101
|
Xu J, Yu B, Hong C, Wang CY. KDM6B epigenetically regulates odontogenic differentiation of dental mesenchymal stem cells. Int J Oral Sci 2013; 5:200-5. [PMID: 24158144 PMCID: PMC3967319 DOI: 10.1038/ijos.2013.77] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 06/04/2013] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been identified and isolated from dental tissues, including stem cells from apical papilla, which demonstrated the ability to differentiate into dentin-forming odontoblasts. The histone demethylase KDM6B (also known as JMJD3) was shown to play a key role in promoting osteogenic commitment by removing epigenetic marks H3K27me3 from the promoters of osteogenic genes. Whether KDM6B is involved in odontogenic differentiation of dental MSCs, however, is not known. Here, we explored the role of KDM6B in dental MSC fate determination into the odontogenic lineage. Using shRNA-expressing lentivirus, we performed KDM6B knockdown in dental MSCs and observed that KDM6B depletion leads to a significant reduction in alkaline phosphate (ALP) activity and in formation of mineralized nodules assessed by Alizarin Red staining. Additionally, mRNA expression of odontogenic marker gene SP7 (osterix, OSX), as well as extracellular matrix genes BGLAP (osteoclacin, OCN) and SPP1 (osteopontin, OPN), was suppressed by KDM6B depletion. When KDM6B was overexpressed in KDM6B-knockdown MSCs, odontogenic differentiation was restored, further confirming the facilitating role of KDM6B in odontogenic commitment. Mechanistically, KDM6B was recruited to bone morphogenic protein 2 (BMP2) promoters and the subsequent removal of silencing H3K27me3 marks led to the activation of this odontogenic master transcription gene. Taken together, our results demonstrated the critical role of a histone demethylase in the epigenetic regulation of odontogenic differentiation of dental MSCs. KDM6B may present as a potential therapeutic target in the regeneration of tooth structures and the repair of craniofacial defects.
Collapse
Affiliation(s)
- Juan Xu
- 1] Department of Stomatology, Chinese People's Liberation Army General Hospital, Beijing, China [2] Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, USA
| | | | | | | |
Collapse
|
102
|
Deng H, Li F, Li H, Deng Y, Liu J, Wang D, Han G, Wang XJ, Zhang Q. CtBP1 overexpression in keratinocytes perturbs skin homeostasis. J Invest Dermatol 2013; 134:1323-1331. [PMID: 24280726 PMCID: PMC4537778 DOI: 10.1038/jid.2013.504] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 09/24/2013] [Accepted: 10/08/2013] [Indexed: 12/20/2022]
Abstract
Carboxyl-terminal binding protein-1 (CtBP1) is a transcriptional co-repressor with multiple in vitro targets, but its in vivo functions are largely unknown. We generated keratinocyte-specific CtBP1 transgenic mice with a keratin 5 promoter (K5.CtBP1) to probe the pathological roles of CtBP1. At transgene expression levels comparable with endogenous CtBP1 in acute skin wounds, K5.CtBP1 epidermis displayed hyperproliferation, loss of E-cadherin, and failed terminal differentiation. Known CtBP1 target genes associated with these processes, e.g., p21, Brca1, and E-cadherin were down-regulated in K5.CtBP1 skin. Surprisingly, K5.CtBP1 pups also exhibited a hair loss phenotype. We found that expression of the Distal-less 3 (Dlx3), a critical regulator of hair follicle differentiation and cycling, was decreased in K5.CtBP1 mice. Molecular studies revealed that CtBP1 directly suppressed Dlx3 transcription. Consistently, K5.CtBP1 mice displayed abnormal hair follicles with decreased expression of Dlx3 downstream targets Gata3, Hoxc13, and hair keratins. In sum, this first CtBP1 transgenic model provides in vivo evidence for certain CtBP1 functions predicted from in vitro studies, reveals to our knowledge previously unreported functions and transcriptional activities of CtBP1 in the context of epithelial-mesenchymal interplay, and suggest CtBP1 has a pathogenesis role in hair follicle morphogenesis and differentiation.
Collapse
Affiliation(s)
- Hui Deng
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Dermatology, The Sixth People's Hospital of Shanghai, Shanghai Jiaotong University, Shanghai, China
| | - Fulun Li
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Dermatology, Yueyang Hospital Affiliated to Shanghai University of TCM, Shanghai, China
| | - Hong Li
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Yu Deng
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Jing Liu
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA
| | - Donna Wang
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA.
| | - Gangwen Han
- Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA
| | - Xiao-Jing Wang
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA.
| | - Qinghong Zhang
- Department of Dermatology, University of Colorado Denver, Aurora, Colorado, USA; Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
103
|
Lian JB, Gordon JA, Stein GS. Redefining the activity of a bone-specific transcription factor: novel insights for understanding bone formation. J Bone Miner Res 2013; 28:2060-3. [PMID: 23966343 DOI: 10.1002/jbmr.2076] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jane B Lian
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | | | | |
Collapse
|
104
|
Kim YI, Lee S, Jung SH, Kim HT, Choi JH, Lee MS, You KH, Yeo SY, Yoo KW, Kwak S, Lee JN, Park R, Choe SK, Kim CH. Establishment of a bone-specific col10a1:GFP transgenic zebrafish. Mol Cells 2013; 36:145-50. [PMID: 23852131 PMCID: PMC3887955 DOI: 10.1007/s10059-013-0117-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/05/2013] [Accepted: 06/10/2013] [Indexed: 01/21/2023] Open
Abstract
During skeletal development, both osteogenic and chondrogenic programs are initiated from multipotent mesenchymal cells, requiring a number of signaling molecules, transcription factors, and downstream effectors to orchestrate the sophisticated process. Col10a1, an important downstream effector gene, has been identified as a marker for maturing chondrocytes in higher vertebrates, such as mammals and birds. In zebrafish, this gene has been shown to be expressed in both osteoblasts and chondrocytes, but no study has reported its role in osteoblast development. To initially delineate the osteogenic program from chondrogenic lineage development, we used the zebrafish col10a1 promoter to establish a transgenic zebrafish expressing a GFP reporter specifically in osteoblast-specific bone structures that do not involve cartilaginous programs. A construct harboring a -2.2-kb promoter region was found to be sufficient to drive the reporter gene in osteoblast-specific bone structures within the endogenous col10a1 expression domain, confirming that separable cis-acting elements exist for distinct cell type-specific expression of col10a1 during zebrafish skeletal development. The -2.2-kb col10a1:GFP transgenic zebrafish marking only bone structures derived from osteoblasts will undoubtedly be an invaluable tool for identifying and characterizing molecular events driving osteoblast development in zebrafish, which may further provide a differential mechanism where col10a1 is involved in the development of chondrocytes undergoing maturation in other vertebrate systems.
Collapse
Affiliation(s)
- Yong-Il Kim
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - Suman Lee
- Department of Biomedical Science, CHA University, Seongnam 463-836,
Korea
| | - Seung-Hyun Jung
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| | - Mi-Sun Lee
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| | - Kwan-Hee You
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| | - Sang-Yeob Yeo
- Department of Biotechnology, Division of Applied Chemistry and Biotechnology, Hanbat National University, Daejeon 305-719,
Korea
| | - Kyeong-Won Yoo
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - SeongAe Kwak
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - Joon No Lee
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - Raekil Park
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - Seong-Kyu Choe
- Center for Metabolic Function Regulation, Department of Microbiology, College of Medicine, Wonkwang University, Iksan 570-749,
Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon 305-764,
Korea
| |
Collapse
|
105
|
Gadi J, Jung SH, Lee MJ, Jami A, Ruthala K, Kim KM, Cho NH, Jung HS, Kim CH, Lim SK. The transcription factor protein Sox11 enhances early osteoblast differentiation by facilitating proliferation and the survival of mesenchymal and osteoblast progenitors. J Biol Chem 2013; 288:25400-25413. [PMID: 23888050 DOI: 10.1074/jbc.m112.413377] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sox11 deletion mice are known to exhibit developmental defects of craniofacial skeletal malformations, asplenia, and hypoplasia of the lung, stomach, and pancreas. Despite the importance of Sox11 in the developing skeleton, the role of Sox11 in osteogenesis has not been studied yet. In this study, we identified that Sox11 is an important transcription factor for regulating the proliferation and survival of osteoblast precursor cells as well as the self-renewal potency of mesenchymal progenitor cells via up-regulation of Tead2. Furthermore, Sox11 also plays an important role in the segregation of functional osteoblast lineage progenitors from osteochondrogenic progenitors. Facilitation of osteoblast differentiation from mesenchymal cells was achieved by enhanced expression of the osteoblast lineage specific transcription factors Runx2 and Osterix. Morpholino-targeted disruption of Sox11 in zebrafish impaired organogenesis, including the bones, which were under mineralized. These results indicated that Sox11 plays a crucial role in the proliferation and survival of mesenchymal and osteoblast precursors by Tead2, and osteogenic differentiation by regulating Runx2 and Osterix.
