151
|
Sharff KA, Song WX, Luo X, Tang N, Luo J, Chen J, Bi Y, He BC, Huang J, Li X, Jiang W, Zhu GH, Su Y, He Y, Shen J, Wang Y, Chen L, Zuo GW, Liu B, Pan X, Reid RR, Luu HH, Haydon RC, He TC. Hey1 basic helix-loop-helix protein plays an important role in mediating BMP9-induced osteogenic differentiation of mesenchymal progenitor cells. J Biol Chem 2009; 284:649-659. [PMID: 18986983 PMCID: PMC2610517 DOI: 10.1074/jbc.m806389200] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 10/29/2008] [Indexed: 11/06/2022] Open
Abstract
Pluripotent mesenchymal stem cells (MSCs) are bone marrow stromal progenitor cells that can differentiate into osteogenic, chondrogenic, adipogenic, and myogenic lineages. We previously demonstrated that bone morphogenetic protein (BMP) 9 is one of the most potent and yet least characterized BMPs that are able to induce osteogenic differentiation of MSCs both in vitro and in vivo. Here, we conducted gene expression-profiling analysis and identified that Hey1 of the hairy/Enhancer of split-related repressor protein basic helix-loop-helix family was among the most significantly up-regulated early targets in BMP9-stimulated MSCs. We demonstrated that Hey1 expression was up-regulated at the immediate early stage of BMP9-induced osteogenic differentiation. Chromatin immunoprecipitation analysis indicated that Hey1 may be a direct target of the BMP9-induced Smad signaling pathway. Silencing Hey1 expression diminished BMP9-induced osteogenic differentiation both in vitro and in vivo and led to chondrogenic differentiation. Likewise, constitutive Hey1 expression augmented BMP9-mediated bone matrix mineralization. Hey1 and Runx2 were shown to act synergistically in BMP9-induced osteogenic differentiation, and Runx2 expression significantly decreased in the absence of Hey1, suggesting that Runx2 may function downstream of Hey1. Accordingly, the defective osteogenic differentiation caused by Hey1 knockdown was rescued by exogenous Runx2 expression. Thus, our findings suggest that Hey1, through its interplay with Runx2, may play an important role in regulating BMP9-induced osteoblast lineage differentiation of MSCs.
Collapse
Affiliation(s)
- Katie A Sharff
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Wen-Xin Song
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Xiaoji Luo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Ni Tang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Jinyong Luo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Jin Chen
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Yang Bi
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Bai-Cheng He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Jiayi Huang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Xinmin Li
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Wei Jiang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Gao-Hui Zhu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Yuxi Su
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Yun He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Jikun Shen
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Yi Wang
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Liang Chen
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Guo-Wei Zuo
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Bo Liu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Xiaochuan Pan
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Russell R Reid
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637.
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637; Molecular Oncology Laboratory, Department of Surgery, The University of Chicago Medical Center, Chicago, Illinois 60637, the Key Laboratory of Diagnostic Medicine designated by Chinese Ministry of Education and The Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China, the Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California 90095, and the Department of Radiology, The University of Chicago Medical Center, Chicago, Illinois 60637.
| |
Collapse
|
152
|
|
153
|
Regulation of Osteoblast Differentiation by Runx2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 658:43-9. [DOI: 10.1007/978-1-4419-1050-9_5] [Citation(s) in RCA: 308] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
154
|
Kitazawa R, Mori K, Yamaguchi A, Kondo T, Kitazawa S. Modulation of mouse RANKL gene expression by Runx2 and vitamin D3. J Cell Biochem 2008; 105:1289-97. [DOI: 10.1002/jcb.21929] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
155
|
The Upregulation of Osteoblast Marker Genes in Mesenchymal Stem Cells Prove the Osteoinductivity of Hydroxyapatite/Tricalcium Phosphate Biomaterial. Transplant Proc 2008; 40:2645-8. [DOI: 10.1016/j.transproceed.2008.07.096] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
156
|
Guillotin B, Bareille R, Bourget C, Bordenave L, Amédée J. Interaction between human umbilical vein endothelial cells and human osteoprogenitors triggers pleiotropic effect that may support osteoblastic function. Bone 2008; 42:1080-91. [PMID: 18387350 DOI: 10.1016/j.bone.2008.01.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 01/24/2008] [Accepted: 01/27/2008] [Indexed: 01/01/2023]
Abstract
Osteogenesis occurs in striking interaction with angiogenesis. There is growing evidence that endothelial cells are involved in the modulation of osteoblast differentiation. We hypothesized that primary human umbilical vein endothelial cells (HUVEC) should be able to modulate primary human osteoprogenitors (HOP) function in an in vitro co-culture model. In a previous study we demonstrated that a 3 day to 3 week co-culture stimulates HOP differentiation markers such as Alkaline Phosphatase (ALP) activity and mineralization. In the present study we addressed the effects induced by the co-culture on HOP within the first 48 hours. As a prerequisite, we validated a method based on immuno-magnetic beads to separate HOP from HUVEC after co-culture. Reverse transcription-real time quantitative PCR studies demonstrated up-regulation of the ALP expression in the co-cultured HOP, confirming previous results. Surprisingly, down-regulation of runx2 and osteocalcin was also shown. Western blot analysis revealed co-culture induced down-regulation of Connexin43 expression in both cell types. Connexin43 function may be altered in co-cultured HOP as well. Stimulation of the cAMP pathway was able to counterbalance the effect of the co-culture on the ALP activity, but was not able to rescue runx2 mRNA level. Co-culture effect on HOP transcriptome was analyzed with GEArray cDNA microarray showing endothelial cells may also modulate HOP extracellular matrix production. In accordance with previous work, we propose endothelial cells may support initial osteoblastic proliferation but do not alter the ability of the osteoblasts to produce extracellular mineralizing matrix.
Collapse
Affiliation(s)
- B Guillotin
- Laboratoire CIRID, UMR 5164 CNRS, Université Victor Segalen Bordeaux2, 146 rue Léo Saignat, 33076 Bordeaux, France.
| | | | | | | | | |
Collapse
|
157
|
Flow perfusion culture of human mesenchymal stem cells on silicate-substituted tricalcium phosphate scaffolds. Biomaterials 2008; 29:2616-27. [DOI: 10.1016/j.biomaterials.2008.03.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 03/04/2008] [Indexed: 11/18/2022]
|
158
|
Frith J, Genever P. Transcriptional control of mesenchymal stem cell differentiation. TRANSFUSION MEDICINE AND HEMOTHERAPY : OFFIZIELLES ORGAN DER DEUTSCHEN GESELLSCHAFT FUR TRANSFUSIONSMEDIZIN UND IMMUNHAMATOLOGIE 2008; 35:216-27. [PMID: 21547119 DOI: 10.1159/000127448s] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Accepted: 01/31/2008] [Indexed: 12/23/2022]
Abstract
SUMMARY In recent years, transcriptomics and proteomics have provided us with a great deal of information about the expression profiles of various cell types and how these change under different conditions. Stem cell research is one area where this has had a major impact by providing an insight into events at the molecular level that control stem cell growth and differentiation. This includes mesenchymal stem cell (MSC) biology where knowledge about the mechanisms governing differentiation is vital for the development of future therapeutic strategies. Although there is still much to learn, we are starting to build up a picture of the main events in these differentiation processes. This review will discuss control of MSC differentiation at the transcriptional level. Not all the factors which have been shown to play a role in lineage-specific mesenchymal differentiation can be covered here. Instead, we will focus specifically on the key factors that contribute to the regulation of osteogenesis, adipogenesis, and chondrogenesis.
Collapse
Affiliation(s)
- Jess Frith
- Department of Biology (Area 9), University of York, UK
| | | |
Collapse
|
159
|
Hu Y, Papagerakis P, Ye L, Feng JQ, Simmer JP, Hu JCC. Distal cis-regulatory elements are required for tissue-specific expression of enamelin (Enam). Eur J Oral Sci 2008; 116:113-23. [PMID: 18353004 DOI: 10.1111/j.1600-0722.2007.00519.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Enamel formation is orchestrated by the sequential expression of genes encoding enamel matrix proteins; however, the mechanisms sustaining the spatio-temporal order of gene transcription during amelogenesis are poorly understood. The aim of this study was to characterize the cis-regulatory sequences necessary for normal expression of enamelin (Enam). Several enamelin transcription regulatory regions, showing high sequence homology among species, were identified. DNA constructs containing 5.2 or 3.9 kb regions upstream of the enamelin translation initiation site were linked to a LacZ reporter and used to generate transgenic mice. Only the 5.2-Enam-LacZ construct was sufficient to recapitulate the endogenous pattern of enamelin tooth-specific expression. The 3.9-Enam-LacZ transgenic lines showed no expression in dental cells, but ectopic beta-galactosidase activity was detected in osteoblasts. Potential transcription factor-binding sites were identified that may be important in controlling enamelin basal promoter activity and in conferring enamelin tissue-specific expression. Our study provides new insights into regulatory mechanisms governing enamelin expression.
Collapse
Affiliation(s)
- Yuanyuan Hu
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI 48108, USA
| | | | | | | | | | | |
Collapse
|
160
|
Tamiya H, Ikeda T, Jeong JH, Saito T, Yano F, Jung YK, Ohba S, Kawaguchi H, Chung UI, Choi JY. Analysis of the Runx2 promoter in osseous and non-osseous cells and identification of HIF2A as a potent transcription activator. Gene 2008; 416:53-60. [PMID: 18442887 DOI: 10.1016/j.gene.2008.03.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 02/21/2008] [Accepted: 03/04/2008] [Indexed: 11/16/2022]
Abstract
Little is known about the upstream regulator of Runx2, a master regulator of osteoblast differentiation in bone tissues. To elucidate the molecular mechanism of Runx2 gene expression, we analyzed Runx2 promoter activity in osseous (MC3T3-E1, KS483, Kusa) and non-osseous (NIH3T3, C3H10T1/2, mouse embryonic fibroblasts) cells and also identified Runx2 upstream regulator using a Runx2 promoter-derived luciferase reporter system. After cloning 15 serial deletion constructs from -6832 bp/+390 bp to -37 bp/+390 bp of the Runx2-P1 promoter, we performed a transient transfection assay in osseous and non-osseous cells. A reduction in Runx2 promoter activity was observed in two regions; one was between -3 kb and -1 kb, and the other was between -155 bp and -75 bp. The step-down pattern in promoter activity between -3 kb and -1 kb was observed only in osseous cells. Interestingly, the step-down pattern between -155 bp and -75 bp was revealed in both cell types. Consistently, beta-galactosidase staining in axial skeleton of -3 kb-Runx2-P1-LacZ transgenic mice was positive, but that of all skeletal tissues of -1 kb-Runx2-P1-LacZ transgenic mice was negative. To identify upstream regulators of the Runx2-P1 promoter, we screened 100 transcription factors using Runx2-P1-luciferase reporter constructs in NIH3T3 fibroblasts and HeLa cells. Among them, HIF2A was identified as the strongest activator of Runx2-P1 promoter activity. A HIF2A-responsive site on the Runx2 promoter was identified between -106 bp and -104 bp by mutation analysis. An electrophoretic mobility shift assay and chromatin immunoprecipitation assay confirmed the binding of HIF2A to the Runx2-P1 promoter in vitro and in vivo, respectively. Knock-down using siRNA against HIF2A confirmed that HIF2A is an important regulator of Runx2 gene expression. Collectively, these results suggest that the region between -3 kb and -1 kb is required for the minimal skeletal tissue-specific expression of Runx2, and that the region between -155 bp and -75 bp is important for its basal transcription, which may be in part mediated by HIF2A in bone tissues.
