR-135a and BMP signaling in early tooth development. When miR-135a was ectopically expressed, no tooth formation was observed after 4 wk of incubation in the kidney capsule. This study suggested that Bmp signaling, specifically Bmpr-Ia and Bmpr-Ib, regulates tooth formation via miR-135a.
Collapse
Affiliation(s)
- E.J. Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - M.J. Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - L. Li
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - K.S. Yoon
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - K.S. Kim
- Graduate School of Biomedical Science and Engineering, College of Medicine, Hanyang University, Seoul, Korea
| | - H.S. Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
- Oral Biosciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR
| |
Collapse
|
88
|
Hu JKH, Mushegyan V, Klein OD. On the cutting edge of organ renewal: Identification, regulation, and evolution of incisor stem cells. Genesis 2014; 52:79-92. [PMID: 24307456 PMCID: PMC4252016 DOI: 10.1002/dvg.22732] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 11/22/2013] [Accepted: 11/25/2013] [Indexed: 12/14/2022]
Abstract
The rodent incisor is one of a number of organs that grow continuously throughout the life of an animal. Continuous growth of the incisor arose as an evolutionary adaptation to compensate for abrasion at the distal end of the tooth. The sustained turnover of cells that deposit the mineralized dental tissues is made possible by epithelial and mesenchymal stem cells residing at the proximal end of the incisor. A complex network of signaling pathways and transcription factors regulates the formation, maintenance, and differentiation of these stem cells during development and throughout adulthood. Research over the past 15 years has led to significant progress in our understanding of this network, which includes FGF, BMP, Notch, and Hh signaling, as well as cell adhesion molecules and micro-RNAs. This review surveys key historical experiments that laid the foundation of the field and discusses more recent findings that definitively identified the stem cell population, elucidated the regulatory network, and demonstrated possible genetic mechanisms for the evolution of continuously growing teeth.
Collapse
Affiliation(s)
- Jimmy Kuang-Hsien Hu
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vagan Mushegyan
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Ophir D. Klein
- Department of Orofacial Sciences and Program in Craniofacial and Mesenchymal Biology, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Pediatrics and Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|