101
|
Soreide E, Denbeigh JM, Lewallen EA, Samsonraj RM, Berglund LJ, Dudakovic A, Cool SM, Nordsletten L, Kakar S, van Wijnen AJ. Fibrin glue mediated delivery of bone anabolic reagents to enhance healing of tendon to bone. J Cell Biochem 2018; 119:5715-5724. [PMID: 29388702 DOI: 10.1002/jcb.26755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/25/2018] [Indexed: 01/12/2023]
Abstract
Tendon graft healing in bone tunnels for the fixation of intra-articular ligament reconstructions may limit clinical outcome by delaying healing. This study assesses the effects of hydrogel-mediated delivery of bone anabolic growth factors in a validated model of tendon-to-bone tunnel healing. Forty-five Wistar rats were randomly allocated into three groups (BMP2-treated, GSK126-treated, and placebo). All animals underwent a tendon-to-bone tunnel reconstruction. Healing was evaluated at 4 weeks by biomechanical assessment, micro-computed tomography (bone mineral density, bone volume, cross sectional area of bone tunnels), and traditional histology. Adverse events associated with the hydrogel-mediated delivery of drugs were not observed. Results of our biomechanical assessment demonstrated favorable trends in animals treated with bone anabolic factors for energy absorption (P = 0.116) and elongation (P = 0.054), while results for force to failure (P = 0.691) and stiffness (P = 0.404) did not show discernible differences. Cross sectional areas for BMP2-treated animals were reduced, but neither BMP2 nor GSK126 administration altered bone mineral density (P = 0.492) or bone volume in the bone tunnel. These results suggest a novel and positive effect of bone anabolic factors on tendon-to-bone tunnel healing. Histological evaluation confirmed absence of collagen fibers crossing the soft tissue-bone interface indicating immature graft integration as expected at this time point. Our study indicates that hydrogel-mediated delivery of BMP2 and GSK126 appears to be safe and has the potential to enhance tendon-to-bone tunnel healing in ligament reconstructions.
Collapse
Affiliation(s)
- Endre Soreide
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Division of Orthopedic Surgery, Oslo University Hospital, Norway & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Janet M Denbeigh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biological Sciences, Hampton University, Hampton, Virginia
| | | | | | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Simon M Cool
- Institute of Medical Biology, A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore
| | - Lars Nordsletten
- Division of Orthopedic Surgery, Oslo University Hospital, Norway & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
102
|
PRC2 Is Dispensable in Vivo for β-Catenin-Mediated Repression of Chondrogenesis in the Mouse Embryonic Cranial Mesenchyme. G3-GENES GENOMES GENETICS 2018; 8:491-503. [PMID: 29223978 PMCID: PMC5919733 DOI: 10.1534/g3.117.300311] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A hallmark of craniofacial development is the differentiation of multiple cell lineages in close proximity to one another. The mouse skull bones and overlying dermis are derived from the cranial mesenchyme (CM). Cell fate selection of the embryonic cranial bone and dermis in the CM requires Wnt/β-catenin signaling, and loss of β-catenin leads to an ectopic chondrogenic cell fate switch. The mechanism by which Wnt/β-catenin activity suppresses the cartilage fate is unclear. Upon conditional deletion of β-catenin in the CM, several key determinants of the cartilage differentiation program, including Sox9, become differentially expressed. Many of these differentially expressed genes are known targets of the Polycomb Repressive Complex 2 (PRC2). Thus, we hypothesized that PRC2 is required for Wnt/β-catenin-mediated repression of chondrogenesis in the embryonic CM. We find that β-catenin can physically interact with PRC2 components in the CM in vivo. However, upon genetic deletion of Enhancer of Zeste homolog 2 (EZH2), the catalytic component of PRC2, chondrogenesis remains repressed and the bone and dermis cell fate is preserved in the CM. Furthermore, loss of β-catenin does not alter either the H3K27me3 enrichment levels genome-wide or on cartilage differentiation determinants, including Sox9. Our results indicate that EZH2 is not required to repress chondrogenesis in the CM downstream of Wnt/β-catenin signaling.
Collapse
|
103
|
Samsonraj RM, Dudakovic A, Manzar B, Sen B, Dietz AB, Cool SM, Rubin J, van Wijnen AJ. Osteogenic Stimulation of Human Adipose-Derived Mesenchymal Stem Cells Using a Fungal Metabolite That Suppresses the Polycomb Group Protein EZH2. Stem Cells Transl Med 2017; 7:197-209. [PMID: 29280310 PMCID: PMC5788881 DOI: 10.1002/sctm.17-0086] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 10/25/2017] [Indexed: 12/19/2022] Open
Abstract
Strategies for musculoskeletal tissue regeneration apply adult mesenchymal stem/stromal cells (MSCs) that can be sourced from bone marrow- and lipo-aspirates. Adipose tissue-derived MSCs are more easily harvested in the large quantities required for skeletal tissue-engineering approaches, but are generally considered to be less osteogenic than bone marrow MSCs. Therefore, we tested a new molecular strategy to improve their osteogenic lineage-differentiation potential using the fungal metabolite cytochalasin D (CytoD). We show that CytoD, which may function by redistributing the intracellular location of β-actin (ACTB), is a potent osteogenic stimulant as reflected by significant increases in alkaline phosphatase activity, extracellular matrix mineralization, and osteoblast-related gene expression (e.g., RUNX2, ALPL, SPARC, and TGFB3). RNA sequencing analyses of MSCs revealed that acute CytoD treatment (24 hours) stimulates a broad program of osteogenic biomarkers and epigenetic regulators. CytoD decreases mRNA and protein levels of the Polycomb chromatin regulator Enhancer of Zeste Homolog 2 (EZH2), which controls heterochromatin formation by mediating trimethylation of histone 3 lysine 27 (H3K27me3). Reduced EZH2 expression decreases cellular H3K27me3 marks indicating a global reduction in heterochromatin. We conclude that CytoD is an effective osteogenic stimulant that mechanistically functions by blocking both cytoplasmic actin polymerization and gene-suppressive epigenetic mechanisms required for the acquisition of the osteogenic phenotype in adipose tissue-derived MSCs. This finding supports the use of CytoD in advancing the osteogenic potential of MSCs in skeletal regenerative strategies. Stem Cells Translational Medicine 2018;7:197-209.