Collapse
Affiliation(s)
- Hiroyuki Tamiya
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, the University of Tokyo, Tokyo, 113-8655, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Kishiya M, Sawada T, Kanemaru K, Kudo H, Numasawa T, Yokoyama T, Tanaka S, Motomura S, Ueyama K, Harata S, Toh S, Furukawa KI. A functional RNAi screen for Runx2-regulated genes associated with ectopic bone formation in human spinal ligaments. J Pharmacol Sci 2008; 106:404-14. [PMID: 18319563 DOI: 10.1254/jphs.fp0072043] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Ossification of the posterior longitudinal ligament of the spine (OPLL) is characterized by ectopic ossification in the spinal ligaments, which enlarges with time and compresses the spinal cord, resulting in serious neurological symptoms. We previously reported that Runx2 expression was enhanced in spinal ligament cells from OPLL patients (OPLL cells). To clarify genes regulated by Runx2, Runx2 expression was first enhanced by culturing primary OPLL cells in osteogenic medium (OS induction) and then inhibited by siRNAs targeted to Runx2. DNA microarray demonstrated that in addition to chondrogenic factors such as connective tissue growth factor and cartilage oligomeric matrix protein, angiopoietin-1 was also significantly increased by OS induction and decreased by siRNAs for Runx2 in OPLL cells, suggesting that these genes are regulated by Runx2. However, these changes were not observed in non-OPLL control cells (from cervical spondylotic myelopathy patients). Furthermore, Runx2 was not decreased by siRNAs for angiopoietin-1. OS induction and RNAi inhibition of angiopoietin-1 expression was also observed in osteoblasts. Both siRNAs for Runx2 and angiopoietin-1 completely inhibited aggrecan-1 expression. These results suggest that angiopoietin-1 is downstream of Runx2 in both OPLL primary cells and osteoblasts. Angiopoietin-1 may play an important role in ectopic ossification.
Collapse
Affiliation(s)
- Masaki Kishiya
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Abstract
The bone marrow mesenchymal compartment contains putative stem/progenitors of skeletal tissue components such as bone, cartilage, haematopoiesis-supporting stroma and adipocytes. Previously appreciated as vital to the support of haematopoiesis, these cells have also been recently recognized as having significant immunomodulatory properties with implications for allogeneic haematopoietic cell transplantation. Despite having been studied for more than three decades and currently being used in different clinical settings, their biology remains elusive. The aim of this review is to critically analyse the field of mesenchymal stem/progenitor cell biology, in respect of their relationship with other mesenchymal cell-types. Several issues concerning lineage commitment and inter-conversion potential between different mesenchymal cell-types are reviewed.
Collapse
Affiliation(s)
- Fernando Anjos-Afonso
- Haematopoietic Stem Cell Laboratory, Cancer Research UK, London Research Institute, London, UK
| | | |
Collapse
|
163
|
Sun S, Wang Z, Hao Y. Osterix overexpression enhances osteoblast differentiation of muscle satellite cells in vitro. Int J Oral Maxillofac Surg 2008; 37:350-6. [PMID: 18272339 DOI: 10.1016/j.ijom.2007.11.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2007] [Revised: 09/15/2007] [Accepted: 11/28/2007] [Indexed: 11/30/2022]
Abstract
Muscle satellite cells have long been considered a distinct myogenic lineage responsible for postnatal growth, repair and maintenance of skeletal muscle. Recent studies have demonstrated that they are multi-potential. Osterix (Osx), a novel zinc-finger-containing transcription factor of the sp family, is required for osteoblast differentiation and bone formation. It was hypothesized that Osx overexpression would enhance osteoblast differentiation of muscle satellite cells in vitro. Recombinant adenovirus-mediated Osx gene (Ad-Osx) was constructed and used to transfect muscle satellite cells. Osx overexpression inhibited myogenesis, as demonstrated by suppression of MyoD and myogenin mRNA levels and reduced myotube formation. Muscle satellite cells transduced with Ad-Osx exhibited apparent osteoblast differentiation as determined by the expression of related osteoblastic genes, increased activity of alkaline phosphatase and the formation of mineralized nodules. These results confirmed the ability of Osx to enhance osteoblast differentiation of muscle satellite cells in vitro, and the competence of muscle satellite cells as promising seed cells for bone tissue engineering.
Collapse
Affiliation(s)
- S Sun
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Tongji University, Shanghai, China
| | | | | |
Collapse
|
164
|
Frith J, Genever P. Transcriptional Control of Mesenchymal Stem Cell Differentiation. Transfus Med Hemother 2008. [DOI: 10.1159/000127448] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
165
|
Zou L, Zou X, Chen L, Li H, Mygind T, Kassem M, Bünger C. Multilineage differentiation of porcine bone marrow stromal cells associated with specific gene expression pattern. J Orthop Res 2008; 26:56-64. [PMID: 17676606 DOI: 10.1002/jor.20467] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
There are increasing reports regarding differentiation of bone marrow stromal cells (BMSC) from human and various species of animals including pigs. The phenotype and function of BMSC along a mesenchymal lineage differentiation are well characterized by specific transcription factors and marker genes. However, it is not fully clear whether multilineage differentiation (osteogenesis, chondrogenesis, and adipogenesis) of BMSC is associated with a specific gene expression pattern. In the present study, we investigated the gene expression pattern of representative transcription factors and marker genes along those three mesenchymal lineages during a particular lineage differentiation of porcine BMSC by means of real-time PCR measurement. In an osteogenic medium, the mRNA levels of cbfa1, osterix, alkaline phosphatase, type 1 collagen, osteonectin, bone sialoprotein, and osteocalcin were induced stepwise. Meanwhile, sox9, specific to chondrogenic differentiation, was inhibited but not PPARgamma2 specific to adipogenic differentiation. In an adipogenic medium, adipogenic differentiation was confirmed by upregulation of PPARgamma2 and aP2 and downregulation of osteogenic genes and sox9. Chondrogenic differentiation was induced in cell pellet culture by expression of sox9, type 2 collagen, and aggrecan. Cbfa1 and PPARgamma2 were inhibited in chondrogenic medium. These results indicate that the differentiation potential of BMSC to a particular mesenchymal lineage relies upon specific gene expression pattern, namely upregulation of genes specific for this lineage and suppression of other lineage differentiation.
Collapse
Affiliation(s)
- Lijin Zou
- Orthopaedic Research Laboratory, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
166
|
Maehira F, Iinuma Y, Eguchi Y, Miyagi I, Teruya S. Effects of soluble silicon compound and deep-sea water on biochemical and mechanical properties of bone and the related gene expression in mice. J Bone Miner Metab 2008; 26:446-55. [PMID: 18758902 DOI: 10.1007/s00774-007-0845-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 11/20/2007] [Indexed: 10/14/2022]
Abstract
Silicon has been known as an essential element for bone formation. The silicon contents of sea water increase with increasing of depth: 1.8 ppm Si in deep-sea water (DW) at 612 m in depth versus 0.06 ppm in surface sea water (SW). The effects of soluble silicon (Si) and DW from which NaCl was eliminated were studied in comparison with tap water (TW) and SW in cell cultures and in animal experiments using the control strain of senescence accelerated mouse, SAMR1. Si at 10 ppm as sodium metasilicate or 10% DW in the alpha-MEM medium stimulated cellular viability, marker enzymes of osteoblast and osteoclast cell lines, and the (45)CaCl(2) uptake in those cells in comparison with the medium control. After weanling SAMR1 were maintained for 6 months on a diet containing 200 ppm Si and 39% of DW and SW, DW and Si improved bone biochemical indices such as femoral weight, mineral and collagen content, and marker enzymes of bone formation and resorption as well as mechanical properties as compared to TW. In the femoral bone marrow of SAMR1, the mRNA expression of bone morphogenetic protein-2 (BMP-2), interleukin-11 (IL-11), and runt-related transcription factor 2 (Runx 2), which stimulate osteoblast development as well as type I procollagen (COL1A1) mRNA, were significantly increased in both DW and Si groups. The expressions of both osteoprotegerin (OPG) and receptor activator of NF-kappaB ligand (RANKL) were also elevated, resulting in distinct increases of the OPG/RANKL ratio in both DW and Si groups. The results indicated that a soluble silicate and deep-sea water as its natural material stimulated cell growth in both osteoblasts and osteoclasts in cell culture and promoted bone metabolic turnover in favor of bone formation through stimulation of the related mRNA expression in animal experiments.
Collapse
Affiliation(s)
- Fusako Maehira
- Laboratory of Biometabolic Chemistry, School of Health Sciences, Faculty of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa, Japan.
| | | | | | | | | |
Collapse
|
167
|
Zou L, Zou X, Li H, Mygind T, Zeng Y, Lü N, Bünger C. Molecular mechanism of osteochondroprogenitor fate determination during bone formation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 585:431-41. [PMID: 17120800 DOI: 10.1007/978-0-387-34133-0_28] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Osteoblasts and chondrocytes, which derive from a common mesenchymal precursor (osteochondroprogenitor), are involved in bone formation and remodeling in vivo. Determination of osteochondroprogenitor fate is under the control of complex hormonal and local factors converging onto a series of temporospatial dependent transcription regulators. Sox9, together with L-Sox5 and Sox6, of the Sox family is required for chondrogenic differentiation commitment, while Runx2/Cbfa 1, a member of runt family and Osterix/Osx, a novel zinc finger-containing transcription factor play a pivotal role in osteoblast differentiation decision and hypertrophic chondrocyte maturation. Recent in vitro and in vivo evidence suggests beta-catenin, a transcriptional activator in the canonical Wnt pathway, can act as a determinant factor for controlling chondrocyte and osteoblast differentiation. Here we focus on several intensively studied transcription factors and Wnt/beta-catenin signal molecules to illustrate the regulatory mechanism in directing commitment between osteoblast and chondrocyte, which will eventually allow us to properly manipulate the mesenchymal progenitor cell differentiation on bone and regeneration of cartilage tissue engineering.