Collapse
Affiliation(s)
- Jogeswar Gadi
- From the Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine
| | - Seung-Hyun Jung
- the Diabetic Complications Research Center, Division of Traditional Korean Medicine (TKM), Integrated Research, Korea Institute of Oriental Medicine (KIOM), Daejeon, Korea 305-811; the Laboratory of Developmental Genetics, Department of Biology, Chungnam National University, Daejeon, Korea 305-764
| | - Min-Jung Lee
- the Brain Korea 21 Project for Medical Sciences,; the Division of Anatomy and Developmental Biology, Department of Oral Biology, Research Center for Orofacial Hard Tissue Regeneration, College of Dentistry, Yonsei University, Seoul, Korea 120-752, and
| | - Ajita Jami
- From the Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine,; the Brain Korea 21 Project for Medical Sciences
| | - Kalyani Ruthala
- the Brain Korea 21 Project for Medical Sciences,; the Department of Anatomy, Embryology Lab, Yonsei University College of Medicine, Seoul, Korea 120-752
| | - Kyoung-Min Kim
- From the Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine,; the Brain Korea 21 Project for Medical Sciences
| | | | - Han-Sung Jung
- the Division of Anatomy and Developmental Biology, Department of Oral Biology, Research Center for Orofacial Hard Tissue Regeneration, College of Dentistry, Yonsei University, Seoul, Korea 120-752, and
| | - Cheol-Hee Kim
- the Laboratory of Developmental Genetics, Department of Biology, Chungnam National University, Daejeon, Korea 305-764
| | - Sung-Kil Lim
- From the Division of Endocrinology and Endocrine Research Institute, Department of Internal Medicine,; the Brain Korea 21 Project for Medical Sciences,.
| |
Collapse
|
106
|
Matsumoto T, Yamada A, Aizawa R, Suzuki D, Tsukasaki M, Suzuki W, Nakayama M, Maki K, Yamamoto M, Baba K, Kamijo R. BMP-2 Induced Expression of Alx3 That Is a Positive Regulator of Osteoblast Differentiation. PLoS One 2013; 8:e68774. [PMID: 23825702 PMCID: PMC3689002 DOI: 10.1371/journal.pone.0068774] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 06/05/2013] [Indexed: 01/12/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) regulate many aspects of skeletal development, including osteoblast and chondrocyte differentiation, cartilage and bone formation, and cranial and limb development. Among them, BMP-2, one of the most potent osteogenic signaling molecules, stimulates osteoblast differentiation, while it inhibits myogenic differentiation in C2C12 cells. To evaluate genes involved in BMP-2-induced osteoblast differentiation, we performed cDNA microarray analyses to compare BMP-2-treated and -untreated C2C12 cells. We focused on Alx3 (aristaless-like homeobox 3) which was clearly induced during osteoblast differentiation. Alx3, a homeobox gene related to the Drosophilaaristaless gene, has been linked to developmental functions in craniofacial structures and limb development. However, little is known about its direct relationship with bone formation. In the present study, we focused on the mechanisms of Alx3 gene expression and function during osteoblast differentiation induced by BMP-2. In C2C12 cells, BMP-2 induced increase of Alx3 gene expression in both time- and dose-dependent manners through the BMP receptors-mediated SMAD signaling pathway. In addition, silencing of Alx3 by siRNA inhibited osteoblast differentiation induced by BMP-2, as showed by the expressions of alkaline phosphatase (Alp), Osteocalcin, and Osterix, while over-expression of Alx3 enhanced osteoblast differentiation induced by BMP-2. These results indicate that Alx3 expression is enhanced by BMP-2 via the BMP receptors mediated-Smad signaling and that Alx3 is a positive regulator of osteoblast differentiation induced by BMP-2.
Collapse
Affiliation(s)
- Takashi Matsumoto
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Atsushi Yamada
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- * E-mail:
| | - Ryo Aizawa
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Periodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Dai Suzuki
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| | - Masayuki Tsukasaki
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| | - Wataru Suzuki
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Mutsuko Nakayama
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
- Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Koutaro Maki
- Department of Orthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Matsuo Yamamoto
- Department of Periodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Kazuyoshi Baba
- Department of Prosthodontics, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryutaro Kamijo
- Department of Biochemistry, School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
107
|
Tao Y, Wu M, Zhou X, Yin W, Hu B, de Crombrugghe B, Sinha KM, Zang J. Structural insights into histone demethylase NO66 in interaction with osteoblast-specific transcription factor osterix and gene repression. J Biol Chem 2013; 288:16430-16437. [PMID: 23620590 PMCID: PMC3675579 DOI: 10.1074/jbc.m112.446849] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 04/22/2013] [Indexed: 11/06/2022] Open
Abstract
Osterix (Osx) is an osteoblast-specific transcriptional factor and is required for osteoblast differentiation and bone formation. A JmjC domain-containing protein NO66 was previously found to participate in regulation of Osx transcriptional activity and plays an important role in osteoblast differentiation through interaction with Osx. Here, we report the crystal structure of NO66 forming in a functional tetramer. A hinge domain links the N-terminal JmjC domain and C-terminal winged helix-turn-helix domain of NO66, and both domains are essential for tetrameric assembly. The oligomerization interface of NO66 interacts with a conserved fragment of Osx. We show that the hinge domain-dependent oligomerization of NO66 is essential for inhibition of Osx-dependent gene activation. Our findings suggest that homo-oligomerization of JmjC domain containing proteins might play a physiological role through interactions with other regulatory factors during gene expression.
Collapse
Affiliation(s)
- Yue Tao
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | - Minhao Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | - Xing Zhou
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | - Wu Yin
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | - Bin Hu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China
| | | | - Krishna M Sinha
- Endocrine Neoplasia and Hormonal Disorders, The University of Texas M.D. Anderson Cancer Center, Houston, Texas.
| | - Jianye Zang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China; Key Laboratory of Structural Biology, Chinese Academy of Sciences, Hefei, Anhui 230027, China.
| |
Collapse
|
108
|
Liu TM, Lee EH. Transcriptional regulatory cascades in Runx2-dependent bone development. TISSUE ENGINEERING. PART B, REVIEWS 2013; 19:254-263. [PMID: 23150948 PMCID: PMC3627420 DOI: 10.1089/ten.teb.2012.0527] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 11/09/2012] [Indexed: 12/11/2022]
Abstract
The development of the musculoskeletal system is a complex process that involves very precise control of bone formation and growth as well as remodeling during postnatal life. Although the understanding of the transcriptional mechanisms of osteogenesis has increased considerably, the molecular regulatory basis, especially the gene regulatory network of osteogenic differentiation, is still poorly understood. This review provides the reader with an overview of the key transcription factors that govern bone formation, highlighting their function and regulation linked to Runt-related transcription factor 2 (Runx2). Runx2 as the master transcription factor of osteoblast differentiation, Twist, Msh homeobox 2 (Msx2), and promyelocytic leukemia zinc-finger protein (PLZF) acting upstream of Runx2, Osterix (Osx) acting downstream of Runx2, and activating transcription factor 4 (ATF4) and zinc-finger protein 521 (ZFP521) acting as cofactors of Runx2 are discussed, and their relevance for tissue engineering is presented. References are provided for more in-depth personal study.
Collapse
Affiliation(s)
- Tong Ming Liu
- Stem Cell and Developmental Biology, Genome Institute of Singapore, Singapore, Singapore
- Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore
- NUS Tissue Engineering Program (NUSTEP), National University of Singapore, Singapore, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, National University of Singapore, Singapore, Singapore
- NUS Tissue Engineering Program (NUSTEP), National University of Singapore, Singapore, Singapore
| |
Collapse
|
109
|
Choi YH, Choi JH, Oh JW, Lee KY. Calmodulin-dependent kinase II regulates osteoblast differentiation through regulation of Osterix. Biochem Biophys Res Commun 2013; 432:248-55. [PMID: 23402759 DOI: 10.1016/j.bbrc.2013.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 02/02/2013] [Indexed: 11/28/2022]
Abstract
Osterix (Osx), a zinc-finger transcription factor, is required for osteoblast differentiation and new bone formation during embryonic development. Calmodulin-dependent kinase II (CaMKII) acts as a key regulator of osteoblast differentiation. However, the precise molecular signaling mechanisms between Osterix and CaMKII are not known. In this study, we focused on the relationship between Osterix and CaMKII during osteoblast differentiation. We examined the role of the CaMKII pathway in the regulation of protein levels and its transcriptional activity on Osterix. We showed that CaMKII interacts with Osterix by increasing the protein levels and enhancing the transcriptional activity of Osterix. Conversely, CaMKII inhibitor KN-93 decreases the protein levels and increases the stability of Osterix. The siRNA-mediated knockdown of CaMKII decreased the protein levels and transcriptional activity of Osterix. These results suggest that Osterix is a novel target of CaMKII and the activity of Osterix can be modulated by a novel mechanism involving CaMKII during osteoblast differentiation.
Collapse
Affiliation(s)
- You Hee Choi
- College of Pharmacy and Research Institute of Drug Development, Chonnam National University, Gwangju 500-757, Republic of Korea
| | | | | | | |
Collapse
|
110
|
Duverger O, Isaac J, Zah A, Hwang J, Berdal A, Lian JB, Morasso MI. In vivo impact of Dlx3 conditional inactivation in neural crest-derived craniofacial bones. J Cell Physiol 2013; 228:654-64. [PMID: 22886599 PMCID: PMC3514657 DOI: 10.1002/jcp.24175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 08/02/2012] [Indexed: 01/05/2023]
Abstract
Mutations in DLX3 in humans lead to defects in craniofacial and appendicular bones, yet the in vivo activities related to Dlx3 function during normal skeletal development have not been fully elucidated. Here we used a conditional knockout approach to analyze the effects of neural crest deletion of Dlx3 on craniofacial bones development. At birth, mutant mice exhibit a normal overall positioning of the skull bones, but a change in the shape of the calvaria was observed. Molecular analysis of the genes affected in the frontal bones and mandibles from these mice identified several bone markers known to affect bone development, with a strong prediction for increased bone formation and mineralization in vivo. Interestingly, while a subset of these genes were similarly affected in frontal bones and mandibles (Sost, Mepe, Bglap, Alp, Ibsp, Agt), several genes, including Lect1 and Calca, were specifically affected in frontal bones. Consistent with these molecular alterations, cells isolated from the frontal bone of mutant mice exhibited increased differentiation and mineralization capacities ex vivo, supporting cell autonomous defects in neural crest cells. However, adult mutant animals exhibited decreased bone mineral density in both mandibles and calvaria, as well as a significant increase in bone porosity. Together, these observations suggest that mature osteoblasts in the adult respond to signals that regulate adult bone mass and remodeling. This study provides new downstream targets for Dlx3 in craniofacial bone, and gives additional evidence of the complex regulation of bone formation and homeostasis in the adult skeleton.