Collapse
Affiliation(s)
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Bushra Manzar
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Buer Sen
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
104
|
Feigenson M, Shull LC, Taylor EL, Camilleri ET, Riester SM, van Wijnen AJ, Bradley EW, Westendorf JJ. Histone Deacetylase 3 Deletion in Mesenchymal Progenitor Cells Hinders Long Bone Development. J Bone Miner Res 2017; 32:2453-2465. [PMID: 28782836 PMCID: PMC5732041 DOI: 10.1002/jbmr.3236] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/19/2017] [Accepted: 08/03/2017] [Indexed: 01/21/2023]
Abstract
Long bone formation is a complex process that requires precise transcriptional control of gene expression programs in mesenchymal progenitor cells. Histone deacetylases (Hdacs) coordinate chromatin structure and gene expression by enzymatically removing acetyl groups from histones and other proteins. Hdac inhibitors are used clinically to manage mood disorders, cancers, and other conditions but are teratogenic to the developing skeleton and increase fracture risk in adults. In this study, the functions of Hdac3, one of the enzymes blocked by current Hdac inhibitor therapies, in skeletal mesenchymal progenitor cells were determined. Homozygous deletion of Hdac3 in Prrx1-expressing cells prevented limb lengthening, altered pathways associated with endochondral and intramembranous bone development, caused perinatal lethality, and slowed chondrocyte and osteoblast differentiation in vitro. Transcriptomic analysis revealed that Hdac3 regulates vastly different pathways in mesenchymal cells expressing the Prxx1-Cre driver than those expressing the Col2-CreERT driver. Notably, Fgf21 was elevated in Hdac3-CKOPrrx1 limbs as well as in chondrogenic cells exposed to Hdac3 inhibitors. Elevated expression of Mmp3 and Mmp10 transcripts was also observed. In conclusion, Hdac3 regulates distinct pathways in mesenchymal cell populations and is required for mesenchymal progenitor cell differentiation and long bone development. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marina Feigenson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lomeli Carpio Shull
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Earnest L Taylor
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
105
|
Li G, Liu M, Zhang S, Wan H, Zhang Q, Yue R, Yan X, Wang X, Wang Z, Sun Y. Essential Role of IFT140 in Promoting Dentinogenesis. J Dent Res 2017; 97:423-431. [PMID: 29195058 DOI: 10.1177/0022034517741283] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Primary cilia, with highly regulated cellular sensory functions, play key roles in tissue development and function maintenance. Intraflagellar transport 140 (IFT140) is a subunit of IFT complex A, which is specialized for retrograde transportation in cilia. Mutations of Ift140 are usually associated with syndromic ciliopathy and may cause isolated diseases such as retinal dystrophy, short ribs, and polycystic kidney. However, the role of IFT140 in tooth development has not been well investigated. In this study, a close relationship between IFT140 and dentin formation is disclosed. During tooth development, IFT140 was highly expressed in odontoblasts. To further understand the role of IFT140 in dentinogenesis, Ift140flox/flox/Osx-Cre mouse was generated. The dentin thickness of Ift140flox/flox/Osx-Cre mouse is thinner and the dentin formation is slower than that in control. In vitro, deletion of IFT140 in odontoblasts led to poor odontogenic differentiation, abnormal primary cilia, and decreased Sonic hedgehog signaling molecules. More important, due to loss of primary cilia in odontoblasts by IFT140 deletion, reparative dentin formation was impaired in a tooth-drilling model. These results suggest that cilia gene IFT140 is essential in promoting dentin formation and reparation.
Collapse
Affiliation(s)
- G Li
- 1 Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - M Liu
- 2 Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai, China
| | - S Zhang
- 1 Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - H Wan
- 1 Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Q Zhang
- 2 Department of Endodontics, School & Hospital of Stomatology, Tongji University, Shanghai, China
| | - R Yue
- 3 School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - X Yan
- 4 State Key Laboratory of Cell Biology, CAS Centre for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - X Wang
- 5 Department of Cell Biology & Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Z Wang
- 1 Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Y Sun
- 1 Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
106
|
Li C, Chai Y, Wang L, Gao B, Chen H, Gao P, Zhou FQ, Luo X, Crane JL, Yu B, Cao X, Wan M. Programmed cell senescence in skeleton during late puberty. Nat Commun 2017; 8:1312. [PMID: 29101351 PMCID: PMC5670205 DOI: 10.1038/s41467-017-01509-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 09/22/2017] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSPCs) undergo rapid self-renewal and differentiation, contributing to fast skeletal growth during childhood and puberty. It remains unclear whether these cells change their properties during late puberty to young adulthood, when bone growth and accrual decelerate. Here we show that MSPCs in primary spongiosa of long bone in mice at late puberty undergo normal programmed senescence, characterized by loss of nestin expression. MSPC senescence is epigenetically controlled by the polycomb histone methyltransferase enhancer of zeste homolog 2 (Ezh2) and its trimethylation of histone H3 on Lysine 27 (H3K27me3) mark. Ezh2 maintains the repression of key cell senescence inducer genes through H3K27me3, and deletion of Ezh2 in early pubertal mice results in premature cellular senescence, depleted MSPCs pool, and impaired osteogenesis as well as osteoporosis in later life. Our data reveals a programmed cell fate change in postnatal skeleton and unravels a regulatory mechanism underlying this phenomenon. Mesenchymal stem cells are essential for bone development, but it is unclear if their activity is maintained after late puberty, when bone growth decelerates. The authors show that during late puberty in mice, these cells undergo senescence under the epigenetic control of Ezh2.