Collapse
Affiliation(s)
- Lijin Zou
- Orthopaedic Research Laboratory, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | | | | | | | | | | | | |
Collapse
|
168
|
Maruyama Z, Yoshida CA, Furuichi T, Amizuka N, Ito M, Fukuyama R, Miyazaki T, Kitaura H, Nakamura K, Fujita T, Kanatani N, Moriishi T, Yamana K, Liu W, Kawaguchi H, Nakamura K, Komori T. Runx2 determines bone maturity and turnover rate in postnatal bone development and is involved in bone loss in estrogen deficiency. Dev Dyn 2007; 236:1876-90. [PMID: 17497678 DOI: 10.1002/dvdy.21187] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Runx2 is an essential transcription factor for osteoblast differentiation. However, the functions of Runx2 in postnatal bone development remain to be clarified. Introduction of dominant-negative (dn)-Runx2 did not inhibit Col1a1 and osteocalcin expression in mature osteoblastic cells. In transgenic mice that expressed dn-Runx2 in osteoblasts, the trabecular bone had increased mineralization, increased volume, and features of compact bone, and the expression of major bone matrix protein genes was relatively maintained. After ovariectomy, neither osteolysis nor bone formation was enhanced and bone was relatively conserved. In wild-type mice, Runx2 was strongly expressed in immature osteoblasts but downregulated during osteoblast maturation. These findings indicate that the maturity and turnover rate of bone are determined by the level of functional Runx2 and Runx2 is responsible for bone loss in estrogen deficiency, but that Runx2 is not essential for maintenance of the expression of major bone matrix protein genes in postnatal bone development and maintenance.
Collapse
Affiliation(s)
- Zenjiro Maruyama
- Department of Cell Biology, Unit of Basic Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Mikami Y, Omoteyama K, Kato S, Takagi M. Inductive effects of dexamethasone on the mineralization and the osteoblastic gene expressions in mature osteoblast-like ROS17/2.8 cells. Biochem Biophys Res Commun 2007; 362:368-73. [PMID: 17707772 DOI: 10.1016/j.bbrc.2007.07.192] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 07/30/2007] [Indexed: 10/23/2022]
Abstract
We examined the effects of dexamethasone (Dex), a synthetic glucocorticoid, on the formation of mineralized bone nodules and the gene expressions of the late osteoblastic markers, bone sialoprotein (BSP), osteocalcin (OC), and osteopontin (OPN) in mature osteoblast ROS17/2.8 cells. Treatment of ROS17/2.8 cells with Dex resulted in the induction of mineralization accompanied with increasing BSP and OC expressions. Previous reports have demonstrated that BSP and OC expressions are regulated by Runx2. Then, we hypothesized that Dex might promote osteoblastic differentiation and mineralization on ROS17/2.8 by Runx2. In this study, no effect was observed in mRNA and protein expression of Runx2. However, the transcriptional activity of Runx2 was enhanced by Dex treatment. Furthermore, the Dex-induced BSP and OC expressions decreased after the transfection of Runx2 small-interfering RNAs (siRNAs). These results suggested that the enhancement of Runx2 transcriptional activity by Dex treatment may be followed by the activation of osteoblast marker genes, such as BSP and OC to thereby produce a bone-specific matrix that subsequently becomes mineralized.
Collapse
Affiliation(s)
- Yoshikazu Mikami
- Department of Anatomy, Division of Functional Morphology, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-surugadai, Chiyoda-ku, Tokyo 101-8310, Japan.
| | | | | | | |
Collapse
|
170
|
Tu Q, Zhang J, James L, Dickson J, Tang J, Yang P, Chen J. Cbfa1/Runx2-deficiency delays bone wound healing and locally delivered Cbfa1/Runx2 promotes bone repair in animal models. Wound Repair Regen 2007; 15:404-12. [PMID: 17537128 PMCID: PMC2837142 DOI: 10.1111/j.1524-475x.2007.00243.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Core binding factor 1 (Cbfa1)/runt-related transcription factor 2 (Runx2) has been identified as a "master gene" in osteoblastic differentiation. In this two-part study, part I of the study was undertaken to test the hypothesis that bone regeneration is compromised in Cbfa1+/- mice. Compared with wild-type mice, wound healing was dramatically delayed in Cbfa1+/- mice characterized by the presence of a small amount of bone near the base of the wounds. The bone defects were largely filled with fibrous connective tissues 3 weeks after surgery. Part II was performed to determine the effects of Cbfa1 in enhancing bone wound healing using a gene-activated matrix (GAM) method. Cbfa1 cDNA was mixed with a biodegradable bovine type I collagen sponge and was inserted into the periodontal window wounds of mice. Control sponges were collagen matrix without Cbfa1 cDNA. Histological analysis and immunohistochemical staining demonstrated that compared with controls, there was increased new bone formation that almost filled the wound defects 14 days after surgery in the Cbfa1-GAM group. The collagen sponge matrix did not seem to elicit significant foreign body reaction in either group. In conclusion, the reduced expression of Cbfa1 interferes with the process of bone wound healing, and local application of Cbfa1 cDNA incorporated into a collagen matrix promotes bone tissue regeneration.
Collapse
Affiliation(s)
- Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, Massachusetts
| | - Jin Zhang
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, Massachusetts
- School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Laji James
- Department of Pediatric Dentistry, University of Texas Health Science Center at San Antonio, Texas
| | - Julia Dickson
- Department of Pediatric Dentistry, University of Texas Health Science Center at San Antonio, Texas
| | - Jean Tang
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, Massachusetts
| | - Pishan Yang
- School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
171
|
Huang W, Yang S, Shao J, Li YP. Signaling and transcriptional regulation in osteoblast commitment and differentiation. FRONT BIOSCI-LANDMRK 2007; 12:3068-92. [PMID: 17485283 PMCID: PMC3571113 DOI: 10.2741/2296] [Citation(s) in RCA: 441] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The major event that triggers osteogenesis is the transition of mesenchymal stem cells into bone forming, differentiating osteoblast cells. Osteoblast differentiation is the primary component of bone formation, exemplified by the synthesis, deposition and mineralization of extracellular matrix. Although not well understood, osteoblast differentiation from mesenchymal stem cells is a well-orchestrated process. Recent advances in molecular and genetic studies using gene targeting in mouse enable a better understanding of the multiple factors and signaling networks that control the differentiation process at a molecular level. Osteoblast commitment and differentiation are controlled by complex activities involving signal transduction and transcriptional regulation of gene expression. We review Wnt signaling pathway and Runx2 regulation network, which are critical for osteoblast differentiation. Many other factors and signaling pathways have been implicated in regulation of osteoblast differentiation in a network manner, such as the factors Osterix, ATF4, and SATB2 and the TGF-beta, Hedgehog, FGF, ephrin, and sympathetic signaling pathways. This review summarizes the recent advances in the studies of signaling transduction pathways and transcriptional regulation of osteoblast cell lineage commitment and differentiation. The knowledge of osteoblast commitment and differentiation should be applied towards the development of new diagnostic and therapeutic alternatives for human bone diseases.
Collapse
Affiliation(s)
- Wei Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Shuying Yang
- Department of Cytokine Biology, Forsyth Institute, Harvard School of Dental Medicine, Boston, Massachusetts
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Jianzhong Shao
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Ping Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Department of Cytokine Biology, Forsyth Institute, Harvard School of Dental Medicine, Boston, Massachusetts
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
172
|
Lambertini E, Penolazzi L, Tavanti E, Schincaglia GP, Zennaro M, Gambari R, Piva R. Human estrogen receptor alpha gene is a target of Runx2 transcription factor in osteoblasts. Exp Cell Res 2007; 313:1548-60. [PMID: 17350616 DOI: 10.1016/j.yexcr.2007.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2007] [Revised: 01/29/2007] [Accepted: 02/01/2007] [Indexed: 11/18/2022]
Abstract
Several studies into the mechanisms involved in control of osteoblast-specific gene expression have identified Runx2 and ERalpha (estrogen receptor alpha) as essential regulators of osteoblast differentiation. Recently, interactions between Runx2 and ERalpha have been described. Here, we investigate the role of Runx2 on the regulation of ERalpha expression by determining its interaction with the F promoter, one of the multiple promoters of the human ERalpha gene and the only one active in bone. We found that, in this promoter, three Runx2-like sites are present. By electrophoretic mobility shift assay in combination with supershift and ChIP experiments, we demonstrated that Runx2 preferentially binds one of the Runx2 motifs of the F promoter. To understand whether or not they are involved in influencing F promoter activity, different promoter-reporter deletion and mutation constructs were transiently transfected into human osteoblastic cells. Comparison of luciferase activities allowed the identification of a prevalent negative role of a sequence context, within the -117,877/-117,426 region, which may be under the control of Runx2 (a) site. Finally, silencing and overexpression of endogenous Runx2 provided evidence that Runx2 has a more complex role than initially expected. In fact, Runx2 (a) and Runx2 (b) sites carried out opposite roles which are conditioned by Runx2 levels in bone cells. Therefore, the resulting F promoter activity may be tightly regulated by a dynamic interplay between these two Runx2 sites, with a predominance of negative effect of the Runx2 (a) site.