Collapse
Affiliation(s)
- Olivier Duverger
- Developmental Skin Biology Section, NIAMS, NIH, Bethesda, MD20892, USA
| | - Juliane Isaac
- Developmental Skin Biology Section, NIAMS, NIH, Bethesda, MD20892, USA
| | - Angela Zah
- Developmental Skin Biology Section, NIAMS, NIH, Bethesda, MD20892, USA
| | - Joonsung Hwang
- Developmental Skin Biology Section, NIAMS, NIH, Bethesda, MD20892, USA
| | - Ariane Berdal
- INSERM, UMRS 872, Universités Paris 5 and 6, Team 5, 75006 Paris, France
| | - Jane B. Lian
- Departments of Cell Biology and Orthopedic Surgery, University of Massachusetts Medical School, MA01655, USA
| | - Maria I. Morasso
- Developmental Skin Biology Section, NIAMS, NIH, Bethesda, MD20892, USA
| |
Collapse
|
111
|
Hovhannisyan H, Zhang Y, Hassan MQ, Wu H, Glackin C, Lian JB, Stein JL, Montecino M, Stein GS, van Wijnen AJ. Genomic occupancy of HLH, AP1 and Runx2 motifs within a nuclease sensitive site of the Runx2 gene. J Cell Physiol 2013; 228:313-21. [PMID: 22886425 DOI: 10.1002/jcp.22109] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Epigenetic mechanisms mediating expression of the Runt-related transcription factor Runx2 are critical for controlling its osteogenic activity during skeletal development. Here, we characterized bona fide regulatory elements within 120 kbp of the endogenous bone-related Runx2 promoter (P1) in osteoblasts by genomic DNase I footprinting and chromatin immuno-precipitations (ChIPs). We identified a ~10 kbp genomic domain spanning the P1 promoter that interacts with acetylated histones H3 and H4 reflecting an open chromatin conformation in MC3T3 osteoblasts. This large chromatin domain contains a single major DNaseI hypersensitive (DHS) region that defines a 0.4 kbp "basal core" promoter. This region encompasses two endogenous genomic protein/DNA interaction sites (i.e., footprints at Activating Protein 1 [AP1], E-box and Runx motifs). Helix-Loop-Helix (HLH)/E-box occupancy and presence of the DHS region persists in several mesenchymal cell types, but AP1 site occupancy occurs only during S phase when Runx2 expression is minimal. Point-mutation of the HLH/E box dramatically reduces basal promoter activity. Our results indicate that the Runx2 P1 promoter utilizes two stable principal protein/DNA interaction domains associated with AP1 and HLH factors. These sites function together with dynamic and developmentally responsive sites in a major DHS region to support epigenetic control of bone-specific transcription when osteoblasts transition into a quiescent or differentiated state.
Collapse
Affiliation(s)
- Hayk Hovhannisyan
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Cook D, Genever P. Regulation of Mesenchymal Stem Cell Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:213-29. [DOI: 10.1007/978-94-007-6621-1_12] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
113
|
Lee HL, Woo KM, Ryoo HM, Baek JH. Distal-less homeobox 5 inhibits adipogenic differentiation through the down-regulation of peroxisome proliferator-activated receptor γ expression. J Cell Physiol 2012; 228:87-98. [PMID: 22553076 DOI: 10.1002/jcp.24106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Distal-less homeobox 5 (Dlx5) is a positive regulator of osteoblast differentiation that contains a homeobox domain. Because there are possible reciprocal relationships between osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (MSCs), we examined the regulatory role of Dlx5 in adipogenic differentiation in this study. Adipogenic stimuli suppressed the expression levels of Dlx5 mRNA in mouse bone marrow stromal cells. Over-expression of Dlx5 inhibited adipogenic differentiation in human bone marrow MSCs and 3T3-L1 preadipocytic cells whereas knockdown of Dlx5 enhanced adipogenic differentiation in 3T3-L1 cells. Over-expression of Dlx5 suppressed the expression of adipogenic marker genes, including CCAAT/enhancer-binding protein α (C/EBPα) and peroxisome proliferator-activated receptor γ (PPARγ). Dlx5-mediated suppression of adipogenic differentiation was overcome by over-expression of PPARγ but not by that of cAMP response element binding protein (CREB) or C/EBPα. Dlx5 decreased the transcriptional activity of CREB and C/EBPα in a dose-dependent manner. Dlx5 directly bound to CREB and C/EBPα and prevented them from binding to and subsequently transactivating the PPARγ promoter. These results suggest that Dlx5 plays an important regulatory role in fate determination of bone marrow MSCs toward the osteoblast lineage through the inhibition of adipocyte differentiation as well as the direct stimulation of osteoblast differentiation.
Collapse
Affiliation(s)
- Hye-Lim Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749, Korea
| | | | | | | |
Collapse
|
114
|
Histone demethylases KDM4B and KDM6B promotes osteogenic differentiation of human MSCs. Cell Stem Cell 2012; 11:50-61. [PMID: 22770241 DOI: 10.1016/j.stem.2012.04.009] [Citation(s) in RCA: 267] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 03/03/2012] [Accepted: 04/09/2012] [Indexed: 01/10/2023]
Abstract
Human bone marrow mesenchymal stem/stromal cells (MSCs) are multipotent progenitor cells with multilineage differentiation potentials including osteogenesis and adipogenesis. While significant progress has been made in understanding transcriptional controls of MSC fate, little is known about how MSC differentiation is epigenetically regulated. Here we show that the histone demethylases KDM4B and KDM6B play critical roles in osteogenic commitment of MSCs by removing H3K9me3 and H3K27me3. Depletion of KDM4B or KDM6B significantly reduced osteogenic differentiation and increased adipogenic differentiation. Mechanistically, while KDM6B controlled HOX expression by removing H3K27me3, KDM4B promoted DLX expression by removing H3K9me3. Importantly, H3K27me3- and H3K9me3-positive MSCs of bone marrow were significantly elevated in ovariectomized and aging mice in which adipogenesis was highly active. Since histone demethylases are chemically modifiable, KDM4B and KDM6B may present as therapeutic targets for controlling MSC fate choices and lead to clues for new treatment in metabolic bone diseases such as osteoporosis.
Collapse
|
115
|
Li X, Yang G, Fan M. Effects of Homeobox Gene Distal-less 3 on Proliferation and Odontoblastic Differentiation of Human Dental Pulp Cells. J Endod 2012; 38:1504-10. [DOI: 10.1016/j.joen.2012.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 07/05/2012] [Accepted: 07/26/2012] [Indexed: 10/27/2022]
|
116
|
Kirkham GR, Lovrics A, Byrne HM, Jensen OE, King JR, Shakesheff KM, Buttery LDK. Early gene regulation of osteogenesis in embryonic stem cells. Integr Biol (Camb) 2012; 4:1470-7. [PMID: 23042286 DOI: 10.1039/c2ib20164j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The early gene regulatory networks (GRNs) that mediate stem cell differentiation are complex, and the underlying regulatory associations can be difficult to map accurately. In this study, the expression profiles of the genes Dlx5, Msx2 and Runx2 in mouse embryonic stem cells were monitored over a 48 hour period after exposure to the growth factors BMP2 and TGFβ1. Candidate GRNs of early osteogenesis were constructed based on published experimental findings and simulation results of Boolean and ordinary differential equation models were compared with our experimental data in order to test the validity of these models. Three gene regulatory networks were found to be consistent with the data, one of these networks exhibited sustained oscillation, a behaviour which is consistent with the general view of embryonic stem cell plasticity. The work cycle presented in this paper illustrates how mathematical modelling can be used to elucidate from gene expression profiles GRNs that are consistent with experimental data.
Collapse
Affiliation(s)
- Glen R Kirkham
- Centre for Biomolecular Sciences, University Park, Nottingham, UK.
| | | | | | | | | | | | | |
Collapse
|
117
|
Biodentine Induces Immortalized Murine Pulp Cell Differentiation into Odontoblast-like Cells and Stimulates Biomineralization. J Endod 2012; 38:1220-6. [DOI: 10.1016/j.joen.2012.04.018] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Revised: 04/12/2012] [Accepted: 04/24/2012] [Indexed: 11/22/2022]
|
118
|
Choi YH, Choi HJ, Lee KY, Oh JW. Akt1 regulates phosphorylation and osteogenic activity of Dlx3. Biochem Biophys Res Commun 2012; 425:800-5. [DOI: 10.1016/j.bbrc.2012.07.155] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 07/27/2012] [Indexed: 02/01/2023]
|
119
|
Okamoto H, Matsumi Y, Hoshikawa Y, Takubo K, Ryoke K, Shiota G. Involvement of microRNAs in regulation of osteoblastic differentiation in mouse induced pluripotent stem cells. PLoS One 2012; 7:e43800. [PMID: 22937097 PMCID: PMC3427148 DOI: 10.1371/journal.pone.0043800] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/26/2012] [Indexed: 11/18/2022] Open
Abstract
Backgoround MicroRNAs (miRNAs), which regulate biological processes by annealing to the 3′-untranslated region (3′-UTR) of mRNAs to reduce protein synthesis, have been the subject of recent attention as a key regulatory factor in cell differentiation. The effects of some miRNAs during osteoblastic differentiation have been investigated in mesenchymal stem cells, however they still remains to be determined in pluripotent stem cells. Methodology/Principal Findings Bone morphogenic proteins (BMPs) are potent activators of osteoblastic differentiation. In the present study, we profiled miRNAs during osteoblastic differentiation of mouse induced pluripotent stem (iPS) cells by BMP-4, in which expression of important osteoblastic markers such as Rux2, osterix, osteopontin, osteocalcin, PTHR1 and RANKL were significantly increased. A miRNA array analysis revealed that six miRNAs including miR-10a, miR-10b, miR-19b, miR-9-3p, miR-124a and miR-181a were significantly downregulated. Interestingly, miR-124a and miR-181a directly target the transcription factors Dlx5 and Msx2, both of which were increased by about 80-and 30-fold, respectively. In addition, transfection of miR-124a and miR-181a into mouse osteo-progenitor MC3T3-E1 cells significantly reduced expression of Dlx5, Runx2, osteocalcin and ALP, and Msx2 and osteocalcin, respectively. Finally, transfection of the anti-miRNAs of these six miRNAs, which are predicted to target Dlx5 and Msx2, into mouse iPS cells resulted in a significant increase in several osteoblastic differentiation markers such as Rux2, Msx2 and osteopontin. Conclusions/Significance In the present study, we demonstrate that six miRNAs including miR-10a, miR-10b, miR-19b, miR-9-3p, miR-124a and miR-181a miRNAs, especially miR-124a and miR-181a, are important regulatory factors in osteoblastic differentiation of mouse iPS cells.