Collapse
Affiliation(s)
- Changjun Li
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu Chai
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lei Wang
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Bo Gao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hao Chen
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Peisong Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.,Department of Pediatric Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Bin Yu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
107
|
Sepulveda H, Aguilar R, Prieto CP, Bustos F, Aedo S, Lattus J, van Zundert B, Palma V, Montecino M. Epigenetic Signatures at the RUNX2-P1 and Sp7 Gene Promoters Control Osteogenic Lineage Commitment of Umbilical Cord-Derived Mesenchymal Stem Cells. J Cell Physiol 2017; 232:2519-2527. [PMID: 27689934 DOI: 10.1002/jcp.25627] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 09/29/2016] [Indexed: 12/16/2023]
Abstract
Wharton's Jelly mesenchymal stem cells (WJ-MSCs) are an attractive potential source of multipotent stem cells for bone tissue replacement therapies. However, the molecular mechanisms involved in their osteogenic conversion are poorly understood. Particularly, epigenetic control operating at the promoter regions of the two master regulators of the osteogenic program, RUNX2/P57 and SP7 has not yet been described in WJ-MSCs. Via quantitative PCR profiling and chromatin immunoprecipitation (ChIP) studies, here we analyze the ability of WJ-MSCs to engage osteoblast lineage. In undifferentiated WJ-MSCs, RUNX2/P57 P1, and SP7 promoters are found deprived of significant levels of the histone post-translational marks that are normally associated with transcriptionally active genes (H3ac, H3K27ac, and H3K4me3). Moreover, the RUNX2 P1 promoter lacks two relevant histone repressive marks (H3K9me3 and H3K27me3). Importantly, RUNX2 P1 promoter is found highly enriched in the H3K4me1 mark, which has been shown recently to mediate gene repression of key regulatory genes. Upon induction of WJ-MSCs osteogenic differentiation, we found that RUNX2/P57, but not SP7 gene expression is strongly activated, in a process that is accompanied by enrichment of activating histone marks (H3K4me3, H3ac, and H3K27ac) at the P1 promoter region. Histone mark analysis showed that SP7 gene promoter is robustly enriched in epigenetic repressive marks that may explain its poor transcriptional response to osteoblast differentiating media. Together, these results point to critical regulatory steps during epigenetic control of WJ-MSCs osteogenic lineage commitment that are relevant for future applications in regenerative medicine. J. Cell. Physiol. 232: 2519-2527, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hugo Sepulveda
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| | - Rodrigo Aguilar
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| | - Catalina P Prieto
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Francisco Bustos
- FONDAP Center for Genome Regulation, Santiago, Chile
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Sócrates Aedo
- Faculty of Medicine, Department of Obstetrics and Gynecology, Campus Oriente, University of Chile, Santiago, Chile
| | - José Lattus
- Faculty of Medicine, Department of Obstetrics and Gynecology, Campus Oriente, University of Chile, Santiago, Chile
| | | | - Veronica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| |
Collapse
|
108
|
Yi X, Jiang X, Li X, Jiang DS. Histone lysine methylation and congenital heart disease: From bench to bedside (Review). Int J Mol Med 2017; 40:953-964. [PMID: 28902362 DOI: 10.3892/ijmm.2017.3115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/21/2017] [Indexed: 11/05/2022] Open
Abstract
Histone post-translational modifications (PTM) as one of the key epigenetic regulatory mechanisms that plays critical role in various biological processes, including regulating chromatin structure dynamics and gene expression. Histone lysine methyltransferase contributes to the establishment and maintenance of differential histone methylation status, which can recognize histone methylated sites and build an association between these modifications and their downstream processes. Recently, it was found that abnormalities in the histone lysine methylation level or pattern may lead to the occurrence of many types of cardiovascular diseases, such as congenital heart disease (CHD). In order to provide new theoretical basis and targets for the treatment of CHD from the view of developmental biology and genetics, this review discusses and elaborates on the association between histone lysine methylation modifications and CHD.
Collapse
Affiliation(s)
- Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ding-Sheng Jiang
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
109
|
Samsonraj RM, Dudakovic A, Zan P, Pichurin O, Cool SM, van Wijnen AJ. A Versatile Protocol for Studying Calvarial Bone Defect Healing in a Mouse Model. Tissue Eng Part C Methods 2017; 23:686-693. [PMID: 28537529 DOI: 10.1089/ten.tec.2017.0205] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal models are vital tools for the preclinical development and testing of therapies aimed at providing solutions for several musculoskeletal disorders. For bone tissue engineering strategies addressing nonunion conditions, rodent models are particularly useful for studying bone healing in a controlled environment. The mouse calvarial defect model permits evaluation of drug, growth factor, or cell transplantation efficacy, together with offering the benefit of utilizing genetic models to study intramembranous bone formation within defect sites. In this study, we describe a detailed methodology for creating calvarial defects in mouse and present our results on bone morphogenetic protein-2-loaded fibrin scaffolds, thus advocating the utility of this functional orthotopic mouse model for the evaluation of therapeutic interventions (such as growth factors or cells) intended for successful bone regeneration therapies.
Collapse
Affiliation(s)
| | - Amel Dudakovic
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Pengfei Zan
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Oksana Pichurin
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Simon M Cool
- 2 Glycotherapeutics Group, Institute of Medical Biology , Agency for Science, Technology and Research (A*STAR), Singapore .,3 Department of Orthopaedic Surgery, National University of Singapore , Singapore
| | - Andre J van Wijnen
- 1 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota.,4 Department of Biochemistry and Molecular Biology, Mayo Clinic , Rochester, Minnesota.,5 Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
110
|
Higashihori N, Lehnertz B, Sampaio A, Underhill T, Rossi F, Richman J. Methyltransferase G9A Regulates Osteogenesis via Twist Gene Repression. J Dent Res 2017. [DOI: 10.1177/0022034517716438] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- N. Higashihori
- Department of Oral Health Sciences, Life Sciences Institute, Faculty of Dentistry, The Biomedical Research Centre, University of British Columbia, Vancouver, Canada
- Department of Medical Genetics, Faculty of Medicine, The Biomedical Research Centre, University of British Columbia, Vancouver, Canada
- Current address: Maxillofacial Orthognathics, Tokyo Medical and Dental University, Tokyo, Japan
| | - B. Lehnertz
- Department of Medical Genetics, Faculty of Medicine, The Biomedical Research Centre, University of British Columbia, Vancouver, Canada
- Current address: Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - A. Sampaio
- Department of Cellular and Physiological Sciences and Biomedical Research Centre, University of British Columbia, Vancouver, Canada
| | - T.M. Underhill
- Department of Cellular and Physiological Sciences and Biomedical Research Centre, University of British Columbia, Vancouver, Canada
| | - F. Rossi
- Department of Medical Genetics, Faculty of Medicine, The Biomedical Research Centre, University of British Columbia, Vancouver, Canada
| | - J.M. Richman
- Department of Oral Health Sciences, Life Sciences Institute, Faculty of Dentistry, The Biomedical Research Centre, University of British Columbia, Vancouver, Canada
| |
Collapse
|
111
|
Yoshioka H, Yoshiko Y. The Roles of Long Non-Protein-Coding RNAs in Osteo-Adipogenic Lineage Commitment. Int J Mol Sci 2017; 18:E1236. [PMID: 28598385 PMCID: PMC5486059 DOI: 10.3390/ijms18061236] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 12/17/2022] Open
Abstract
Osteoblasts and adipocytes share a common mesenchymal progenitor in the bone marrow. This implies that a reciprocal relationship exists between osteogenic and adipogenic differentiation. Further, cells of osteoblast lineage transdifferentiate into adipocytes under some circumstances. Dysregulation of osteo-adipogenic fate-determination leads to bone diseases such as osteoporosis, accompanied by an increase in bone marrow adipose tissue. Thus, the fine-tuning of osteo-adipogenesis is necessary for bone homeostasis. Osteo-adipogenic progression is governed by a complex crosstalk of extrinsic signals, transcription factors, and epigenetic factors. Long non-protein-coding RNAs (lncRNAs) act in part as epigenetic regulators in a broad range of biological activities, such as chromatin organization, transcriptional regulation, post-translational modifications, and histone modification. In this review, we highlight the roles of epigenetic regulators, particularly lncRNAs, in the osteo-adipogenic lineage commitment of bone marrow mesenchymal stem cells and the adipogenic transdifferentiation of osteoblasts.