Collapse
Affiliation(s)
- Elisabetta Lambertini
- Department of Biochemistry and Molecular Biology, Molecular Biology Section, Ferrara University, Via Fossato di Mortara, 74, 44100 Ferrara, Italy
| | | | | | | | | | | | | |
Collapse
|
173
|
Wang Q, Wei X, Zhu T, Zhang M, Shen R, Xing L, O’Keefe RJ, Chen D. Bone morphogenetic protein 2 activates Smad6 gene transcription through bone-specific transcription factor Runx2. J Biol Chem 2007; 282:10742-8. [PMID: 17215250 PMCID: PMC2636961 DOI: 10.1074/jbc.m610997200] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BMP-2 plays an essential role in osteoblast and chondrocyte differentiation, but its signaling mechanism has not been fully defined. In the present studies, we investigated the mechanism through which BMP-2 activates the Smad6 gene. A -2006/+45 Smad6 promoter-luciferase construct was generated along with deletions and Runx2 binding site mutations to examine the role of Smad1 and Runx2 signaling following BMP-2 stimulation in osteoblasts. Transfection of Runx2 or treatment with BMP-2-stimulated promoter activity of the -2006/+45 and -1191/+45 reporters but not the -829/+45 and -374/+45 reporters. No Smad1/5 binding site is present in the -1191/-829 region of the Smad6 promoter. Mutation of the OSE2-a site (-1036/-1031) completely abolished the stimulatory effect of Runx2 as well as BMP-2 on the -2006/+45 and -1191/+45 Smad6 reporters. Gel shift and chromatin immunoprecipitation (ChIP) assays showed that Runx2 binds the OSE2-a element. ChIP assays demonstrated that Smad1 also interacts with the OSE2-a site at the Smad6 promoter through Runx2. The protein degradation of Runx2 is mediated by the E3 ubiquitin ligase Smurf1. In the present studies, we found that Smurf1 binds the OSE2-a site through Runx2 and inhibits Smad6 gene transcription. Treatment with BMP-2 and transfection of Smad1 abolished Smurf1 binding to the OSE2 site. These results show that Smad1 binding excludes Smurf1 interaction with the OSE2 site and promotes Smad6 gene transcription.
Collapse
Affiliation(s)
- Qing Wang
- Medical College, Nankai University, Tianjin 300071, China
| | - Xiaochao Wei
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, New York 14642
| | - Tianhui Zhu
- Medical College, Nankai University, Tianjin 300071, China
| | - Ming Zhang
- Medical College, Nankai University, Tianjin 300071, China
| | - Run Shen
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, New York 14642
| | - Lianping Xing
- Department of Pathology, University of Rochester School of Medicine, Rochester, New York 14642
| | - Regis J. O’Keefe
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, New York 14642
| | - Di Chen
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, New York 14642
- To whom correspondence should be addressed: Dept. of Orthopaedics, University of Rochester Medical Center, Rochester, NY 14642. Fax: 585-275-1121; E-mail:
| |
Collapse
|
174
|
Foster BL, Popowics TE, Fong HK, Somerman MJ. Advances in defining regulators of cementum development and periodontal regeneration. Curr Top Dev Biol 2007; 78:47-126. [PMID: 17338915 DOI: 10.1016/s0070-2153(06)78003-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Substantial advancements have been made in defining the cells and molecular signals that guide tooth crown morphogenesis and development. As a result, very encouraging progress has been made in regenerating crown tissues by using dental stem cells and recombining epithelial and mesenchymal tissues of specific developmental ages. To date, attempts to regenerate a complete tooth, including the critical periodontal tissues of the tooth root, have not been successful. This may be in part due to a lesser degree of understanding of the events leading to the initiation and development of root and periodontal tissues. Controversies still exist regarding the formation of periodontal tissues, including the origins and contributions of cells, the cues that direct root development, and the potential of these factors to direct regeneration of periodontal tissues when they are lost to disease. In recent years, great strides have been made in beginning to identify and characterize factors contributing to formation of the root and surrounding tissues, that is, cementum, periodontal ligament, and alveolar bone. This review focuses on the most exciting and important developments over the last 5 years toward defining the regulators of tooth root and periodontal tissue development, with special focus on cementogenesis and the potential for applying this knowledge toward developing regenerative therapies. Cells, genes, and proteins regulating root development are reviewed in a question-answer format in order to highlight areas of progress as well as areas of remaining uncertainty that warrant further study.
Collapse
Affiliation(s)
- Brian L Foster
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, Washington 98195, USA
| | | | | | | |
Collapse
|
175
|
Pregizer S, Barski A, Gersbach CA, García AJ, Frenkel B. Identification of novel Runx2 targets in osteoblasts: Cell type-specific BMP-dependent regulation of Tram2. J Cell Biochem 2007; 102:1458-71. [PMID: 17486635 DOI: 10.1002/jcb.21366] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Runx2 is an osteoblast master transcription factor and a target for bone morphogenetic protein (BMP) signaling, but our knowledge of events downstream of Runx2 is limited. In this study, we used ChIP Display to discover seven novel genomic regions occupied by Runx2 in living MC3T3-E1 osteoblastic cells. Six of these regions are found within or up to 1-kb away from annotated genes, but only two are found within 5'-gene flanking sequences. One of the newly identified Runx2 target genes is Tram2, whose product facilitates proper folding of type I collagen. We demonstrate that Tram2 mRNA is suppressed in non-osteoblasts when Runx2 is over-expressed, and that this suppression is alleviated upon treatment with BMP-2. Moreover, we show that BMP-induced Runx2 expression in the C3H10T1/2, ST2, C2C12, and MC3T3-E1 cell lines coincides with an increase in Tram2 mRNA levels. Thus, Runx2 may regulate Tram2 expression in a BMP-dependent manner, and Tram2 may participate in the overall osteogenic function of Runx2. Among the other Runx2 target genes discovered in this study are Lnx2, an intracellular scaffolding protein that may play a role in Notch signaling, and Tnfrsf12a, a Tumor Necrosis Factor receptor family member that influences both osteoblast and osteoclast differentiation. Expanding our knowledge of Runx2 target genes, and manipulation of these genes, are warranted to better understand the regulation of osteoblast function and to provide opportunities for the development of new bone anabolics.
Collapse
Affiliation(s)
- Steven Pregizer
- Department of Biochemistry & Molecular Biology, Institute for Genetic Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | |
Collapse
|
176
|
Mori K, Kitazawa R, Kondo T, Maeda S, Yamaguchi A, Kitazawa S. Modulation of mouse RANKL gene expression by Runx2 and PKA pathway. J Cell Biochem 2006; 98:1629-44. [PMID: 16598781 DOI: 10.1002/jcb.20891] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Runx2 regulates the target genes characteristic of osteoblastic phenotypes, while exerting diverse and sometimes controversial effects on osteoblastic cells depending on their differentiation stage. Receptor activator of nuclear factor-kappaB (RANK) ligand (RANKL) is a membrane bound cytokine essential for osteo(chondro)clastogenesis. During endochondral ossification, while Runx2-positive hypertrophic chondrocytes express RANKL, the steady-state expression of the RANKL gene in osteoblastic cells is, at later stages, kept at a relatively low level to sustain the established bone. The aim of this study was to elucidate the mechanism whereby Runx2 and the protein kinase A (PKA) pathway modulate RANKL expression, especially from the viewpoint of their functions in RANKL basic promoter activity and in chromatin structural changes in osteoblastic/stromal cells. Osteoblastic/stromal cell lines derived from normal and Runx2-deficient mice were used to analyze endogenous RANKL gene expression by real-time reverse transcription (RT)-PCR, the acetylation status of the H3 and H4 histone proteins associated with the 5'-flanking region of the RANKL gene by chromatin immunoprecipitation, and the exogenously transfected RANKL gene promoter activity both in the steady-state and under PKA-activated conditions. Here, we demonstrate that Runx2 suppresses steady-state RANKL gene expression by condensing chromatin, while showing a slightly positive effect on RANKL basic promoter activity. Besides acting through the CRE-like region (-0.96 kb) of the RANKL gene promoter, forskolin (FK) treatment transactivates the RANKL gene by antagonizing the function of Runx2, by reducing Runx2 mRNA expression and by opening the chromatin conformation far upstream (more than 40 kb) of the RANKL gene.
Collapse
Affiliation(s)
- Kiyoshi Mori
- Department of Bioinformatics, Division of Molecular Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
177
|
Selvamurugan N, Jefcoat SC, Kwok S, Kowalewski R, Tamasi JA, Partridge NC. Overexpression of Runx2 directed by the matrix metalloproteinase-13 promoter containing the AP-1 and Runx/RD/Cbfa sites alters bone remodeling in vivo. J Cell Biochem 2006; 99:545-57. [PMID: 16639721 DOI: 10.1002/jcb.20878] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The activator protein-1 (AP-1) and runt domain binding (Runx/RD/Cbfa) sites and their respective binding proteins, c-Fos/c-Jun and Runx2 (Cbfa1), regulate the rat matrix metalloproteinase-13 (MMP-13) promoter in both parathyroid hormone (PTH)-treated and differentiating osteoblastic cells in culture. To determine the importance of these regulatory sites in the expression of MMP-13 in vivo, transgenic mice containing either wild-type (-456 or -148) or AP-1 and Runx/RD/Cbfa sites mutated (-148A3R3) MMP-13 promoters fused with the E. coli lacZ reporter were generated. The wild-type transgenic lines expressed higher levels of bacterial beta-galactosidase in bone, teeth, and skin compared to the mutant and non-transgenic lines. Next, we investigated if overexpression of Runx2 directed by the MMP-13 promoter regulated expression of bone specific genes in vivo, and whether this causes morphological changes in these animals. Real time RT-PCR experiments identified increased mRNA expression of bone forming genes and decreased MMP-13 in the tibiae of transgenic mice (14 days and 6 weeks old). Histomorphometric analyses of the proximal tibiae showed increased bone mineralization surface, mineral apposition rate, and bone formation rate in the transgenic mice which appears to be due to decreased osteoclast number. Since MMP-13 is likely to play a role in recruiting osteoclasts to the bone surface, decreased expression of MMP-13 may cause reduced osteoclast-mediated bone resorption, resulting in greater bone formation in transgenic mice. In summary, we show here that the 148 bp upstream of the MMP-13 transcriptional start site is sufficient and necessary for gene expression in bone, teeth, and skin in vivo and the AP-1 and Runx/RD/Cbfa sites are likely to regulate this. Overexpression of Runx2 by these regulatory elements appears to alter the balance between the bone formation-bone resorption processes in vivo.