Collapse
Affiliation(s)
- Hideharu Okamoto
- Division of Molecular and Genetic Medicine, Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medicine, Tottori University, Yonago, Tottori, Japan
- Division of Oral and Maxillofacial Biopathological Surgery, Department of Medicine of Sensory and Motor Organs, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshiaki Matsumi
- Division of Molecular and Genetic Medicine, Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Yoshiko Hoshikawa
- Division of Molecular and Genetic Medicine, Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Kazuko Takubo
- Division of Oral and Maxillofacial Biopathological Surgery, Department of Medicine of Sensory and Motor Organs, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Kazuo Ryoke
- Division of Oral and Maxillofacial Biopathological Surgery, Department of Medicine of Sensory and Motor Organs, School of Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Goshi Shiota
- Division of Molecular and Genetic Medicine, Department of Genetic Medicine and Regenerative Therapeutics, Graduate School of Medicine, Tottori University, Yonago, Tottori, Japan
- * E-mail:
| |
Collapse
|
120
|
Viale-Bouroncle S, Felthaus O, Schmalz G, Brockhoff G, Reichert TE, Morsczeck C. The transcription factor DLX3 regulates the osteogenic differentiation of human dental follicle precursor cells. Stem Cells Dev 2012; 21:1936-1947. [PMID: 22107079 PMCID: PMC3396153 DOI: 10.1089/scd.2011.0422] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 11/22/2011] [Indexed: 01/28/2023] Open
Abstract
The transcription factor DLX3 plays a decisive role in bone development of vertebrates. In neural-crest derived stem cells from the dental follicle (DFCs), DLX3 is differentially expressed during osteogenic differentiation, while other osteogenic transcription factors such as DLX5 or RUNX2 are not highly induced. DLX3 has therefore a decisive role in the differentiation of DFCs, but its actual biological effects and regulation are unknown. This study investigated the DLX3-regulated processes in DFCs. After DLX3 overexpression, DFCs acquired a spindle-like cell shape with reorganized actin filaments. Here, marker genes for cell morphology, proliferation, apoptosis, and osteogenic differentiation were significantly regulated as shown in a microarray analysis. Further experiments showed that DFCs viability is directly influenced by the expression of DLX3, for example, the amount of apoptotic cells was increased after DLX3 silencing. This transcription factor stimulates the osteogenic differentiation of DFCs and regulates the BMP/SMAD1-pathway. Interestingly, BMP2 did highly induce DLX3 and reverse the inhibitory effect of DLX3 silencing in osteogenic differentiation. However, after DLX3 overexpression in DFCs, a BMP2 supplementation did not improve the expression of DLX3 and the osteogenic differentiation. In conclusion, DLX3 influences cell viability and regulates osteogenic differentiation of DFCs via a BMP2-dependent pathway and a feedback control.
Collapse
Affiliation(s)
- Sandra Viale-Bouroncle
- Department of Oral and Maxillofacial Surgery, University of Regensburg, Regensburg, Germany
- Department of Operative Dentistry and Periodontology, University of Regensburg, Regensburg, Germany
| | - Oliver Felthaus
- Department of Oral and Maxillofacial Surgery, University of Regensburg, Regensburg, Germany
- Department of Operative Dentistry and Periodontology, University of Regensburg, Regensburg, Germany
| | - Gottfried Schmalz
- Department of Operative Dentistry and Periodontology, University of Regensburg, Regensburg, Germany
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University of Regensburg, Regensburg, Germany
| | - Torsten E Reichert
- Department of Oral and Maxillofacial Surgery, University of Regensburg, Regensburg, Germany
| | - Christian Morsczeck
- Department of Oral and Maxillofacial Surgery, University of Regensburg, Regensburg, Germany
- Department of Operative Dentistry and Periodontology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
121
|
Athanassiou-Papaefthymiou M, Kim D, Harbron L, Papagerakis S, Schnell S, Harada H, Papagerakis P. Molecular and circadian controls of ameloblasts. Eur J Oral Sci 2012; 119 Suppl 1:35-40. [PMID: 22243224 DOI: 10.1111/j.1600-0722.2011.00918.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Stage-specific expression of ameloblast-specific genes is controlled by differential expression of transcription factors. In addition, ameloblasts follow daily rhythms in their main activities (i.e. enamel protein secretion and enamel mineralization). This time-related control is orchestrated by oscillations of clock proteins involved in the regulation of circadian rhythms. Our aim was to identify the potential links between daily rhythms and developmental controls of ameloblast differentiation. The effects of the transcription factors distal-less homeobox 3 (Dlx3) and runt-related transcription factor 2 (Runx2), and the clock gene nuclear receptor subfamily 1, group D, member 1 (Nr1d1), on secretory and maturation ameloblasts [using stage-specific markers amelogenin (Amelx), enamelin (Enam), and kallikrein-related peptidase 4 (Klk4)] were evaluated in the HAT-7 ameloblast cell line. Amelx and Enam steady-state mRNA expression levels were down-regulated in Runx2 over-expressing cells and up-regulated in Dlx3 over-expressing cells. In contrast, Klk4 mRNA was up-regulated by both Dlx3 and Runx2. Furthermore, a temporal and spatial relationship between clock genes and ameloblast differentiation markers was detected. Of interest, clock genes not only affected rhythmic expression of ameloblast-specific genes but also influenced the expression of Runx2. Multiscale mathematical modeling is being explored to further understand the temporal and developmental controls of ameloblast differentiation. Our study provides novel insights into the regulatory mechanisms sustaining ameloblast differentiation.
Collapse
Affiliation(s)
- Maria Athanassiou-Papaefthymiou
- Department of Orthodontics and Pediatric Medicine, Center for Computational Medicine and Bioinformatics, University of Michigan Schools of Dentistry and Medicine, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
122
|
Cobourne MT, Sharpe PT. Diseases of the tooth: the genetic and molecular basis of inherited anomalies affecting the dentition. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2012; 2:183-212. [DOI: 10.1002/wdev.66] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
123
|
PKC signaling inhibits osteogenic differentiation through the regulation of Msx2 function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:1225-32. [PMID: 22633971 DOI: 10.1016/j.bbamcr.2012.05.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 04/27/2012] [Accepted: 05/16/2012] [Indexed: 11/23/2022]
Abstract
Protein kinase C (PKC) signaling regulates osteoblast differentiation, but little is known about its downstream effectors. We examined the effect of modulating PKC activity on osteogenic transcription factors and found that the protein level of Msx2 is affected. Msx2 is induced by osteogenic signals such as BMPs and it plays critical roles in bone formation and osteoblast differentiation. Here, we examined the role of PKC signaling in regulating the function of Msx2. We found that the inhibition of PKC signaling enhances osteogenic differentiation in BMP2-stimulated C2C12 cells. Treatment with inhibitors of PKC activity or overexpression of kinase-defective (KD), dominant-negative mutant PKC isoforms strongly reduced the level of Msx2 protein. Several PKC isoforms (α, β, δ, and ζ) interacted with Msx2, and PKCβ phosphorylated Msx2 at Thr135 and Thr141. Msx2 repressed the transcriptional activity of the osteogenic transcription factor Runx2, and this repression was relieved by inhibition of PKC activity or overexpression of the KD mutant PKC isoforms. In addition, PKC prolonged the half-life of Msx2 protein. These results suggest that PKC signaling modulates osteoblast differentiation, at least in part, through the regulation of Msx2.
Collapse
|
124
|
Bolaños A, Hotton D, Ferbus D, Loiodice S, Berdal A, Babajko S. Regulation of calbindin-D(28k) expression by Msx2 in the dental epithelium. J Histochem Cytochem 2012; 60:603-10. [PMID: 22614360 DOI: 10.1369/0022155412450641] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Amelogenesis involves the coordinated expression of a set of molecules that includes enamel matrix proteins and calcium-binding proteins. Msx2 is a member of the divergent homeobox gene family and is instrumental in dental morphogenesis and biomineralization. This study focused on an EF-hand calcium-binding protein, calbindin-D(28k), which is highly expressed in dental epithelium. In vivo data showed that calbindin-D(28k) levels were higher in ameloblasts from Msx2(+/-) mice than Msx2(+/+) mice. Consistent with this finding, calbindin-D(28k) distribution was affected in transgenic mice with ectopic expression in root epithelium in rests of Malassez in Msx2(+/-) and more clearly in Msx2(-/-) mice. In accordance with these in vivo data, calbindin-D(28k) protein and mRNA levels were decreased in LS8 ameloblast-like cells by exogenous Msx2 overexpression. Furthermore, calbindin-D(28k) promoter activity (nt-1075/+34) was specifically diminished in the presence of Msx2 overexpression, showing that Msx2 behave as a transcriptional repressor for calbindin-D(28k) gene expression. In conclusion, Msx2 may control the spatiotemporally restricted frame of calbindin-D(28k) production in the dental epithelium in relation to enamel mineralization, as previously shown for amelogenin.