Collapse
Affiliation(s)
- Hirotaka Yoshioka
- Department of Calcified Tissue Biology, Hiroshima University Institute of Biomedical and Health Sciences, 734-8553 Hiroshima, Japan.
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University Institute of Biomedical and Health Sciences, 734-8553 Hiroshima, Japan.
| |
Collapse
|
112
|
Dudakovic A, van Wijnen AJ. Epigenetic Control of Osteoblast Differentiation by Enhancer of Zeste Homolog 2 (EZH2). ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s40610-017-0064-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
113
|
Farzaneh K, Thaler R, Paradise CR, Deyle DR, Julio MKD, Galindo M, Gordon JA, Stein GS, Dudakovic A, van Wijnen AJ. Histone H4 Methyltransferase Suv420h2 Maintains Fidelity of Osteoblast Differentiation. J Cell Biochem 2017; 118:1262-1272. [PMID: 27862226 PMCID: PMC5357582 DOI: 10.1002/jcb.25787] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022]
Abstract
Osteogenic lineage commitment and progression is controlled by multiple signaling pathways (e.g., WNT, BMP, FGF) that converge on bone-related transcription factors. Access of osteogenic transcription factors to chromatin is controlled by epigenetic regulators that generate post-translational modifications of histones ("histone code"), as well as read, edit and/or erase these modifications. Our understanding of the biological role of epigenetic regulators in osteoblast differentiation remains limited. Therefore, we performed next-generation RNA sequencing (RNA-seq) and established which chromatin-related proteins are robustly expressed in mouse bone tissues (e.g., fracture callus, calvarial bone). These studies also revealed that cells with increased osteogenic potential have higher levels of the H4K20 methyl transferase Suv420h2 compared to other methyl transferases (e.g., Suv39h1, Suv39h2, Suv420h1, Ezh1, Ezh2). We find that all six epigenetic regulators are transiently expressed at different stages of osteoblast differentiation in culture, with maximal mRNAs levels of Suv39h1 and Suv39h2 (at day 3) preceding maximal expression of Suv420h1 and Suv420h2 (at day 7) and developmental stages that reflect, respectively, early and later collagen matrix deposition. Loss of function analysis of Suv420h2 by siRNA depletion shows loss of H4K20 methylation and decreased expression of bone biomarkers (e.g., alkaline phosphatase/Alpl) and osteogenic transcription factors (e.g., Sp7/Osterix). Furthermore, Suv420h2 is required for matrix mineralization during osteoblast differentiation. We conclude that Suv420h2 controls the H4K20 methylome of osteoblasts and is critical for normal progression of osteoblastogenesis. J. Cell. Biochem. 118: 1262-1272, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Khani Farzaneh
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Mario Galindo
- Millennium Institute on Immunology and Immunotherapy, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jonathan A. Gordon
- Department of Biochemistry, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, Vermont 05405
| | - Gary S. Stein
- Department of Biochemistry, University of Vermont College of Medicine, 89 Beaumont Avenue, Burlington, Vermont 05405
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre J. van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Corresponding author: Andre J. van Wijnen, Ph.D., Mayo Clinic, 200 First Street SW, Rochester, MN 55905, Phone: 507- 293-2105, Fax: 507-284-5075,
| |
Collapse
|
114
|
Sen B, Uzer G, Samsonraj RM, Xie Z, McGrath C, Styner M, Dudakovic A, van Wijnen AJ, Rubin J. Intranuclear Actin Structure Modulates Mesenchymal Stem Cell Differentiation. Stem Cells 2017; 35:1624-1635. [PMID: 28371128 DOI: 10.1002/stem.2617] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/19/2017] [Accepted: 03/10/2017] [Indexed: 01/05/2023]
Abstract
Actin structure contributes to physiologic events within the nucleus to control mesenchymal stromal cell (MSC) differentiation. Continuous cytochalasin D (Cyto D) disruption of the MSC actin cytoskeleton leads to osteogenic or adipogenic differentiation, both requiring mass transfer of actin into the nucleus. Cyto D remains extranuclear, thus intranuclear actin polymerization is potentiated by actin transfer: we asked whether actin structure affects differentiation. We show that secondary actin filament branching via the Arp2/3 complex is required for osteogenesis and that preventing actin branching stimulates adipogenesis, as shown by expression profiling of osteogenic and adipogenic biomarkers and unbiased RNA-seq analysis. Mechanistically, Cyto D activates osteoblast master regulators (e.g., Runx2, Sp7, Dlx5) and novel coregulated genes (e.g., Atoh8, Nr4a3, Slfn5). Formin-induced primary actin filament formation is critical for Arp2/3 complex recruitment: osteogenesis is prevented by silencing of the formin mDia1, but not its paralog mDia2. Furthermore, while inhibition of actin, branching is a potent adipogenic stimulus, silencing of either mDia1 or mDia2 blocks adipogenic gene expression. We propose that mDia1, which localizes in the cytoplasm of multipotential MSCs and traffics into the nucleus after cytoskeletal disruption, joins intranuclear mDia2 to facilitate primary filament formation before mediating subsequent branching via Arp2/3 complex recruitment. The resulting intranuclear branched actin network specifies osteogenic differentiation, while actin polymerization in the absence of Arp2/3 complex-mediated secondary branching causes adipogenic differentiation. Stem Cells 2017;35:1624-1635.