Collapse
Affiliation(s)
- Nagarajan Selvamurugan
- Department of Physiology and Biophysics, UMDNJ-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | | | | | |
Collapse
|
178
|
Mygind T, Stiehler M, Baatrup A, Li H, Zou X, Flyvbjerg A, Kassem M, Bünger C. Mesenchymal stem cell ingrowth and differentiation on coralline hydroxyapatite scaffolds. Biomaterials 2006; 28:1036-47. [PMID: 17081601 DOI: 10.1016/j.biomaterials.2006.10.003] [Citation(s) in RCA: 240] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Accepted: 10/09/2006] [Indexed: 12/13/2022]
Abstract
Culture of osteogenic cells on a porous scaffold could offer a new solution to bone grafting using autologous human mesenchymal stem cells (hMSC) from the patient. We compared coralline hydroxyapatite scaffolds with pore sizes of 200 and 500 microm for expansion and differentiation of hMSCs. We cultivated the hMSC statically or in spinner flasks for 1, 7, 14 and 21 days and found that the 200-microm pore scaffolds exhibited a faster rate of osteogenic differentiation than did the 500-microm pore scaffolds as shown by an alkaline phosphatase activity assay and real-time reverse transcriptase polymerase chain reaction for 10 osteogenic markers. The 500-microm scaffolds had increased proliferation rates and accommodated a higher number of cells (shown by DNA content, scanning electron microscopy and fluorescence microscopy). Thus the porosity of a 3D microporous biomaterial may be used to steer hMSC in a particular direction. We found that dynamic spinner flask cultivation of hMSC/scaffold constructs resulted in increased proliferation, differentiation and distribution of cells in scaffolds. Therefore, spinner flask cultivation is an easy-to-use inexpensive system for cultivating hMSCs on small to intermediate size 3D scaffolds.
Collapse
Affiliation(s)
- Tina Mygind
- Orthopedic Research Laboratory, Laboratory for Molecular Orthopedics, Clinical Institute, Aarhus University Hospital, Norrebrogade 44 bldg 1A, DK-8000 Aarhus C, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Smith A, Avaron F, Guay D, Padhi BK, Akimenko MA. Inhibition of BMP signaling during zebrafish fin regeneration disrupts fin growth and scleroblast differentiation and function. Dev Biol 2006; 299:438-54. [PMID: 16959242 DOI: 10.1016/j.ydbio.2006.08.016] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Revised: 08/04/2006] [Accepted: 08/07/2006] [Indexed: 12/17/2022]
Abstract
The zebrafish caudal fin provides a simple model to study molecular mechanisms of dermal bone regeneration. We previously showed that misexpression of Bone morphogenetic protein 2b (Bmp2b) induces ectopic bone formation within the regenerate. Here we show that in addition to bmp2b and bmp4 another family member, bmp6, is involved in fin regeneration. We further investigated the function of BMP signaling by ectopically expressing the BMP signaling inhibitor Chordin which caused: (1) inhibition of regenerate outgrowth due to a decrease of blastema cell proliferation and downregulation of msxb and msxC expression and (2) reduced bone matrix deposition resulting from a defect in the maturation and function of bone-secreting cells. We then identified targets of BMP signaling involved in regeneration of the bone of the fin rays. runx2a/b and their target col10a1 were downregulated following BMP signaling inhibition. Unexpectedly, the sox9a/b transcription factors responsible for chondrocyte differentiation were detected in the non-cartilaginous fin rays, sox9a and sox9b were not only differentially expressed but also differentially regulated since sox9a, but not sox9b, was downregulated in the absence of BMP signaling. Finally, this analysis revealed the surprising finding of the expression, in the fin regenerate, of several factors which are normally the signatures of chondrogenic elements during endochondral bone formation although fin rays form through dermal ossification, without a cartilage intermediate.
Collapse
Affiliation(s)
- A Smith
- Ottawa Health Research Institute, 725 Parkdale Avenue, Ottawa, ON, Canada K1Y4E9
| | | | | | | | | |
Collapse
|
180
|
Vaes BLT, Ducy P, Sijbers AM, Hendriks JMA, van Someren EP, de Jong NG, van den Heuvel ER, Olijve W, van Zoelen EJJ, Dechering KJ. Microarray analysis on Runx2-deficient mouse embryos reveals novel Runx2 functions and target genes during intramembranous and endochondral bone formation. Bone 2006; 39:724-38. [PMID: 16774856 DOI: 10.1016/j.bone.2006.04.024] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2006] [Revised: 04/10/2006] [Accepted: 04/20/2006] [Indexed: 10/24/2022]
Abstract
A major challenge in developmental biology is to correlate genome-wide gene expression modulations with developmental processes in vivo. In this study, we analyzed the role of Runx2 during intramembranous and endochondral bone development, by comparing gene expression profiles in 14.5 dpc wild-type and Runx2 (-/-) mice. A total of 1277, 606 and 492 transcripts were found to be significantly modulated by Runx2 in calvaria, forelimbs and hindlimbs, respectively. Bioinformatics analysis indicated that Runx2 not only controls the processes of osteoblast differentiation and chondrocyte maturation, but may also play a role in axon formation and hematopoietic cell commitment during bone development. A total of 41 genes are affected by the Runx2 deletion in both intramembranous and endochondral bone, indicating common pathways between these two developmental modes of bone formation. In addition, we identified genes that are specifically involved in endochondral ossification. In conclusion, our data show that a comparative genome-wide expression analysis of wild-type and mutant mouse models allows the examination of mutant phenotypes in complex tissues.
Collapse
Affiliation(s)
- Bart L T Vaes
- Department of Applied Biology FNWI, Radboud University Nijmegen, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Naitoh M, Kubota H, Ikeda M, Tanaka T, Shirane H, Suzuki S, Nagata K. Gene expression in human keloids is altered from dermal to chondrocytic and osteogenic lineage. Genes Cells 2006; 10:1081-91. [PMID: 16236136 DOI: 10.1111/j.1365-2443.2005.00902.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Keloids are a dermal fibrotic disease whose etiology remains totally unknown and for which there is no successful treatment. Here, we employed cDNA microarray analysis to examine gene expression in keloid lesions and control skin. We found that 32 genes among the 9000 tested were strongly up-regulated in keloid lesions, of which 21 were confirmed by Northern blotting. These included at least seven chondrocyte/osteoblast marker genes, and RT-PCR analysis revealed that transcription factors specific for these genes, SOX9 and CBFA1, were induced. Immunostaining and in situ hybridization further supported that these markers are expressed in keloid lesions. Intriguingly, scleraxis, a transcription factor known as a marker of tendons and ligaments, was also induced in keloid fibroblasts. We propose that reprogramming of gene expression or disordered differentiation from a dermal pattern to that of a chondrocytic/osteogenic lineage, probably closer to that of tendon/ligament lineage, may be involved in the etiology of keloids.
Collapse
Affiliation(s)
- Motoko Naitoh
- Department of Molecular and Cellular Biology, Institute for Frontier Medical Science, Kyoto University, Kyoto 606-8397, Japan
| | | | | | | | | | | | | |
Collapse
|
182
|
Zhang X, Yang M, Lin L, Chen P, Ma KT, Zhou CY, Ao YF. Runx2 overexpression enhances osteoblastic differentiation and mineralization in adipose--derived stem cells in vitro and in vivo. Calcif Tissue Int 2006; 79:169-78. [PMID: 16969589 DOI: 10.1007/s00223-006-0083-6] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Accepted: 05/22/2006] [Indexed: 10/24/2022]
Abstract
Like bone marrow stromal cells, adipose tissue-derived stem cells (ADSCs) possess multilineage potential, a capacity for self-renewal and long-term viability. To confirm whether ADSCs represent a promising source of cells for gene-enhanced bone tissue-engineering, the osteogenic potential of ADSCs under the control of certain osteoinductive genes has been evaluated. Runx2, a transcription factor at the downstream end of bone morphogenetic protein (BMP) signaling pathways, is essential for osteoblast differentiation and bone formation. In this study we used adenovirus vector to deliver Runx2 to ADSCs and then examined the enhancement of osteogenic activity. Overexpression of Runx2 inhibited adipogenesis, as demonstrated by suppression of LPL and PPARgamma expression at the mRNA level and reduced lipid droplet formation. Moreover, ADSCs transduced with Ad-Runx2 underwent rapid and marked osteoblast differentiation as determined by osteoblastic gene expression, alkaline phosphatase activity and mineral deposition. Additionally, histological examination revealed that implantation of Runx2 modified ADSCs could induce mineral deposition and bone-like tissue formation in vivo. These results confirmed, firstly, the ability of Runx2 to promote osteogenesis and cell differentiation and, secondly, the competence of ADSCs as target cells for bone tissue engineering. Our work demonstrates a potential new approach for bone repair using Runx2-modified ADSCs for bone tissue engineering.
Collapse
Affiliation(s)
- X Zhang
- Department of Sports Medicine, Peking University Third Hospital, Beijing 100083, China
| | | | | | | | | | | | | |
Collapse
|
183
|
Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J Cell Biochem 2006; 98:538-54. [PMID: 16317727 DOI: 10.1002/jcb.20719] [Citation(s) in RCA: 156] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSC) are multipotent, self-renewing, mesodermal-origin stem cells that are sequestered in the endosteal compartment. MSC are maintained in a relative state of quiescence in vivo but in response to a variety of physiological and pathological stimuli, proliferate and differentiate into osteoblasts, chondrocytes, adipocytes, or hematopoiesis-supporting stromal cells. Little is understood regarding the cellular or molecular events underlying MSC fate decisions. We report that human MSC (hMSC) cultured in defined, serum-free conditions respond to a narrow spectrum of growth factors with osteogenic commitment, differentiation, and hydroxyapatite deposition. Of the osteogenic factors we examined, only treatment with bone morphogenetic protein (BMP) results in osteoinduction under defined serum-free conditions. Among BMP-2, 4, 6, and 7, BMP-6 is the most consistent and potent regulator of osteoblast differentiation and, of these BMPs, only BMP-6 gene expression is detected prior to hMSC osteoblast differentiation. Addition of exogenous BMP-6 to hMSC induces the expression or upregulation of a repertoire of osteoblast-related genes including type I collagen, osteocalcin, bone sialoprotein, and their regulatory transcription factors Cbfa1/Runx2, and Osterix. This translates into increased production of osteogenic extracellular matrix (ECM) with subsequent hydroxyapatite deposition.
Collapse
Affiliation(s)
- Michael S Friedman
- Graduate Program in Cellular and Molecular Biology, University of Michigan, USA
| | | | | |
Collapse
|
184
|
Jackson RA, Nurcombe V, Cool SM. Coordinated fibroblast growth factor and heparan sulfate regulation of osteogenesis. Gene 2006; 379:79-91. [PMID: 16797878 DOI: 10.1016/j.gene.2006.04.028] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 04/17/2006] [Accepted: 04/20/2006] [Indexed: 01/13/2023]
Abstract
Growth and lineage-specific differentiation constitute crucial phases in the development of stem cells. Control over these processes is exerted by particular elements of the extracellular matrix, which ultimately trigger a cascade of signals that regulate uncommitted cells, by modulating their survival and cell cycle progression, to shape developmental processes. Uncontrolled, constitutive activation of fibroblast growth factor receptors (FGFR) results in bone abnormalities, underlining the stringent control over fibroblast growth factor (FGF) activity that must be maintained for normal osteogenesis to proceed. Mounting evidence suggests that FGF signalling, together with a large number of other growth and adhesive factors, is controlled by the extracellular glycosaminoglycan sugar, heparan sulfate (HS). In this review, we focus on FGF activity during osteogenesis, their receptors, and the use of HS as a therapeutic adjuvant for bone repair.