Collapse
Affiliation(s)
- Alba Bolaños
- Centre de Recherche des Cordeliers, INSERM UMRS 872, Team 5, Laboratory of Molecular Oral Physiopathology, Paris, France
| | | | | | | | | | | |
Collapse
|
125
|
Morozumi A. High concentration of sodium butyrate suppresses osteoblastic differentiation and mineralized nodule formation in ROS17/2.8 cells. J Oral Sci 2012; 53:509-16. [PMID: 22167038 DOI: 10.2334/josnusd.53.509] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Periodontitis is a destructive disease that is likely the result of the activities of different microbial complexes, including anaerobic Gram-negative periodontopathic bacteria. Butyric acid (sodium butyrate; BA) is a major metabolic by-product of anaerobic Gram-negative periodontopathic bacteria present in subgingival plaque. This study was undertaken to examine the effect of BA on the expression of osteogenesis-related transcription factors and mineralized nodule formation in osteoblastic ROS17/2.8 cells. The cells were cultured with 0 (control), 10(-5), 10(-4), or 10(-3) M BA for up to 7 days. The gene and protein expression levels of transcription factors such as Runx2, Osterix, Dlx5, Msx2, and AJ18, as well as extracellular matrix proteins such as bone sialoprotein (BSP) and osteocalcin, were examined using real-time PCR and Western blotting, respectively. Mineralized nodule formation was detected by alizarin red staining. The expression of Runx2, Osterix, Dlx5, and Msx2 decreased significantly in the presence of 10(-3 )M BA compared to the control, whereas AJ18 expression increased significantly. Mineralized nodule formation decreased markedly in the presence of 10(-3) M BA. Alkaline phosphatase activity and the expression of bone sialoprotein and osteocalcin decreased significantly in the presence of 10(-3) M BA compared to the control. These results suggest that 10(-3) M BA suppresses osteoblastic differentiation and mineralized nodule formation in ROS17/2.8 cells.
Collapse
Affiliation(s)
- Akira Morozumi
- Department of Oral Health Sciences, Nihon University School of Dentistry, Tokyo, Japan.
| |
Collapse
|
126
|
Tanimoto Y, Veistinen L, Alakurtti K, Takatalo M, Rice DPC. Prevention of premature fusion of calvarial suture in GLI-Kruppel family member 3 (Gli3)-deficient mice by removing one allele of Runt-related transcription factor 2 (Runx2). J Biol Chem 2012; 287:21429-38. [PMID: 22547067 DOI: 10.1074/jbc.m112.362145] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations in the gene encoding the zinc finger transcription factor GLI3 (GLI-Kruppel family member 3) have been identified in patients with Grieg cephalopolysyndactyly syndrome in which premature fusion of calvarial suture (craniosynostosis) is an infrequent but important feature. Here, we show that Gli3 acts as a repressor in the developing murine calvaria and that Dlx5, Runx2 type II isoform (Runx2-II), and Bmp2 are expressed ectopically in the calvarial mesenchyme, which results in aberrant osteoblastic differentiation in Gli3-deficient mouse (Gli3(Xt-J/Xt-J)) and resulted in craniosynostosis. At the same time, enhanced activation of phospho-Smad1/5/8 (pSmad1/5/8), which is a downstream mediator of canonical Bmp signaling, was observed in Gli3(Xt-J/Xt-J) embryonic calvaria. Therefore, we generated Gli3;Runx2 compound mutant mice to study the effects of decreasing Runx2 dosage in a Gli3(Xt-J/Xt-J) background. Gli3(Xt-J/Xt-J) Runx2(+/-) mice have neither craniosynostosis nor additional ossification centers in interfrontal suture and displayed a normalization of Dlx5, Runx2-II, and pSmad1/5/8 expression as well as sutural mesenchymal cell proliferation. These findings suggest a novel role for Gli3 in regulating calvarial suture development by controlling canonical Bmp-Smad signaling, which integrates a Dlx5/Runx2-II cascade. We propose that targeting Runx2 might provide an attractive way of preventing craniosynostosis in patients.
Collapse
Affiliation(s)
- Yukiho Tanimoto
- Department of Orthodontics, Institute of Dentistry, University of Helsinki, Helsinki 00014, Finland
| | | | | | | | | |
Collapse
|
127
|
Lin GL, Hankenson KD. Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J Cell Biochem 2012; 112:3491-501. [PMID: 21793042 DOI: 10.1002/jcb.23287] [Citation(s) in RCA: 379] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are multipotent progenitors that can commit to osteoblast, chondrocyte, adipocyte, and several other lineages. The proper utilization of stem cells for clinical applications requires an integrated understanding of multiple signal inputs that control maintenance of stemness, proliferation, commitment, and differentiation. Various signaling pathways have been implicated in the regulation of MSC differentiation; however, complexities of pathway interactions, as well as seemingly contradictory results in the literature, create an often confusing and disjointed knowledge base. Several recent publications explore the integration of signaling pathways such as BMP, Wnt, Notch, Hedgehog, and Fibroblast Growth Factors in MSC osteoblast differentiation. The transcription factor Cbfa1/Runx2 has been implicated in these pathways as a potential focal point for signaling integration. This review will outline the current understanding of these pathways and indicate where both spatiotemporal effects during differentiation and comparable experimental conditions need to be considered in order to clarify the outcome(s) of differing regulatory levels of these signaling pathways.
Collapse
Affiliation(s)
- Grace L Lin
- Medical Scientist Training Program, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
128
|
Kim BK, Lee HY, Choi JH, Kim JK, Yoon JB, Yoon SK. Hairless plays a role in formation of inner root sheath via regulation of Dlx3 gene. J Biol Chem 2012; 287:16681-8. [PMID: 22442153 DOI: 10.1074/jbc.m111.320770] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Hairless (Hr), a transcription factor, is expressed in the suprabasal cell layer of the interfollicular epidermis and the lower portion of the hair follicle epithelium, where its expression is dependent on the hair cycle. Recently, we reported a new Hr mutant mouse, Hr(Hp), in which the hairless protein (HR) was overexpressed. In this study, we documented abnormal formation of inner root sheath (IRS), suppressed expression of Dlx3, and IRS keratins in the Hr(Hp)/Hr(Hp) skin. We also found that HR down-regulated Dlx3 mRNA expression through suppression of Dlx3 promoter activity. In addition, we showed that Dlx3 regulated the expression of IRS keratins. Our results demonstrate that regulation of Dlx3 by HR affects the IRS keratin expression, thus modulating the formation of IRS of hair follicle.
Collapse
Affiliation(s)
- Bong-Kyu Kim
- Department of Medical Life Sciences, The Catholic University of Korea, Seoul, 137-701, Korea
| | | | | | | | | | | |
Collapse
|
129
|
Chen H, Kolman K, Lanciloti N, Nerney M, Hays E, Robson C, Chandar N. p53 and MDM2 are involved in the regulation of osteocalcin gene expression. Exp Cell Res 2012; 318:867-76. [PMID: 22405968 DOI: 10.1016/j.yexcr.2012.02.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 11/19/2022]
Abstract
Osteocalcin (OC) is a major noncollagenous bone matrix protein and an osteoblast marker whose expression is limited to mature osteoblasts during the late differentiation stage. In previous studies we have shown osteosarcomas to lose p53 function with a corresponding loss of osteocalcin gene expression. Introduction of wild type p53 resulted in re expression of the osteocalcin gene. Using gel shift and chromatin immunoprecipitation assays, we have identified a putative p53 binding site within the rat OC promoter region and observed an increase in OC promoter activity when p53 accumulates using a CAT assay. The p53 inducible gene Mdm2 is a well-known downstream regulator of p53 levels. Our results showed a synergistic increase in the OC promoter activity when both p53 and MDM2 were transiently overexpressed. We further demonstrate that p53 is not degraded during overexpression of MDM2 protein. Increased OC expression was observed with concomitantly increased p53, VDR, and MDM2 levels in ROS17/2.8 cells during treatment with differentiation promoting (DP) media, but was significantly decreased when co-treated with DP media and the small molecule inhibitor of MDM2-p53 interaction, Nutlin-3. We have also observed a dramatic increase of the OC promoter activity in the presence of p53 and Mdm2 with inclusion of Cbfa-1 and p300 factors. Our results suggest that under some physiological conditions the oncoprotein MDM2 may cooperate with p53 to regulate the osteocalcin gene during osteoblastic differentiation.
Collapse
Affiliation(s)
- Hankui Chen
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | | | | | | | | | | | | |
Collapse
|
130
|
Duverger O, Zah A, Isaac J, Sun HW, Bartels AK, Lian JB, Berdal A, Hwang J, Morasso MI. Neural crest deletion of Dlx3 leads to major dentin defects through down-regulation of Dspp. J Biol Chem 2012; 287:12230-40. [PMID: 22351765 DOI: 10.1074/jbc.m111.326900] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During development, Dlx3 is expressed in ectodermal appendages such as hair and teeth. Thus far, the evidence that Dlx3 plays a crucial role in tooth development comes from reports showing that autosomal dominant mutations in DLX3 result in severe enamel and dentin defects leading to abscesses and infections. However, the normal function of DLX3 in odontogenesis remains unknown. Here, we use a mouse model to demonstrate that the absence of Dlx3 in the neural crest results in major impairment of odontoblast differentiation and dentin production. Mutant mice develop brittle teeth with hypoplastic dentin and molars with an enlarged pulp chamber and underdeveloped roots. Using this mouse model, we found that dentin sialophosphoprotein (Dspp), a major component of the dentin matrix, is strongly down-regulated in odontoblasts lacking Dlx3. Using ChIP-seq, we further demonstrate the direct binding of Dlx3 to the Dspp promoter in vivo. Luciferase reporter assays determined that Dlx3 positively regulates Dspp expression. This establishes a regulatory pathway where the transcription factor Dlx3 is essential in dentin formation by directly regulating a crucial matrix protein.