Collapse
Affiliation(s)
- Buer Sen
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gunes Uzer
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Mechanical/Biomedical Engineering, Boise State University, Boise, Idaho, USA
| | - Rebekah M Samsonraj
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhihui Xie
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Cody McGrath
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Maya Styner
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
115
|
van de Peppel J, Strini T, Tilburg J, Westerhoff H, van Wijnen AJ, van Leeuwen JP. Identification of Three Early Phases of Cell-Fate Determination during Osteogenic and Adipogenic Differentiation by Transcription Factor Dynamics. Stem Cell Reports 2017; 8:947-960. [PMID: 28344004 PMCID: PMC5390132 DOI: 10.1016/j.stemcr.2017.02.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 01/08/2023] Open
Abstract
Age-related skeletal degeneration in patients with osteoporosis is characterized by decreased bone mass and occurs concomitant with an increase in bone marrow adipocytes. Using microarray expression profiling with high temporal resolution, we identified gene regulatory events in early stages of osteogenic and adipogenic lineage commitment of human mesenchymal stromal cells (hMSCs). Data analysis revealed three distinct phases when cells adopt a committed expression phenotype: initiation of differentiation (0-3 hr, phase I), lineage acquisition (6-24 hr, phase II), and early lineage progression (48-96 hr, phase III). Upstream regulator analysis identified 34 transcription factors (TFs) in phase I with a role in hMSC differentiation. Interestingly, expression levels of identified TFs did not always change and indicate additional post-transcriptional regulatory mechanisms. Functional analysis revealed that forced expression of IRF2 enhances osteogenic differentiation. Thus, IRF2 and other early-responder TFs may control osteogenic cell fate of MSCs and should be considered in mechanistic models that clarify bone-anabolic changes during clinical progression of osteoporosis.
Collapse
Affiliation(s)
- Jeroen van de Peppel
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Tanja Strini
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Julia Tilburg
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands
| | - Hans Westerhoff
- Synthetic Systems Biology, University of Amsterdam, 1081 HZ Amsterdam, the Netherlands; Molecular Cell Physiology, VU University Amsterdam, 1081 HZ Amsterdam, the Netherlands; Systems Biology, MCISB, University of Manchester, Manchester M1 7DN, UK
| | - Andre J van Wijnen
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands; Department of Orthopedic Surgery, Biochemistry & Molecular Biology, and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Johannes P van Leeuwen
- Bone and Calcium Metabolism, Department Internal Medicine, Erasmus MC, Wytemaweg 80, Postbus 2040, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
116
|
Pollock K, Samsonraj RM, Dudakovic A, Thaler R, Stumbras A, McKenna DH, Dosa PI, van Wijnen AJ, Hubel A. Improved Post-Thaw Function and Epigenetic Changes in Mesenchymal Stromal Cells Cryopreserved Using Multicomponent Osmolyte Solutions. Stem Cells Dev 2017; 26:828-842. [PMID: 28178884 DOI: 10.1089/scd.2016.0347] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Current methods for freezing mesenchymal stromal cells (MSCs) result in poor post-thaw function, which limits the clinical utility of these cells. This investigation develops a novel approach to preserve MSCs using combinations of sugars, sugar alcohols, and small-molecule additives. MSCs frozen using these solutions exhibit improved post-thaw attachment and a more normal alignment of the actin cytoskeleton compared to cells exposed to dimethylsulfoxide (DMSO). Osteogenic and chondrogenic differentiation assays show that cells retain their mesenchymal lineage properties. Genomic analysis indicates that the different freezing media evaluated have different effects on the levels of DNA hydroxymethylation, which are a principal epigenetic mark and a key step in the demethylation of CpG doublets. RNA sequencing and quantitative real time-polymerase chain reaction validation demonstrate that transcripts for distinct classes of cytoprotective genes, as well as genes related to extracellular matrix structure and growth factor/receptor signaling are upregulated in experimental freezing solutions compared to DMSO. For example, the osmotic regulator galanin, the antiapoptotic marker B cell lymphoma 2, as well as the cell surface adhesion molecules CD106 (vascular cell adhesion molecule 1) and CD54 (intracellular adhesion molecule 1) are all elevated in DMSO-free solutions. These studies validate the concept that DMSO-free solutions improve post-thaw biological functions and are viable alternatives for freezing MSCs. These novel solutions promote expression of cytoprotective genes, modulate the CpG epigenome, and retain the differentiation ability of MSCs, suggesting that osmolyte-based freezing solutions may provide a new paradigm for therapeutic cell preservation.
Collapse
Affiliation(s)
- Kathryn Pollock
- 1 Department of Biomedical Engineering, University of Minnesota , Minneapolis, Minnesota
| | | | - Amel Dudakovic
- 2 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Roman Thaler
- 2 Department of Orthopedic Surgery, Mayo Clinic , Rochester, Minnesota
| | - Aron Stumbras
- 3 Stem Cell Institute, University of Minnesota , Minneapolis, Minnesota
| | - David H McKenna
- 4 Department of Laboratory Medicine and Pathology, University of Minnesota , Minneapolis, Minnesota
| | - Peter I Dosa
- 5 Institute for Therapeutics Discovery and Development, University of Minnesota , Minneapolis, Minnesota
| | | | - Allison Hubel
- 6 Department of Mechanical Engineering, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
117
|
Wang FS, Lian WS, Lee MS, Weng WT, Huang YH, Chen YS, Sun YC, Wu SL, Chuang PC, Ko JY. Histone demethylase UTX counteracts glucocorticoid deregulation of osteogenesis by modulating histone-dependent and -independent pathways. J Mol Med (Berl) 2017; 95:499-512. [PMID: 28130569 DOI: 10.1007/s00109-017-1512-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/27/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
Abstract
Excess glucocorticoid administration impairs osteogenic activities, which raises the risk of osteoporotic disorders. Epigenetic methylation of DNA and histone regulates the lineage commitment of progenitor cells. This study was undertaken to delineate the actions of histone lysine demethylase 6a (UTX) with regard to the glucocorticoid impediment of osteogenic differentiation. Osteogenic progenitor cells responded to supraphysiological glucocorticoid by elevating CpG dinucleotide methylation proximal to transcription start sites within Runx2 and osterix promoters and Wnt inhibitor Dickkopf-1 (Dkk1) expression concomitant with low UTX expression. 5'-Aza-deoxycystidine demethylation of Runx2 and osterix promoters abolished the glucocorticoid inhibition of mineralized matrix accumulation. Gain of UTX function attenuated the glucocorticoid-induced loss of osteogenic differentiation, whereas UTX silencing escalated adipogenic gene expression and adipocyte formation. UTX sustained osteogenic gene transcription through maintaining its occupancy to Runx2 and osterix promoters. It also mitigated the trimethylation of histone 3 at lysine 27 (H3K27me3), which reduced H3K27me3 enrichment to Dkk1 promoter and thereby lowered Dkk1 transcription. Modulation of β-catenin and Dkk1 actions restored UTX signaling in glucocorticoid-stressed cells. In vivo, UTX inhibition by exogenous methylprednisolone and GSK-J4 administration, an effect that disturbed H3K27me3, β-catenin, Dkk1, Runx2, and osterix levels, exacerbated trabecular microarchitecture loss and marrow adiposity. Taken together, glucocorticoid reduction of UTX function hindered osteogenic differentiation. Epigenetic hypomethylation of osteogenic transcription factor promoters and H3K27 contributed to the UXT alleviation of Dkk1 transcription and osteogenesis in glucocorticoid-stressed osteogenic progenitor cells. Control of UTX action has an epigenetic perspective of curtailing glucocorticoid impairment of osteogenic differentiation and bone mass. KEY MESSAGES UTX attenuates glucocorticoid deregulation of osteogenesis and adipogenesis. UTX reduces Runx2 promoter methylation and H3K27me3 enrichment in the Dkk1 promoter. β-catenin and Dkk1 modulate the glucocorticoid inhibition of UTX signaling. UTX inhibition exacerbates bone mass, trabecular microstructure and fatty marrow. UTX signaling is indispensable in fending off glucocorticoid-impaired osteogenesis.