Collapse
Affiliation(s)
- Rebecca A Jackson
- Laboratory of Stem Cells and Tissue Repair, Institute of Molecular and Cell Biology, Proteos, Singapore.
| | | | | |
Collapse
|
185
|
Shen R, Wang X, Drissi H, Liu F, O'Keefe RJ, Chen D. Cyclin D1-cdk4 induce runx2 ubiquitination and degradation. J Biol Chem 2006; 281:16347-53. [PMID: 16613857 PMCID: PMC2649830 DOI: 10.1074/jbc.m603439200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Runx2 is a Runt domain transcription factor involved in the activation of genes encoding osteoblast and chondrocyte-specific proteins. Runx2 activity is regulated by transcriptional and post-transcriptional mechanisms. The functional significance of the post-translational modification of Runx2 has not been fully defined. We show that cyclin D1-Cdk4 induce Runx2 degradation in an ubiquitination-proteasome-dependent manner. Mutagenesis of Runx2 serine-472, a consensus Cdk site, to alanine increases the half-life of Runx2 and causes loss of sensitivity to cyclin D1-induced Runx2 degradation. The targeted Runx2 degradation by cyclin D1 identifies a novel mechanism through which Runx2 activity is regulated coordinately with the cell cycle machinery in bone cells.
Collapse
Affiliation(s)
- Run Shen
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
186
|
Kanatani N, Fujita T, Fukuyama R, Liu W, Yoshida CA, Moriishi T, Yamana K, Miyazaki T, Toyosawa S, Komori T. Cbf beta regulates Runx2 function isoform-dependently in postnatal bone development. Dev Biol 2006; 296:48-61. [PMID: 16797526 DOI: 10.1016/j.ydbio.2006.03.039] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 03/21/2006] [Accepted: 03/28/2006] [Indexed: 01/10/2023]
Abstract
Runx2 and Cbfbeta are essential for skeletal development during the embryonic stage. Runx2 has two isoforms with different N-termini. We examined the functions of the Runx2 isoforms and Cbfbeta in postnatal bone development. On luciferase and electrophoretic mobility shift assays, Runx2-I was less active than Runx2-II in the absence of Cbfb, but the two Runx2 isoforms had similar activity levels in the presence of Cbfb. We generated Runx2-I transgenic mice under the control of Col1a1 promoter and Runx2-I/Cbfb and Runx2-II/Cbfb double transgenic mice. Runx2-I transgenic mice showed less severe osteopenia and fragility than Runx2-II transgenic mice due to milder inhibition of both osteoblast maturation and transition to osteocytes, even though the former mice showed higher transgene expression. However, Runx2-I/Cbfb and Runx2-II/Cbfb double transgenic mice had enhanced inhibition of osteoblast maturation, resulting in similar severity of osteopenia and fragility, although the latter mice had less osteocytes. These findings indicate that (1) Runx2-II more strongly inhibits osteoblast maturation and transition to osteocytes than Runx2-I; (2) Cbfbeta regulates Runx2 function isoform-dependently; and (3) Runx2-I activity is highly dependent on Cbfbeta. These findings demonstrate that Runx2 isoforms exert their functions through at least partly different mechanisms and Cbfbeta regulates bone development by regulating Runx2 function isoform-dependently.
Collapse
Affiliation(s)
- Naoko Kanatani
- Department of Developmental and Reconstructive Medicine, Division of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Takahashi T, Kato S, Suzuki N, Kawabata N, Takagi M. Autoregulatory mechanism of Runx2 through the expression of transcription factors and bone matrix proteins in multipotential mesenchymal cell line, ROB-C26. J Oral Sci 2006; 47:199-207. [PMID: 16415564 DOI: 10.2334/josnusd.47.199] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Runx2 is essential for osteoblast differentiation and gene expression of bone matrix proteins, however, little is known about the mechanism regulating its activity. In this study, the role of Runx2 on gene expression of transcription factors, AJ18, Msx2, and Dlx5, was examined in vitro. It is known that AJ18 and Msx2 act as repressors to inhibit activity of Runx2, whereas Dlx5 promotes its activity. An expression vector inserted Runx2 cDNA was transiently overexpressed in a rat multipotential mesenchymal cell line, ROB-C26 (C26). Real time reverse transcription-PCR analysis showed that, in exogenous Runx2-overexpressing C26 cells (C26-Rx), AJ18 expression increased 1.8-fold, Msx2 expression increased 3.0-fold, and Dlx5 expression increased 2.7-fold compared to the cells transfected with vector alone (C26-Co). Luciferase assay also showed that, in C26-Rx, AJ18 promoter activity increased 2.1-fold compared to C26-Co. Furthermore, gene expression of alkaline phosphatase (ALP) and bone matrix proteins including type I collagen (Col1), osteocalcin (OC), osteopontin (OPN), and matrix Gla protein (MGP) was examined. In C26-Rx, MGP expression increased 1.8-fold, and OPN expression increased 1.4-fold compared to C26-Co. However, no significant difference in Col1, ALP, and OC expressions was detected between C26-Rx and C26-Co. These results suggest that the existence of autoregulatory feed back loops, which inhibit Runx2 activity through the interaction of AJ18, Dlx5, and Msx2 cooperating with that of MGP and OPN, interferes with the differentiation of C26 cells toward mature osteoblasts.
Collapse
Affiliation(s)
- Tomihisa Takahashi
- Department of Anatomy, Nihon University School of Dentistry, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
188
|
Mehrotra M, Saegusa M, Voznesensky O, Pilbeam C. Role of Cbfa1/Runx2 in the fluid shear stress induction of COX-2 in osteoblasts. Biochem Biophys Res Commun 2006; 341:1225-30. [PMID: 16476583 DOI: 10.1016/j.bbrc.2006.01.084] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/20/2006] [Indexed: 10/25/2022]
Abstract
Induction of cyclooxygenase-2 (COX-2) is thought to be important for the anabolic effects of mechanical loading. The transcription factor Cbfa1/Runx2 is essential for osteoblastic differentiation. We examined the role of Cbfa1 in the fluid shear stress (FSS) induction of COX-2 in MC3T3-E1 cells stably transfected with a COX-2 promoter-luciferase reporter. Cells were subjected to FSS for 30 min and returned to static culture (post-FSS). COX-2 mRNA and promoter activity peaked 0.5-1h and 2-3h, respectively, post-FSS. Mutation of the Cbfa1 consensus sequence at -267/-261 bp decreased the FSS fold-induction of luciferase activity by 50%. On electrophoretic mobility shift assay (EMSA), proteins binding to an oligonucleotide spanning the Cbfa1 site were supershifted by specific antibody to Cbfa1. FSS did not increase Cbfa1 binding on EMSA or Cbfa1 mRNA or protein levels. These data suggest that transcriptional activity of Cbfa1, independent of its level of expression, is necessary for maximal FSS induction of COX-2 in osteoblasts.
Collapse
Affiliation(s)
- Meenal Mehrotra
- Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | |
Collapse
|
189
|
Jackson RA, Kumarasuriyar A, Nurcombe V, Cool SM. Long-term loading inhibits ERK1/2 phosphorylation and increases FGFR3 expression in MC3T3-E1 osteoblast cells. J Cell Physiol 2006; 209:894-904. [PMID: 16972271 DOI: 10.1002/jcp.20779] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bone tissue homeostasis relies upon the ability of cells to detect and interpret extracellular signals that direct changes in tissue architecture. This study utilized a four-point bending model to create both fluid shear and strain forces (loading) during the time-dependent progression of MC3T3-E1 preosteoblasts along the osteogenic lineage. Loading was shown to increase cell number, alkaline phosphatase (ALP) activity, collagen synthesis, and the mRNA expression levels of Runx2, osteocalcin (OC), osteopontin, and cyclo-oxygenase-2. However, mineralization in these cultures was inhibited, despite an increase in calcium accumulation, suggesting that loading may inhibit mineralization in order to increase matrix deposition. Loading also increased fibroblast growth factor receptor-3 (FGFR3) expression coincident with an inhibition of FGFR1, FGFR4, FGF1, and extracellular signal-related kinase (ERK)1/2 phosphorylation. To examine whether these loading-induced changes in cell phenotype and FGFR expression could be attributed to the inhibition of ERK1/2 phosphorylation, cells were grown for 25 days in the presence of the MEK1/2 inhibitor, U0126. Significant increases in the expression of FGFR3, ALP, and OC were observed, as well as the inhibition of FGFR1, FGFR4, and FGF1. However, U0126 also increased matrix mineralization, demonstrating that inhibition of ERK1/2 phosphorylation cannot fully account for the changes observed in response to loading. In conclusion, this study demonstrates that preosteoblasts are mechanoresponsive, and that long-term loading, whilst increasing proliferation and differentiation of preosteoblasts, inhibits matrix mineralization. In addition, the increase in FGFR3 expression suggests that it may have a role in osteoblast differentiation.