Collapse
Affiliation(s)
- Olivier Duverger
- Developmental Skin Biology Section, NIAMS, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Gade P, Kalvakolanu DV. Chromatin immunoprecipitation assay as a tool for analyzing transcription factor activity. Methods Mol Biol 2012; 809:85-104. [PMID: 22113270 DOI: 10.1007/978-1-61779-376-9_6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Differential gene expression is facilitated by transcriptional regulatory mechanisms and chromatin modifications through DNA-protein interactions. One of the widely used assays to study this is chromatin immunoprecipitation (ChIP) assay, which enables analysis of association of regulatory molecules to specific promoters and histone modifications in vivo. This is of immense value as ChIP assays can provide glimpse of the regulatory mechanisms involved in gene expression in vivo. This article outlines the general strategies and protocols to study ChIP assays in differential recruitment of transcriptional factors (TFs) and also global analysis of transcription factor recruitment is discussed. Further, the applications of ChIP assays for discovering novel genes that are dependent on specific transcription factors were addressed.
Collapse
Affiliation(s)
- Padmaja Gade
- Department of Microbiology & Immunology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
132
|
Singh M, Del Carpio-Cano FE, Monroy MA, Popoff SN, Safadi FF. Homeodomain transcription factors regulate BMP-2-induced osteoactivin transcription in osteoblasts. J Cell Physiol 2011; 227:390-9. [PMID: 21503878 DOI: 10.1002/jcp.22791] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Osteoactivin (OA) is required for the differentiation of osteoblast cells. OA expression is stimulated by bone morphogenetic protein-2 (BMP-2). BMP-2 recruits homeodomain transcription factors Dlx3, Dlx5, and Msx2 to selectively activate or repress transcription of osteogenic genes and hence tightly regulate their transcription during osteoblast differentiation. Considering the key roles of Dlx3, Dlx5, and Msx2 in osteoblast differentiation, here we hypothesize that homeodomain proteins regulate BMP-2-induced OA transcription during osteoblast differentiation. Four classical homeodomain binding sites were identified in the proximal 0.96 kb region of rat OA promoter. Deletions and mutagenesis studies of the OA promoter region indicated that all four homeodomain binding sites are crucial for BMP-2-induced OA promoter activity. Simultaneous disruption of homeodomain binding sites at -852 and -843 of the transcription start site of OA gene significantly decreased the BMP-2-induced OA transcription and inhibited binding of Dlx3, Dlx5, and Msx2 proteins to the OA promoter. Dlx3 and Dlx5 proteins were found to activate the OA transcription, whereas, Msx2 suppressed BMP-2-induced OA transcription. Using chromatin immunoprecipitation assays, we demonstrated that the OA promoter is predominantly occupied by Dlx3 and Dlx5 during the proliferation and matrix maturation stages of osteoblast differentiation, respectively. During the matrix mineralization stage, BMP-2 robustly enhanced the recruitment of Dlx5 and to a lesser extent of Dlx3 and Msx2 to the OA promoter region. Collectively, our results show that the BMP-2-induced OA transcription is differentially regulated by Dlx3, Dlx5, and Msx2 during osteoblast differentiation.
Collapse
Affiliation(s)
- Maneet Singh
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | |
Collapse
|
133
|
Chen H, Hays E, Liboon J, Neely C, Kolman K, Chandar N. Osteocalcin gene expression is regulated by wild-type p53. Calcif Tissue Int 2011; 89:411-8. [PMID: 21964930 DOI: 10.1007/s00223-011-9533-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 09/03/2011] [Indexed: 01/14/2023]
Abstract
The tumor-suppressor p53 is a transcription factor that regulates a number of genes in the process of cell-cycle inhibition, apoptosis, and DNA damage. Recent studies have revealed a crucial role for p53 in bone remodeling. In our previous studies we have shown that p53 is an important regulator of osteoblast differentiation. In this study we investigated the role of p53 in the regulation of human osteocalcin gene expression. We observed that osteocalcin promoter activity could be upregulated by both exogenous and endogenous p53 and downregulated by p53-specific small interfering RNA. DNA affinity immunoblotting assay showed that p53 can bind to the human osteocalcin promoter in vitro. We further identified a p53 response element within the osteocalcin promoter region using a chromatin immunoprecipitation assay. Furthermore, we observed an additive effect of p53 and VDR on the regulation of osteocalcin promoter activity. Our findings suggest that p53 may directly target the human osteocalcin gene and positively affect osteocalcin gene expression.
Collapse
Affiliation(s)
- Hankui Chen
- Department of Biochemistry, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | | | | | | | | | | |
Collapse
|
134
|
Petri AD, Teixeira LN, Crippa GE, Beloti MM, de Oliveira PT, Rosa AL. Effects of low-level laser therapy on human osteoblastic cells grown on titanium. Braz Dent J 2011; 21:491-8. [PMID: 21271038 DOI: 10.1590/s0103-64402010000600003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to investigate the effects of low-level laser therapy (LLLT) by using gallium aluminum arsenide (GaAlAs) diode laser on human osteoblastic cells grown on titanium (Ti). Osteoblastic cells were obtained by enzymatic digestion of human alveolar bone and cultured on Ti discs for up to 17 days. Cells were exposed to LLLT at 3 J/cm2 (wavelength of 780 nm) at days 3 and 7 and non-irradiated cultures were used as control. LLLT treatment did not influence culture growth, ALP activity, and mineralized matrix formation. Analysis of cultures by epifluorescence microscopy revealed an area without cells in LLLT treated cultures, which was repopulated latter with proliferative and less differentiated cells. Gene expression of ALP, OC, BSP, and BMP-7 was higher in LLLT treated cultures, while Runx2, OPN, and OPG were lower. These results indicate that LLLT modulates cell responses in a complex way stimulating osteoblastic differentiation, which suggests possible benefits on implant osseointegration despite a transient deleterious effect immediately after laser irradiation.
Collapse
Affiliation(s)
- Alice Dias Petri
- Cell Culture Laboratory, Ribeirão Preto Dental School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | | |
Collapse
|
135
|
Xu ZS, Wang XY, Xiao DM, Hu LF, Lu M, Wu ZY, Bian JS. Hydrogen sulfide protects MC3T3-E1 osteoblastic cells against H2O2-induced oxidative damage-implications for the treatment of osteoporosis. Free Radic Biol Med 2011; 50:1314-23. [PMID: 21354302 DOI: 10.1016/j.freeradbiomed.2011.02.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 02/15/2011] [Accepted: 02/16/2011] [Indexed: 01/16/2023]
Abstract
Osteoporosis is a bone disease that leads to an increased risk of fracture. Oxidative damage is an important contributor to the morphological and functional changes in the development of osteoporosis. We found in this study that hydrogen sulfide (H2S), a novel endogenous gaseous mediator, protected MC3T3-E1 osteoblastic cells against hydrogen peroxide (H2O2)-induced oxidative injury. NaHS, an H2S donor, increased cell viability and reduced cell apoptosis caused by H2O2. NaHS also stimulated osteoblast proliferation by enhancing both transcription and activity of alkaline phosphatase in MC3T3-E1 osteoblastic cells. Moreover, treatment with NaHS stimulated the transcriptional level of osteocalcin, the main bone matrix protein, and the protein expression of collagen, a major constituent of bone tissue. The above effects were mediated by the antioxidant effect of H2S. NaHS reversed the reduced superoxide dismutase activity, decreased reactive oxygen species production, and suppressed NADPH oxidase activity in H2O2-treated osteoblasts. In addition, NaHS treatment also produced anti-inflammatory effects via inhibition of the production of nitric oxide and TNF-α, suggesting an anti-inflammatory effect of H2S. Cell viability and Western blotting analysis demonstrated that the protective effects of H2S were mediated by p38 and ERK1/2 MAPKs. In conclusion, H2S protects osteoblastic cells against oxidative stress-induced cell injury and suppression of proliferation and differentiation via a MAPK (p38 and ERK1/2)-dependent mechanism. Our findings suggest that H2S may have a potentially therapeutic value for osteoporosis.
Collapse
Affiliation(s)
- Zhong-Shi Xu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore
| | | | | | | | | | | | | |
Collapse
|
136
|
Sellars EA, Zimmerman SL, Smolarek T, Hopkin RJ. Ventricular noncompaction and absent thumbs in a newborn with tetrasomy 5q35.2-5q35.3: An association with Hunter-McAlpine syndrome? Am J Med Genet A 2011; 155A:1409-13. [DOI: 10.1002/ajmg.a.33997] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 02/13/2011] [Indexed: 11/08/2022]
|
137
|
Di Costanzo A, Festa L, Roscigno G, Vivo M, Pollice A, Morasso M, La Mantia G, Calabrò V. A dominant mutation etiologic for human tricho-dento-osseous syndrome impairs the ability of DLX3 to downregulate ΔNp63α. J Cell Physiol 2011; 226:2189-97. [DOI: 10.1002/jcp.22553] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
138
|
Acquafreda T, Nunes FD, Soprano DR, Soprano KJ. Expression of homeobox genes in oral squamous cell carcinoma cell lines treated with all-trans retinoic acid. J Cell Biochem 2011; 111:1437-44. [PMID: 20830740 DOI: 10.1002/jcb.22871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Oral squamous cell carcinoma (OSCC) may arise from potentially malignant oral lesions. All-trans retinoic acid (atRA), which plays a role in cell growth and differentiation, has been studied as a possible chemotherapeutic agent in the prevention of this progression. While the mechanism by which atRA suppresses cell growth has not been completely elucidated, it is known that homeobox genes are atRA targets. To determine if these genes are involved in the atRA-mediated OSCC growth inhibition, PCR array was performed to evaluate the expression of 84 homeobox genes in atRA-sensitive SCC-25 cells compared to atRA-resistant SCC-9 cells following 7 days with atRA treatment. Results showed that the expression of 8 homeobox genes was downregulated and expression of 4 was upregulated in SCC-25 cells but not in SCC-9 cells. Gene expression levels were confirmed for seven of these genes by RT-qPCR. Expression of three genes that showed threefold downregulation was evaluated in SCC-25 cells treated with atRA for 3, 5, and 7 days. Three different patterns of atRA-dependent gene expression were observed. ALX1 showed downregulation only on day 7. DLX3 showed reduced expression on day 3 and further reduced on day 7. TLX1 showed downregulation only on days 5 and 7. Clearly the expression of homeobox genes is modulated by atRA in OSCC cell lines. However, the time course of this modulation suggests that these genes are not direct targets of atRA mediating OSCC growth suppression. Instead they appear to act as downstream effectors of atRA signaling.