Collapse
Affiliation(s)
- Feng-Sheng Wang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Wei-Shiung Lian
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Mel S Lee
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Wen-Tsan Weng
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Ying-Hsien Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Yu-Shan Chen
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Yi-Chih Sun
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Shing-Long Wu
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.,Core Laboratory for Phenomics and Diagonistics, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan
| | - Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.
| | - Jih-Yang Ko
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan. .,Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital, 123, Ta-Pei Road, Niao-Sung District, Kaohsiung, 83303, Taiwan.
| |
Collapse
|
118
|
Dudakovic A, Gluscevic M, Paradise CR, Dudakovic H, Khani F, Thaler R, Ahmed FS, Li X, Dietz AB, Stein GS, Montecino MA, Deyle DR, Westendorf JJ, van Wijnen AJ. Profiling of human epigenetic regulators using a semi-automated real-time qPCR platform validated by next generation sequencing. Gene 2017; 609:28-37. [PMID: 28132772 DOI: 10.1016/j.gene.2017.01.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 01/20/2017] [Indexed: 12/11/2022]
Abstract
Epigenetic mechanisms control phenotypic commitment of mesenchymal stromal/stem cells (MSCs) into osteogenic, chondrogenic or adipogenic lineages. To investigate enzymes and chromatin binding proteins controlling the epigenome, we developed a hybrid expression screening strategy that combines semi-automated real-time qPCR (RT-qPCR), next generation RNA sequencing (RNA-seq), and a novel data management application (FileMerge). This strategy was used to interrogate expression of a large cohort (n>300) of human epigenetic regulators (EpiRegs) that generate, interpret and/or edit the histone code. We find that EpiRegs with similar enzymatic functions are variably expressed and specific isoforms dominate over others in human MSCs. This principle is exemplified by analysis of key histone acetyl transferases (HATs) and deacetylases (HDACs), H3 lysine methyltransferases (e.g., EHMTs) and demethylases (KDMs), as well as bromodomain (BRDs) and chromobox (CBX) proteins. Our results show gender-specific expression of H3 lysine 9 [H3K9] demethylases (e.g., KDM5D and UTY) as expected and upregulation of distinct EpiRegs (n>30) during osteogenic differentiation of MSCs (e.g., HDAC5 and HDAC7). The functional significance of HDACs in osteogenic lineage commitment of MSCs was functionally validated using panobinostat (LBH-589). This pan-deacetylase inhibitor suppresses osteoblastic differentiation as evidenced by reductions in bone-specific mRNA markers (e.g., ALPL), alkaline phosphatase activity and calcium deposition (i.e., Alizarin Red staining). Thus, our RT-qPCR platform identifies candidate EpiRegs by expression screening, predicts biological outcomes of their corresponding inhibitors, and enables manipulation of the human epigenome using molecular or pharmacological approaches to control stem cell differentiation.
Collapse
Affiliation(s)
- Amel Dudakovic
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Farzaneh Khani
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Roman Thaler
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Farah S Ahmed
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Xiaodong Li
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Allan B Dietz
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Gary S Stein
- Department of Biochemistry, University of Vermont Medical School, Burlington, VT, USA
| | - Martin A Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | | | - Jennifer J Westendorf
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA; Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
119
|
Maria S, Swanson MH, Enderby LT, D'Amico F, Enderby B, Samsonraj RM, Dudakovic A, van Wijnen AJ, Witt-Enderby PA. Melatonin-micronutrients Osteopenia Treatment Study (MOTS): a translational study assessing melatonin, strontium (citrate), vitamin D3 and vitamin K2 (MK7) on bone density, bone marker turnover and health related quality of life in postmenopausal osteopenic women following a one-year double-blind RCT and on osteoblast-osteoclast co-cultures. Aging (Albany NY) 2017; 9:256-285. [PMID: 28130552 PMCID: PMC5310667 DOI: 10.18632/aging.101158] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/15/2017] [Indexed: 11/25/2022]
Abstract
This one-year double blind randomized control trial assessed the effects of nightly melatonin, strontium (citrate), vitamin D3 and vitamin K2 (MK7; MSDK) on bone mineral density (BMD) and quality of life (QOL) in postmenopausal osteopenic women (ages 49-75). Compared to placebo, MSDK treatment increased BMD in lumbar spine (4.3%) and left femoral neck (2.2%), with an upward trend for total left hip (p=0.069). MSDK increased serum P1NP levels and reduced bone turnover (CTx:P1NP). Psychometric analyses indicated that mood and sleep quality improved for the MSDK group. MSDK-exposed human mesenchymal stem cells (hMSCs) and human peripheral blood monocytes (hPBMCs) plated in transwells or layered demonstrated increases in osteoblastogenesis, decreases in osteoclastogenesis, increases in OPG (TNFRSF11B) and decreases in RANKL (TNFSF11) levels. In transwell osteoblasts, MSDK increased pERK1/2 (MAPK1/MAPK3) and RUNX2 levels; decreased ERK5 (MAPK7); and did not affect the expression of NFκB (NFKB1) and β1integrin (ITGB1). In layered osteoblasts, MSDK also decreased expression of the metabolic proteins PPARγ (PPARG) and GLUT4 (SLC2A4). In adipose-derived human MSCs, MSDK induced osteoblastogenesis. These findings provide both clinical and mechanistic support for the use of MSDK for the prevention or treatment of osteopenia, osteoporosis or other bone-related diseases.