Collapse
Affiliation(s)
- Rebecca A Jackson
- School of Biomedical Sciences, University of Queensland, Queensland, Australia
| | | | | | | |
Collapse
|
190
|
Maehata Y, Takamizawa S, Ozawa S, Kato Y, Sato S, Kubota E, Hata RI. Both direct and collagen-mediated signals are required for active vitamin D3-elicited differentiation of human osteoblastic cells: roles of osterix, an osteoblast-related transcription factor. Matrix Biol 2006; 25:47-58. [PMID: 16266799 DOI: 10.1016/j.matbio.2005.09.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 08/11/2005] [Accepted: 09/07/2005] [Indexed: 12/19/2022]
Abstract
In order to investigate the mechanisms by which 1alpha,25(OH)2 vitamin D3 (VD3) stimulates the differentiation of human osteoblasts, we cultured MG-63, which is a human osteoblastic cell line, in the presence or absence of VD3 and/or L-ascorbic acid 2-phosphate (Asc 2-P), a long-acting vitamin C derivative. The cell growth rate was decreased by the presence of VD3 in the culture medium. Type I collagen synthesis and alkaline phosphatase (ALP) activity, which are markers of early stage osteoblast differentiation, were stimulated by the presence of VD3 as well as by that of Asc 2-P. The co-presence of Asc 2-P and VD3 had a synergistic effect on the collagen synthesis and ALP activity of the cells. Inhibition of collagen synthesis by the addition of inhibitors of collagen synthesis to the medium attenuated the stimulative effect of VD3 and Asc 2-P on the ALP activity. Transfection of the cells with siRNA-expressing vectors for COL1A1 decreased the expression level of ALP mRNA in addition to that of COL1A1. On the other hand, ALP activity was significantly increased, and the growth rate was decreased, when the cells were cultured on type I collagen-coated dishes. These effects were not seen when the cells were cultured on dishes coated with heat-denatured collagen. VD3 also increased the mRNA levels for Runx2 and osterix, which are transcription factors critical for osteoblast differentiation, as well as those of differentiation markers such as bone/liver/kidney type ALP, COL1A1, (the gene for the alpha1 chain of type I collagen), and osteocalcin, in the cells. Normal human osteoblasts and human bone marrow-derived mesenchymal stem cells (hBMSC) showed quite similar responses to VD3. These results indicate that VD3-stimulated gene expression of type I collagen and that mature type I collagen produced in the presence of Asc 2-P mediates at least a part of the stimulative effects of Asc 2-P and VD3 on the differentiation of these human osteoblastic cells. Levels of mRNAs for ALP and COL1A1 were increased, but the level of Runx2 was decreased, by the expression of osterix in MG-63 cells. These results also suggest that VD3 controls the growth and differentiation of human osteoblastic cells by regulating the gene expression of osteoblast-related transcription factors as well as that of type I collagen, and that the co-presence of both signals is essential for VD3 to express full activity toward the differentiation of human osteoblasts.
Collapse
Affiliation(s)
- Yojiro Maehata
- Department of Biochemistry and Molecular Biology, Kanagawa Dental College, Yokosuka, 238-8580, Japan
| | | | | | | | | | | | | |
Collapse
|
191
|
Jackson RA, Murali S, van Wijnen AJ, Stein GS, Nurcombe V, Cool SM. Heparan sulfate regulates the anabolic activity of MC3T3-E1 preosteoblast cells by induction of Runx2. J Cell Physiol 2006; 210:38-50. [PMID: 17051597 DOI: 10.1002/jcp.20813] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The transcription factor Runx2 can be controlled by a number of upstream regulators involved in intracellular signalling, including the activation ERK1/2 signaling by fibroblast growth factor-2 (FGF-2). FGFs interact with their cell surface receptors (FGFRs) through an obligate cross-binding interaction with heparan sulfate proteoglycan (HSPG) co-receptors; exogenous HS sugar chains have been shown to potently modulate changes in cell phenotype depending on the stage of tissue differentiation when the HS is harvested, suggesting that HS chain structure and function varies depending on the stage of cell maturity. This study examined the potential of bone-derived heparan sulfate (HS), harvested from differentiating osteoblasts, for the enhancement of preosteoblast growth and differentiation. HS was harvested from conditioned media, cell surface and matrix compartments of postconfluent (differentiating) MC3T3-E1 osteoblasts and dosed back onto preconfluent MC3T3-E1 cells. We show that HS can increase the expression Runx2, ALP, and OPN in preosteoblast cells, suggesting the potential for exogenous HS to shift cells from proliferative to differentiative phenotypes. In line with their structural differences, only HS released into the media was found to co-stimulate the mitogenic effect of FGF-2, whilst exogenous application of all the HSs together with FGF-2 served to increase the expression of OPN. Only the application of cell surface-derived HS triggered a synergistic increase in FGFR1 expression together with FGF-2, although all three HS preparations could trigger transient increases in PI3K, ERK1/2, and stat3 phosphorylation levels. These findings demonstrate that the compartmentally distinct HS species expressed by differentiating MC3T3-E1 cells act in complex ways to coordinate the extracellular conditions that lead to osteoblast differentiation, with the cell surface species coordinating the FGF response.
Collapse
Affiliation(s)
- Rebecca A Jackson
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | | | | | | | | | | |
Collapse
|
192
|
Shen R, Chen M, Wang YJ, Kaneki H, Xing L, O'keefe RJ, Chen D. Smad6 interacts with Runx2 and mediates Smad ubiquitin regulatory factor 1-induced Runx2 degradation. J Biol Chem 2005; 281:3569-76. [PMID: 16299379 PMCID: PMC2647593 DOI: 10.1074/jbc.m506761200] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Runx2 is a bone-specific transcription factor that plays a critical role in bone development, postnatal bone formation, and chondrocyte maturation. The protein levels of Runx2 are regulated by the ubiquitin-proteasome pathway. In previous studies we discovered that E3 ubiquitin ligase Smad ubiquitin regulatory factor 1 (Smurf1) induces Runx2 degradation in a ubiquitin-proteasome-dependent manner, and Smurf1 plays an important role in osteoblast function and bone formation. In the present studies we investigated the molecular mechanism of Smurf1-induced Runx2 degradation. Smurf1 interacts with the PY motif of substrate proteins, and a PY motif has been identified in the C terminus of the Runx2 protein. To determine whether Smurf1 induces Runx2 degradation through the interaction with the PY motif of Runx2, we created a mutant Runx2 with a PY motif deletion and found that Smurf1 retained some of its ability to induce the degradation of the mutant Runx2, suggesting that Smurf1 could induce Runx2 degradation through an indirect mechanism. Smurf1 has been shown to interact with Smads 1, 5, 6, and 7, and Smads 1 and 5 also interact with Runx2. In the present studies we found that Smads 1 and 5 had no effect on Smurf1-induced Runx2 degradation. Although Smads 6 and 7 bind Smurf1, it is not known if Smads 6 or 7 interacts with Runx2 and mediate Runx2 degradation. We performed immunoprecipitation assays and found that Smad6 but not Smad7 interacts with Runx2. Smad6 enhances Smurf1-induced Runx2 degradation in an ubiquitin-proteasome-dependent manner. These results demonstrate that in addition to its interaction with the PY motif of Runx2, Smurf1 induces Runx2 degradation in a Smad6-dependent manner. Smurf1-induced Runx2 degradation serves as a negative regulatory mechanism for the BMP-Smad-Runx2 signaling pathway.
Collapse
Affiliation(s)
- Run Shen
- Department of Orthopaedics and Department of Pathology, Center for Musculoskeletal Research, University of Rochester, School of Medicine, Rochester, New York 14642, USA
| | | | | | | | | | | | | |
Collapse
|
193
|
Akiyama H, Kim JE, Nakashima K, Balmes G, Iwai N, Deng JM, Zhang Z, Martin JF, Behringer RR, Nakamura T, de Crombrugghe B. Osteo-chondroprogenitor cells are derived from Sox9 expressing precursors. Proc Natl Acad Sci U S A 2005; 102:14665-70. [PMID: 16203988 PMCID: PMC1239942 DOI: 10.1073/pnas.0504750102] [Citation(s) in RCA: 423] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The transcription factor Sox9 is expressed in all chondroprogenitors and has an essential role in chondrogenesis. Sox9 is also expressed in other tissues, including central nervous system, neural crest, intestine, pancreas, testis, and endocardial cushions, and plays a crucial role in cell proliferation and differentiation in several of these tissues. To determine the cell fate of Sox9-expressing cells during mouse embryogenesis, we generated mice in which a Cre recombinase gene preceded by an internal ribosome entry site was inserted into the 3' untranslated region of the Sox9 gene (Sox9-Cre knock-in). In the developing skeleton, Sox9 was expressed before Runx2, an early osteoblast marker gene. Cell fate mapping by using Sox9-Cre;ROSA26 reporter (R26R) mice revealed that Sox9-expressing limb bud mesenchymal cells gave rise to both chondrocytes and osteoblasts. Furthermore, a mutant in which the Osterix gene was inactivated in Sox9-expressing cells exhibited a lack of endochondral and intramembranous ossification and a lack of mature osteoblasts comparable with Osterix-null mutants. In addition, Sox9-expressing limb bud mesenchymal cells also contributed to tendon and synovium formation. By using Sox9-Cre;R26R mice, we also were able to systematically follow Sox9-expressing cells from embryonic day 8.0 to 17.0. Our results showed that Sox9-expressing cells contributed to the formation of all cell types of the spinal cord, epithelium of the intestine, pancreas, and mesenchyme of the testis. Thus, our results strongly suggest that all osteo-chondroprogenitor cells, as well as progenitors in a variety of tissues, are derived from Sox9-expressing precursors during mouse embryogenesis.
Collapse
Affiliation(s)
- Haruhiko Akiyama
- Department of Molecular Genetics, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Chikazu D, Li X, Kawaguchi H, Sakuma Y, Voznesensky OS, Adams DJ, Xu M, Hoshi K, Katavic V, Herschman HR, Raisz LG, Pilbeam CC. Bone morphogenetic protein 2 induces cyclo-oxygenase 2 in osteoblasts via a Cbfa1 binding site: role in effects of bone morphogenetic protein 2 in vitro and in vivo. 2002. J Bone Miner Res 2005; 20:1888-98. [PMID: 16355502 DOI: 10.1359/jbmr.2005.20.10.1887] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
195
|
Roca H, Phimphilai M, Gopalakrishnan R, Xiao G, Franceschi RT. Cooperative interactions between RUNX2 and homeodomain protein-binding sites are critical for the osteoblast-specific expression of the bone sialoprotein gene. J Biol Chem 2005; 280:30845-55. [PMID: 16000302 DOI: 10.1074/jbc.m503942200] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The bone sialoprotein (Bsp) gene provides an excellent model for studying mechanisms controlling osteoblast-specific gene expression. Although the RUNX2 transcription factor directly regulates many osteoblast-related genes, its function in Bsp expression remains uncertain. By using chromatin immunoprecipitation (ChIP) analysis in MC3T3-E1 (clone MC-4) preosteoblast cells, RUNX2 was shown to bind a chromatin fragment containing the proximal Bsp promoter. Two putative RUNX2-binding sites (R1 and R2) were identified within this region of the mouse, rat, and human genes and were shown to bind RUNX2 in vitro and in vivo (by ChIP assay). Site-specific mutagenesis established that both sites act as osteoblast-specific transcriptional enhancers and together account for nearly two-thirds of the total promoter activity. In addition, functional cooperativity was observed between the R2 site and an adjacent homeodomain protein-binding site previously characterized by this laboratory (the C site). All three sites (R1, R2, and C) are necessary for maximal promoter activity in osteoblasts. DLX5 in MC-4 cell nuclear extracts binds to the C site in vitro. Furthermore, ChIP assays revealed that DLX5 is selectively associated with chromatin in the vicinity of the C site only when Bsp is transcriptionally active. Finally, co-immunoprecipitation assays detected a physical complex containing DLX5 and RUNX2. Taken together, our data show that RUNX2 is a direct regulator of Bsp in osteoblasts and that it functions in cooperation with DLX5 or a related factor to activate osteoblast-specific gene expression.