Collapse
Affiliation(s)
- Thais Acquafreda
- Department of Oral Pathology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
139
|
Duverger O, Chen S, Lee D, Li T, Chock P, Morasso M. SUMOylation of DLX3 by SUMO1 promotes its transcriptional activity. J Cell Biochem 2011; 112:445-52. [PMID: 21268066 PMCID: PMC3180851 DOI: 10.1002/jcb.22891] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Small ubiquitin-like modifiers (SUMO) are post-translational modifiers that regulate target protein activity in diverse ways. The most common group of SUMO substrates is transcription factors, whose transcriptional activity can be altered positively or negatively as a result of SUMOylation. DLX3 is a homeodomain transcription factor involved in placental development, in the differentiation of structures involving epithelial-mesenchymal interactions, such as hair, teeth and nails, and in bone mineralization. We identified two potential SUMOylation sites in the N-terminal domain of DLX3 at positions K83 and K112. Among the six members of the Distal-less family, DLX3 is the only member containing these sites, which are highly conserved among vertebrates. Co-expression experiments demonstrated that DLX3 can be SUMOylated by SUMO1. Site-directed mutagenesis of lysines 83 and 112 to arginines (K83R and K112R) demonstrated that only K112 is involved in SUMOylation. Immunocytochemical analysis determined that SUMOylation does not affect DLX3 translocation to the nucleus and favors perinuclear localization. Moreover, using electrophoresis mobility shift assay (EMSA), we found that DLX3 is still able to bind DNA when SUMOylated. Using luciferase reporter assays, we showed that DLX3(K112R) exhibits a significantly lower transcriptional activity compared to DLX3(WT), suggesting that SUMOylation has a positive effect on DLX3 activity. We identified a new level of regulation in the activity of DLX3 that may play a crucial role in the regulation of hair, teeth, and bone development.
Collapse
Affiliation(s)
- O. Duverger
- Developmental Skin Biology Section, NIAMS/NIH, Bethesda, Maryland, USA
| | - S.X. Chen
- Developmental Skin Biology Section, NIAMS/NIH, Bethesda, Maryland, USA
| | - D. Lee
- Developmental Skin Biology Section, NIAMS/NIH, Bethesda, Maryland, USA
| | - T. Li
- Laboratory of Biochemistry, NHLBI/NIH, Bethesda, Maryland, USA
| | - P.B. Chock
- Laboratory of Biochemistry, NHLBI/NIH, Bethesda, Maryland, USA
| | - M.I. Morasso
- Developmental Skin Biology Section, NIAMS/NIH, Bethesda, Maryland, USA
| |
Collapse
|
140
|
Hesse E, Saito H, Kiviranta R, Correa D, Yamana K, Neff L, Toben D, Duda G, Atfi A, Geoffroy V, Horne WC, Baron R. Zfp521 controls bone mass by HDAC3-dependent attenuation of Runx2 activity. ACTA ACUST UNITED AC 2010; 191:1271-83. [PMID: 21173110 PMCID: PMC3010073 DOI: 10.1083/jcb.201009107] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Runx2 is indispensable for osteoblast lineage commitment and early differentiation but also blocks osteoblast maturation, thereby causing bone loss in Runx2 transgenic mice. Zinc finger protein 521 (Zfp521) antagonizes Runx2 in vivo. Eliminating one Zfp521 allele mitigates the cleidocranial dysplasia-like phenotype of newborn Runx2(+/-) mice, whereas overexpressing Zfp521 exacerbates it. Overexpressing Zfp521 also reverses the severe osteopenia of adult Runx2 transgenic mice. Zfp521 binds to both Runx2 and histone deacetylase 3 (HDAC3), promotes their association, and antagonizes Runx2 transcriptional activity in an HDAC3-dependent manner. Mutating the Zfp521 zinc finger domains 6 and 26 reduces the binding of Zfp521 to Runx2 and inhibition of Runx2 activity. These data provide evidence that Zfp521 antagonizes Runx2 in vivo and thereby regulates two stages of osteoblast development, early during mesenchymal cell lineage commitment and later during osteoblast maturation. Thus, the balance and molecular interplay between Zfp521 and Runx2 contribute to the control of osteoblast differentiation, skeletal development, and bone homeostasis.
Collapse
Affiliation(s)
- Eric Hesse
- Department of Medicine, Harvard Medical School, Harvard School of Dental Medicine, Harvard University, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Li Y, Liu J, Hudson M, Kim S, Hatch NE. FGF2 promotes Msx2 stimulated PC-1 expression via Frs2/MAPK signaling. J Cell Biochem 2010; 111:1346-58. [DOI: 10.1002/jcb.22861] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
142
|
Bakshi R, Hassan MQ, Pratap J, Lian JB, Montecino MA, van Wijnen AJ, Stein JL, Imbalzano AN, Stein GS. The human SWI/SNF complex associates with RUNX1 to control transcription of hematopoietic target genes. J Cell Physiol 2010; 225:569-76. [PMID: 20506188 DOI: 10.1002/jcp.22240] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The acute myeloid leukemia 1 (AML1, RUNX1) transcription factor is a key regulator of hematopoietic differentiation that forms multi-protein complexes with co-regulatory proteins. These complexes are assembled at target gene promoters in nuclear microenvironments to mediate phenotypic gene expression and chromatin-related epigenetic modifications. Here, immunofluorescence microscopy and biochemical assays are used to show that RUNX1 associates with the human ATP-dependent SWI/SNF chromatin remodeling complex. The SWI/SNF subunits BRG1 and INI1 bind in vivo to RUNX1 target gene promoters (e.g., GMCSF, IL3, MCSF-R, MIP, and p21). These interactions correlate with histone modifications characteristic of active chromatin, including acetylated H4 and dimethylated H3 lysine 4. Downregulation of RUNX1 by RNA interference diminishes the binding of BRG1 and INI1 at selected target genes. Taken together, our findings indicate that RUNX1 interacts with the human SWI/SNF complex to control hematopoietic-specific gene expression.
Collapse
Affiliation(s)
- Rachit Bakshi
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Lee HL, Yi T, Woo KM, Ryoo HM, Kim GS, Baek JH. Msx2 mediates the inhibitory action of TNF-alpha on osteoblast differentiation. Exp Mol Med 2010; 42:437-45. [PMID: 20440096 DOI: 10.3858/emm.2010.42.6.045] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
TNF-alpha, a proinflammatory cytokine, inhibits osteoblast differentiation under diverse inflammatory conditions; however, the underlying mechanisms in terms of the TNF-alpha signaling pathway remain unclear. In this study, we examined the role of Msx2 in TNF-alpha-mediated inhibition of alkaline phosphatase (ALP) expression and the signaling pathways involved. TNF-alpha down-regulated ALP expression induced by bone morphogenetic protein 2 (BMP2) in C2C12 and Runx2(-/-)calvarial cells. Over-expression of Msx2 suppressed BMP2-induced ALP expression. Furthermore, TNF-alpha induced Msx2 expression, and the knockdown of Msx2 by small interfering RNAs rescued ALP expression, which was inhibited by TNF-alpha. TNF-alpha activated the NF-kappaB and the JNK pathways. Inhibition of NF-kappaB or JNK activation reduced the inhibitory effect of TNF-alpha on ALP expression, whereas TNF-alpha-induced Msx2 expression was only suppressed by the inhibition of the NF-kappaB pathway. Taken together, these results indicate that Msx2 mediates the inhibitory action of TNF-alpha on BMP2-regulated osteoblast differentiation and that the TNF-alpha-activated NF-kappaB pathway is responsible for Msx2 induction.
Collapse
Affiliation(s)
- Hye-Lim Lee
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749, Korea
| | | | | | | | | | | |
Collapse
|
144
|
Choi S, Song I, Feng J, Gao T, Haruyama N, Gautam P, Robey P, Hart TC. Mutant DLX 3 disrupts odontoblast polarization and dentin formation. Dev Biol 2010; 344:682-92. [PMID: 20510228 PMCID: PMC2945701 DOI: 10.1016/j.ydbio.2010.05.499] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 05/17/2010] [Accepted: 05/19/2010] [Indexed: 11/30/2022]
Abstract
Tricho-dento-osseous (TDO) syndrome is an autosomal dominant disorder characterized by abnormalities in the thickness and density of bones and teeth. A 4-bp deletion mutation in the Distal-Less 3 (DLX3) gene is etiologic for most cases of TDO. To investigate the in vivo role of mutant DLX3 (MT-DLX3) on dentin development, we generated transgenic (TG) mice expressing MT-DLX3 driven by a mouse 2.3 Col1A1 promoter. Dentin defects were radiographically evident in all teeth and the size of the nonmineralized pulp was enlarged in TG mice, consistent with clinical characteristics in patients with TDO. High-resolution radiography, microcomputed tomography, and SEM revealed a reduced zone of mineralized dentin with anomalies in the number and organization of dentinal tubules in MT-DLX3 TG mice. Histological and immunohistochemical studies demonstrated that the decreased dentin was accompanied by altered odontoblast cytology that included disruption of odontoblast polarization and reduced numbers of odontoblasts. TUNEL assays indicated enhanced odontoblast apoptosis. Expression levels of the apoptotic marker caspase-3 were increased in odontoblasts in TG mice as well as in odontoblastic-like MDPC-23 cells transfected with MT-DLX3 cDNA. Expression of Runx2, Wnt 10A, and TBC1D19 colocalized with DLX3 expression in odontoblasts, and MT-DLX3 significantly reduced expression of all three genes. TBC1D19 functions in cell polarity and decreased TBC1D19 expression may contribute to the observed disruption of odontoblast polarity and apoptosis. These data indicate that MT-DLX3 acts to disrupt odontoblast cytodifferentiation leading to odontoblast apoptosis, and aberrations of dentin tubule formation and dentin matrix production, resulting in decreased dentin and taurodontism. In summary, this TG model demonstrates that MT-DLX3 has differential effects on matrix production and mineralization in dentin and bone and provides a novel tool for the investigation of odontoblast biology.