Collapse
Affiliation(s)
- Sifat Maria
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA 15282, USA
| | - Mark H. Swanson
- Functional Medicine, Heart Preventics, LLC, Sequim, WA 98382, USA
| | - Larry T. Enderby
- Enderby Healthcare/Legal Consulting, LLC, Pittsburgh, PA 15102, USA
| | - Frank D'Amico
- Department of Mathematics, Duquesne University School of Liberal Arts, Pittsburgh, PA 15282, USA
| | - Brianna Enderby
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA 15282, USA
| | | | - Amel Dudakovic
- Mayo Clinic, Department of Orthopedic Surgery, Rochester, MN 55905, USA
| | | | - Paula A. Witt-Enderby
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, PA 15282, USA
| |
Collapse
|
120
|
Adamik J, Jin S, Sun Q, Zhang P, Weiss KR, Anderson JL, Silbermann R, Roodman GD, Galson DL. EZH2 or HDAC1 Inhibition Reverses Multiple Myeloma-Induced Epigenetic Suppression of Osteoblast Differentiation. Mol Cancer Res 2017; 15:405-417. [PMID: 28119431 DOI: 10.1158/1541-7786.mcr-16-0242-t] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/16/2016] [Accepted: 12/21/2016] [Indexed: 01/12/2023]
Abstract
In multiple myeloma, osteolytic lesions rarely heal because of persistent suppressed osteoblast differentiation resulting in a high fracture risk. Herein, chromatin immunoprecipitation analyses reveal that multiple myeloma cells induce repressive epigenetic histone changes at the Runx2 locus that prevent osteoblast differentiation. The most pronounced multiple myeloma-induced changes were at the Runx2-P1 promoter, converting it from a poised bivalent state to a repressed state. Previously, it was observed that multiple myeloma induces the transcription repressor GFI1 in osteoblast precursors, which correlates with decreased Runx2 expression, thus prompting detailed characterization of the multiple myeloma and TNFα-dependent GFI1 response element within the Runx2-P1 promoter. Further analyses reveal that multiple myeloma-induced GFI1 binding to Runx2 in osteoblast precursors and recruitment of the histone modifiers HDAC1, LSD1, and EZH2 is required to establish and maintain Runx2 repression in osteogenic conditions. These GFI1-mediated repressive chromatin changes persist even after removal of multiple myeloma. Ectopic GFI1 is sufficient to bind to Runx2, recruit HDAC1 and EZH2, increase H3K27me3 on the gene, and prevent osteogenic induction of endogenous Runx2 expression. Gfi1 knockdown in MC4 cells blocked multiple myeloma-induced recruitment of HDAC1 and EZH2 to Runx2, acquisition of repressive chromatin architecture, and suppression of osteoblast differentiation. Importantly, inhibition of EZH2 or HDAC1 activity in pre-osteoblasts after multiple myeloma exposure in vitro or in osteoblast precursors from patients with multiple myeloma reversed the repressive chromatin architecture at Runx2 and rescued osteoblast differentiation.Implications: This study suggests that therapeutically targeting EZH2 or HDAC1 activity may reverse the profound multiple myeloma-induced osteoblast suppression and allow repair of the lytic lesions. Mol Cancer Res; 15(4); 405-17. ©2017 AACR.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shunqian Jin
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Quanhong Sun
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Peng Zhang
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Cancer Stem Cell Laboratory, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Judith L Anderson
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - Rebecca Silbermann
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana
| | - G David Roodman
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, Indiana. .,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Deborah L Galson
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. .,McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
121
|
Hemming S, Cakouros D, Codrington J, Vandyke K, Arthur A, Zannettino A, Gronthos S. EZH2 deletion in early mesenchyme compromises postnatal bone microarchitecture and structural integrity and accelerates remodeling. FASEB J 2016; 31:1011-1027. [PMID: 27934660 DOI: 10.1096/fj.201600748r] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 11/22/2016] [Indexed: 02/03/2023]
Abstract
In this study, we examined the functional importance of EZH2 during skeletal development and homeostasis using the conditional deletion of Ezh2 (Ezh2fl/fl ) in early mesenchyme with the use of a Prrx-1-cre driver mouse (Ezh2+/+). Heterozygous (Ezh2+/-) newborn and 4-wk-old mice exhibited increased skeletal size, growth plate size, and weight when compared to the wild-type control (Ezh2+/+), whereas homozygous deletion of Ezh2 (Ezh2-/-) resulted in skeletal deformities and reduced skeletal size, growth plate size, and weight in newborn and 4-wk-old mice. Ezh2-/- mice exhibited enhanced trabecular patterning. Osteogenic cortical and trabecular bone formation was enhanced in Ezh2+/- and Ezh2-/- animals. Ezh2+/- and Ezh2-/- mice displayed thinner cortical bone and decreased mechanical strength compared to the wild-type control. Differences in cortical bone thickness were attributed to an increased number of osteoclasts, corresponding with elevated levels of the bone turnover markers cross-linked C-telopeptide-1 and tartrate-resistant acid phosphatase, detected within serum. Moreover, Ezh2+/- mice displayed increased osteoclastogenic potential coinciding with an upregulation of Rankl and M-csf expression by mesenchymal stem cells (MSCs). MSCs isolated from Ezh2+/- mice also exhibited increased trilineage potential compared with wild-type bone marrow stromal/stem cells (BMSCs). Gene expression studies confirmed the upregulation of known Ezh2 target genes in Ezh2-/- bone tissue, many of which are involved in Wnt/BMP signaling as promoters of osteogenesis and inhibitors of adipogenesis. In summary, EZH2 appears to be an important orchestrator of skeletal development, postnatal bone remodelling and BMSC fate determination in vitro and in vivo-Hemming, S., Cakouros, D., Codrington, J., Vandyke, K., Arthur, A., Zannettino, A., Gronthos, S. EZH2 deletion in early mesenchyme compromises postnatal bone microarchitecture and structural integrity and accelerates remodeling.