Collapse
Affiliation(s)
- Hernan Roca
- Department of Periodontics, Prevention, and Geriatrics and Center for Craniofacial Regeneration, School of Dentistry, University of Michigan, Ann Arbor 48109-1078, USA
| | | | | | | | | |
Collapse
|
196
|
Chen S, Rani S, Wu Y, Unterbrink A, Gu TT, Gluhak-Heinrich J, Chuang HH, Macdougall M. Differential regulation of dentin sialophosphoprotein expression by Runx2 during odontoblast cytodifferentiation. J Biol Chem 2005; 280:29717-27. [PMID: 15980071 DOI: 10.1074/jbc.m502929200] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dentin sialophosphoprotein (DSPP) consists of dentin sialoprotein (DSP) and dentin phosphoprotein (DPP). The spatial-temporal expression of DSPP is largely restricted during differentiational stages of dental cells. DSPP plays a vital role in tooth development. It is known that an osteoblast-specific transcription factor, Runx2, is essential for osteoblast differentiation. However, effects of Runx2 on DSPP transcription remain unknown. Here, we studied different roles of Runx2 in controlling DSPP expression in mouse preodontoblast (MD10-F2) and odontoblast (MO6-G3) cells. Two Runx2 isoforms were expressed in preodontoblast and odontoblast cells, and in situ hybridization assay showed that DSPP expression increased, whereas Runx2 was down-regulated during odontoblast differentiation and maturation. Three potential Runx2 sites are present in promoters of mouse and rat DSPP genes. Runx2 binds to these sites as demonstrated by electrophoretic mobility shift assay and supershift experiments. Mutations of Runx2 sites in mouse DSPP promoter resulted in a decline of promoter activity in MD10-F2 cells compared with an increase of its activity in MO6-G3 cells. Multiple Runx2 sites were more active than a single site in regulating the DSPP promoter. Furthermore, forced overexpression of Runx2 isoforms induced increases of endogenous DSPP protein levels in MD10-F2 cells but reduced its expression in MO6-G3 cells consistent with the DSPP promoter analysis. Thus, our results suggest that differential positive and negative regulation of DSPP by Runx2 is dependent on use of cytodifferentiation of dental ectomesenchymal-derived cells that may contribute to the spatial-temporal expression of DSPP during tooth development.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Pediatric Dentistry, The University of Texas Health Science Center, San Antonio, 78229-3900, USA.
| | | | | | | | | | | | | | | |
Collapse
|
197
|
Lietman SA, Ding C, Cooke DW, Levine MA. Reduction in Gsalpha induces osteogenic differentiation in human mesenchymal stem cells. Clin Orthop Relat Res 2005:231-8. [PMID: 15864058 DOI: 10.1097/01.blo.0000153279.90512.38] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We hypothesized that a decrease in Gsalpha expression occurs with osteogenic differentiation and that when Gsalpha expression was decreased by antisense oligonucleotides or direct inhibition of protein kinase A there was a concomitant increase in Runx2/Cbfa1. We also investigated the mechanism involved in the change in Runx2/Cbfa1 levels and whether the expression of other genes known to be involved in bone formation was altered. There was a decrease in Gsalpha expression with osteogenic differentiation and antisense oligonucleotides, and protein kinase A inhibition led to increased expression and DNA binding of the osteoblast-specific Runx2/Cbfa1. Additionally, with decreased Gsalpha expression or protein kinase A inhibition, Runx2/Cbfa1 protein was serine phosphorylated and ubiquitinated less. Microarray analysis, after the addition of antisense Gsalpha, showed a more than 10-fold increase in collagen Type I Alpha 2 mRNA (a target of Runx2/Cbfa1). These data show that reduced expression of Gsalpha can induce an osteoblast-like phenotype. The results also indicate a potential pathophysiologic role in patients with heterozygous inactivating mutations in GNAS1, the gene for the alpha chain (Gsalpha) of the heterotrimeric G protein, present in three disorders with ectopic intramembranous bone: Albright's hereditary osteodystrophy, progressive osseous heteroplasia, and osteoma cutis.
Collapse
Affiliation(s)
- Steven A Lietman
- Department of Orthopaedic Surgery, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| | | | | | | |
Collapse
|
198
|
Varghese S, Rydziel S, Canalis E. Bone morphogenetic protein-2 suppresses collagenase-3 promoter activity in osteoblasts through a runt domain factor 2 binding site. J Cell Physiol 2005; 202:391-9. [PMID: 15389594 DOI: 10.1002/jcp.20130] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transforming growth factor-beta (TGFbeta) superfamily of growth factors, which include bone morphogenetic proteins (BMPs), have multiple effects in osteoblasts. In this study, we examined the regulation of collagenase-3 promoter activity by BMP-2 in osteoblast-enriched (Ob) cells from fetal rat calvariae. BMP-2 suppressed the activity of a -2 kb collagenase-3 promoter/luciferase recombinant in a time- and dose-dependent manner. The BMP-2 effect on the collagenase-3 promoter was further tested in several collagenase-3 promoter deletion constructs and it was narrowed down to a -148 to -94 nucleotide segment of the promoter containing a runt domain factor 2 (Runx2) site at nucleotide -132 to -126. The effect of BMP-2 was obliterated in a collagenase-3 promoter/luciferase construct containing a mutated Runx2 (mRunx2) sequence indicating that the Runx2 site mediates the BMP-2 response. Electrophoretic mobility shift assays, using nuclear extracts from control and BMP-2-treated Ob cells, indicated that the Runx2 protein is a component of the specific DNA-protein complex formed on the Runx2 site and that the BMP-2 effect may be associated with minor protein modifications rather than major changes in the composition of specific proteins interacting with the Runx2 site. We confirmed that other members of the TGFbeta family can down-regulate the collagenase-3 promoter by showing that TGFbeta1 also suppresses the promoter activity in a time- and dose-dependent manner. In conclusion, this study demonstrates that BMP-2 and TGFbeta1 suppress collagenase-3 promoter activity in osteoblasts and establishes a link between BMP-2 action and collagenase-3 expression via Runx2, a major regulator of osteoblast formation and function.
Collapse
Affiliation(s)
- Samuel Varghese
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut 06105, USA.
| | | | | |
Collapse
|
199
|
Chen S, Santos L, Wu Y, Vuong R, Gay I, Schulze J, Chuang HH, MacDougall M. Altered gene expression in human cleidocranial dysplasia dental pulp cells. Arch Oral Biol 2005; 50:227-36. [PMID: 15721154 DOI: 10.1016/j.archoralbio.2004.10.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Accepted: 10/07/2004] [Indexed: 11/13/2022]
Abstract
Cleidocranial dysplasia (CCD) is an autosomal dominant disorder characterised by defects of bone and tooth development. The dental manifestations in CCD patients include supernumerary teeth, delayed tooth eruption, tooth hypoplasia and absence of cellular cementum formation. This disorder is associated with mutations in the osteoblast-specific transcription factor Runx2. To identify morphological and molecular alterations associated with CCD dental tissues, human primary dental pulp cell cultures were established from age- and sex-matched CCD and normal patients. Dental pulp cells were compared for general morphology, proliferation rates, and gene expression profiles using cDNA microarray technology. CCD pulp cells were about four-fold larger than normal cells, however the normal pulp proliferation rates were two- and three-fold greater at time points tested than the CCD cells. Of the 226 genes analysed by blot microarray, 18.6% displayed significant differences at least two-fold in expression levels. This includes 25 genes (11.1%) that were up-regulated, while 17 (7.5%) that were down-regulated in the CCD cells as compared to the normal cells. Expression of selected genes was further verified by quantitative real-time polymerase chain reaction (qRT-PCR). Comparison between the CDD and normal cells revealed that gene expression of cytokines and growth factors, such as leukemia inhibitory factor (LIF), interleukin-6 (IL-6) and transforming growth factor beta receptor II (TGF-betaRII) and vascular endothelial growth factor B (VEGFB) were higher while bone morphogenetic protein 2 (BMP2) was lower in the CCD cells. Furthermore, potential Runx2 binding sites were found in all putative target gene promoters. This study suggests that in addition to bone and tooth cell differentiation, Runx2 may be involved in controlling cell growth during tooth development.
Collapse
Affiliation(s)
- Shuo Chen
- Department of Pediatric Dentistry, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | | | | | |
Collapse
|
200
|
Byers BA, García AJ. Exogenous Runx2 Expression Enhances in Vitro Osteoblastic Differentiation and Mineralization in Primary Bone Marrow Stromal Cells. ACTA ACUST UNITED AC 2004; 10:1623-32. [PMID: 15684671 DOI: 10.1089/ten.2004.10.1623] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Bone marrow stromal cells represent a promising cell source for cell-based therapeutic and bone tissue-engineering applications, but are restricted by a low frequency in healthy marrow, an age-related decrease in osteogenic capacity, and a propensity for dedifferentiation during in vitro expansion. To address these limitations, retroviral gene delivery was used to examine the effects of sustained and elevated expression of the Runx2 osteoblastic transcription factor on osteoblastic gene and protein expression and mineralization in primary rat bone marrow stromal cells. Runx2 overexpression upregulated several osteoblast-specific genes, including collagen type I and osteocalcin, and enhanced alkaline phosphatase activity and biological mineral deposition. Forced Runx2 expression in combination with dexamethasone increased matrix mineralization compared with exogenous Runx2 expression or dexamethasone treatment alone, whereas dexamethasone-free control cultures displayed minimal mineralization. These additive effects suggest complementary interactions between Runx2 and dexamethasone-responsive regulatory factors. Finally, Runx2 overexpression in stromal cell cultures undergoing considerable in vitro expansion resulted in higher matrix mineralization capacity compared with controls, which completely lost the ability to produce mineralized matrix even in the presence of dexamethasone. These findings provide a novel strategy for cell-based therapeutic applications requiring significant numbers of osteogenic cells to synthesize mineralized constructs for the treatment of large bone defects.
Collapse
Affiliation(s)
- Benjamin A Byers
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | | |
Collapse
|