Collapse
Affiliation(s)
- S.J. Choi
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - I.S. Song
- Asan Institute of Life Sciences, Laboratory of National Investment Project, Dept. of Rheumatology, Ulsan University Medical School, Seoul, Republic of Korea
| | - J.Q. Feng
- Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX, USA
| | - T. Gao
- Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX, USA
| | - N. Haruyama
- Department of Maxillofacial Orthognathics, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - P. Gautam
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - P.G. Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Thomas C. Hart
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
145
|
Hu R, Li H, Liu W, Yang L, Tan YF, Luo XH. Targeting miRNAs in osteoblast differentiation and bone formation. Expert Opin Ther Targets 2010; 14:1109-20. [DOI: 10.1517/14728222.2010.512916] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
146
|
Breen KM, Thackray VG, Coss D, Mellon PL. Runt-related transcription factors impair activin induction of the follicle-stimulating hormone {beta}-subunit gene. Endocrinology 2010; 151:2669-80. [PMID: 20357224 PMCID: PMC2875819 DOI: 10.1210/en.2009-0949] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 03/04/2010] [Indexed: 02/02/2023]
Abstract
Synthesis of the FSH beta-subunit (FSHbeta) is critical for normal reproduction in mammals, and its expression within the pituitary gonadotrope is tightly regulated by activin. Here we show that Runt-related (RUNX) proteins, transcriptional regulators known to interact with TGFbeta signaling pathways, suppress activin induction of FSHbeta gene expression. Runx2 is expressed within the murine pituitary gland and dramatically represses activin-induced FSHbeta promoter activity, without affecting basal expression in LbetaT2 cells, an immortalized mouse gonadotrope cell line. This repressive effect is specific, because RUNX2 induces LHbeta transcription (with or without activin) and does not interfere with GnRH induction of either gonadotropin beta-subunit gene. Analysis of the murine FSHbeta promoter by transfection and gel shift assays reveals that RUNX2 repression localizes to a Runx-binding element at -159/-153, which is adjacent to a previously recognized region critical for activin induction. Mutation of this -153 activin-response element or, indeed, any of the five activin-responsive regions prevents activin induction and, in fact, RUNX2 suppression, instead converting RUNX2 to an activator of the FSHbeta gene. Although the Runx-binding element is required for RUNX2-mediated repression of FSHbeta induction by either activin or Smad3, confirming a functional role of this novel site, protein interactions in addition to those between RUNX2 and Smads are necessary to account for full repression of activin induction. In summary, the present study provides evidence for Runx2-mediated repression of activin-induced FSHbeta gene expression and reveals the context dependence of Runx2 action in hormonal regulation of the gonadotropin genes.
Collapse
Affiliation(s)
- Kellie M Breen
- Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | | | |
Collapse
|
147
|
Pbx1 represses osteoblastogenesis by blocking Hoxa10-mediated recruitment of chromatin remodeling factors. Mol Cell Biol 2010; 30:3531-41. [PMID: 20439491 DOI: 10.1128/mcb.00889-09] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abdominal-class homeodomain-containing (Hox) factors form multimeric complexes with TALE-class homeodomain proteins (Pbx, Meis) to regulate tissue morphogenesis and skeletal development. Here we have established that Pbx1 negatively regulates Hoxa10-mediated gene transcription in mesenchymal cells and identified components of a Pbx1 complex associated with genes in osteoblasts. Expression of Pbx1 impaired osteogenic commitment of C3H10T1/2 multipotent cells and differentiation of MC3T3-E1 preosteoblasts. Conversely, targeted depletion of Pbx1 by short hairpin RNA (shRNA) increased expression of osteoblast-related genes. Studies using wild-type and mutated osteocalcin and Bsp promoters revealed that Pbx1 acts through a Pbx-binding site that is required to attenuate gene activation by Hoxa10. Chromatin-associated Pbx1 and Hoxa10 were present at osteoblast-related gene promoters preceding gene expression, but only Hoxa10 was associated with these promoters during transcription. Our results show that Pbx1 is associated with histone deacetylases normally linked with chromatin inactivation. Loss of Pbx1 from osteoblast promoters in differentiated osteoblasts was associated with increased histone acetylation and CBP/p300 recruitment, as well as decreased H3K9 methylation. We propose that Pbx1 plays a central role in attenuating the ability of Hoxa10 to activate osteoblast-related genes in order to establish temporal regulation of gene expression during osteogenesis.
Collapse
|
148
|
Saugspier M, Felthaus O, Viale-Bouroncle S, Driemel O, Reichert TE, Schmalz G, Morsczeck C. The differentiation and gene expression profile of human dental follicle cells. Stem Cells Dev 2010; 19:707-717. [PMID: 20491563 DOI: 10.1089/scd.2010.0027] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human dental follicle cells (DFCs) are progenitor cells. Recent studies supposed that osteogenic differentiation of DFCs is controlled by growth factors such as BMP2 and IGF2, but their influence on the differentiation of DFCs has not been investigated in detail. We examined DFCs after the induction of osteogenic differentiation with BMP2, IGF2 and a standard osteogenic differentiation medium (ODM) with dexamethasone. The alkaline phosphatase (ALP) activity and the calcium accumulation demonstrated osteogenic differentiation after all treatments, but with the most effective differentiation by ODM. Interestingly, markers of the process of osteoblast differentiation were much higher up-regulated in BMP2- or IGF2-treated cells than in ODM-treated cells. To evaluate the reason of these differences, we compared genome-wide expression profiles at an early stage of differentiation. Chondroblast markers in BMP2-differentiated cells and general markers for cell differentiation/proliferation in IGF2-treated cells were significantly regulated. However, ODM-treated DFCs expressed late markers of osteogenic-differentiated DFCs such as the transcription factor ZBTB16 that is not expressed in BMP2- or IGF2-differentiated cells. Importantly, although the BMP-antagonist noggin (NOG) diminishes the phosphorylation of SMAD1 in DFCs, it did not inhibit osteogenic differentiation by ODM and the expression of ZBTB16. In conclusion, this study demonstrates that osteogenic differentiation of DFCs can be stimulated with all tested inducers but also independently of BMP signaling. To evaluate this mechanism, the transcription factor ZBTB16 is a target for further investigations.
Collapse
Affiliation(s)
- Michael Saugspier
- Institute of Human Genetics, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
149
|
Wu DD, Zhang YP. Positive selection drives population differentiation in the skeletal genes in modern humans. Hum Mol Genet 2010; 19:2341-6. [PMID: 20233747 DOI: 10.1093/hmg/ddq107] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
During the course of evolution, the human skeletal system has evolved rapidly leading to an incredible array of phenotypic diversity, including variations in height and bone mineral density. However, the genetic basis of this phenotypic diversity and the relatively rapid tempo of evolution have remained largely undocumented. Here, we discover that skeletal genes exhibit a significantly greater level of population differentiation among humans compared with other genes in the genome. The pattern is exceptionally evident at amino acid-altering sites within these genes. Divergence is greater between Africans and both Europeans and East Asians. In contrast, relatively weak differentiation is observed between Europeans and East Asians. SNPs with higher levels of differentiation have correspondingly higher derived allele frequencies in Europeans and East Asians. Thus, it appears that positive selection has operated on skeletal genes in the non-African populations and this may have been initiated with the human colonization of Eurasia. In conclusion, we provide genetic evidence supporting the rapid evolution of the human skeletal system and the associated diversity of phenotypes.
Collapse
Affiliation(s)
- Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | | |
Collapse
|
150
|
Feldman G, Dalsey C, Fertala K, Azimi D, Fortina P, Devoto M, Pacifici M, Parvizi J. The Otto Aufranc Award: Identification of a 4 Mb region on chromosome 17q21 linked to developmental dysplasia of the hip in one 18-member, multigeneration family. Clin Orthop Relat Res 2010; 468:337-44. [PMID: 19756907 PMCID: PMC2807013 DOI: 10.1007/s11999-009-1073-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 08/19/2009] [Indexed: 01/31/2023]
Abstract
Developmental dysplasia of the hip (DDH) is a disabling condition that, depending on geography, can afflict between 20% and 80% of patients with end-stage arthritis of the hip. Despite its prevalence, the etiology of this disease remains unknown. DDH is a complex disorder with both environmental and genetic causes. Based on the literature the candidate genes for the disease are HOXB9, collagen type I alpha1, and DLX 3. The purpose of our study was to map and characterize the gene or genes responsible for this disorder by family linkage analysis. We recruited one 18-member, multigeneration affected family to provide cheek swabs and blood samples for isolation of DNA. Amplified DNA underwent a total genome scan using GeneChip Mapping 250 K Assay (Affymetrix, Santa Clara, CA). We observed only one region with a LOD score greater than 1.5: a 4 Mb region on chromosome 17q21.32, yielding a LOD score of 1.82. While a LOD score of 1.82 does not meet the accepted standard for linkage we interpret these data as suggesting the responsible gene could be linked to this region, which includes a cluster of homeobox genes (HOX genes) that are part of the developmental regulatory system providing cells with specific positional identities along the developing joint and spine. Discovering the genetic basis of the disease would be an important step in understanding the etiology of this disabling condition.
Collapse
Affiliation(s)
- George Feldman
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| | - Chelsea Dalsey
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| | - Kasia Fertala
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| | - David Azimi
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| | - Paolo Fortina
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Marcella Devoto
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Maurizio Pacifici
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| | - Javad Parvizi
- Thomas Jefferson University and the Rothman Institute of Orthopaedics, 925 Chestnut Street, 5th Floor, Philadelphia, PA 19107 USA
| |
Collapse
|