Collapse
Affiliation(s)
- Sarah Hemming
- Mesenchymal Stem Cell Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Dimitrios Cakouros
- Mesenchymal Stem Cell Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - John Codrington
- School of Mechanical Engineering, University of Adelaide, Adelaide, South Australia, Australia
| | - Kate Vandyke
- Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Myeloma Research Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; and.,South Australia Pathology, Adelaide, South Australia, Australia
| | - Agneiszka Arthur
- Mesenchymal Stem Cell Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Andrew Zannettino
- Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Myeloma Research Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; and
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, South Australia, Australia; .,Cancer Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
122
|
Lui JC, Garrison P, Nguyen Q, Ad M, Keembiyehetty C, Chen W, Jee YH, Landman E, Nilsson O, Barnes KM, Baron J. EZH1 and EZH2 promote skeletal growth by repressing inhibitors of chondrocyte proliferation and hypertrophy. Nat Commun 2016; 7:13685. [PMID: 27897169 PMCID: PMC5477487 DOI: 10.1038/ncomms13685] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022] Open
Abstract
Histone methyltransferases EZH1 and EZH2 catalyse the trimethylation of histone H3 at lysine 27 (H3K27), which serves as an epigenetic signal for chromatin condensation and transcriptional repression. Genome-wide associated studies have implicated EZH2 in the control of height and mutations in EZH2 cause Weaver syndrome, which includes skeletal overgrowth. Here we show that the combined loss of Ezh1 and Ezh2 in chondrocytes severely impairs skeletal growth in mice. Both of the principal processes underlying growth plate chondrogenesis, chondrocyte proliferation and hypertrophy, are compromised. The decrease in chondrocyte proliferation is due in part to derepression of cyclin-dependent kinase inhibitors Ink4a/b, while ineffective chondrocyte hypertrophy is due to the suppression of IGF signalling by the increased expression of IGF-binding proteins. Collectively, our findings reveal a critical role for H3K27 methylation in the regulation of chondrocyte proliferation and hypertrophy in the growth plate, which are the central determinants of skeletal growth.
Collapse
Affiliation(s)
- Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Presley Garrison
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Quang Nguyen
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Michal Ad
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Chithra Keembiyehetty
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg8, Room 1A11, 8 Center Drive, Bethesda, Maryland 20892, USA
| | - Weiping Chen
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg8, Room 1A11, 8 Center Drive, Bethesda, Maryland 20892, USA
| | - Youn Hee Jee
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Ellie Landman
- Division of Pediatric Endocrinology. Q2:08, Department of Women's and Children's Health, Karolinska Institutet and University Hospital, 171 76 Stockholm, Sweden
| | - Ola Nilsson
- Division of Pediatric Endocrinology. Q2:08, Department of Women's and Children's Health, Karolinska Institutet and University Hospital, 171 76 Stockholm, Sweden.,Department of Medical Sciences, Rm C1213, Örebro University and University Hospital, 701 85 Örebro, Sweden
| | - Kevin M Barnes
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| | - Jeffrey Baron
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, CRC, Room 1-3330, 10 Center Drive, MSC-1103, Bethesda, Maryland 20892, USA
| |
Collapse
|
123
|
Dudakovic A, Camilleri ET, Riester SM, Paradise CR, Gluscevic M, O'Toole TM, Thaler R, Evans JM, Yan H, Subramaniam M, Hawse JR, Stein GS, Montecino MA, McGee-Lawrence ME, Westendorf JJ, van Wijnen AJ. Enhancer of Zeste Homolog 2 Inhibition Stimulates Bone Formation and Mitigates Bone Loss Caused by Ovariectomy in Skeletally Mature Mice. J Biol Chem 2016; 291:24594-24606. [PMID: 27758858 DOI: 10.1074/jbc.m116.740571] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/06/2016] [Indexed: 11/06/2022] Open
Abstract
Perturbations in skeletal development and bone degeneration may result in reduced bone mass and quality, leading to greater fracture risk. Bone loss is mitigated by bone protective therapies, but there is a clinical need for new bone-anabolic agents. Previous work has demonstrated that Ezh2 (enhancer of zeste homolog 2), a histone 3 lysine 27 (H3K27) methyltransferase, suppressed differentiation of osteogenic progenitors. Here, we investigated whether inhibition of Ezh2 can be leveraged for bone stimulatory applications. Pharmacologic inhibition and siRNA knockdown of Ezh2 enhanced osteogenic commitment of MC3T3 preosteoblasts. Next generation RNA sequencing of mRNAs and real time quantitative PCR profiling established that Ezh2 inactivation promotes expression of bone-related gene regulators and extracellular matrix proteins. Mechanistically, enhanced gene expression was linked to decreased H3K27 trimethylation (H3K27me3) near transcriptional start sites in genome-wide sequencing of chromatin immunoprecipitations assays. Administration of an Ezh2 inhibitor modestly increases bone density parameters of adult mice. Furthermore, Ezh2 inhibition also alleviated bone loss in an estrogen-deficient mammalian model for osteoporosis. Ezh2 inhibition enhanced expression of Wnt10b and Pth1r and increased the BMP-dependent phosphorylation of Smad1/5. Thus, these data suggest that inhibition of Ezh2 promotes paracrine signaling in osteoblasts and has bone-anabolic and osteoprotective potential in adults.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jared M Evans
- Statistics and Informatics, Mayo Clinic, Rochester, Minnesota 55905
| | - Huihuang Yan
- Statistics and Informatics, Mayo Clinic, Rochester, Minnesota 55905
| | | | | | - Gary S Stein
- the Department of Biochemistry, University of Vermont Medical School, Burlington, Vermont 05405
| | - Martin A Montecino
- the Centro de Investigaciones Biomedicas and FONDAP Center for Genome Regulation, Universidad Andres Bello, 837-0146 Santiago, Chile, and
| | - Meghan E McGee-Lawrence
- the Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia 30912
| | | | - Andre J van Wijnen
- From the Departments of Orthopedic Surgery,; Biochemistry & Molecular Biology,.
| |
Collapse
|
124
|
Guo C, Li C, Yang K, Kang H, Xu X, Xu X, Deng L. Increased EZH2 and decreased osteoblastogenesis during local irradiation-induced bone loss in rats. Sci Rep 2016; 6:31318. [PMID: 27499068 PMCID: PMC4976370 DOI: 10.1038/srep31318] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023] Open
Abstract
Radiation therapy is commonly used to treat cancer patients but exhibits adverse effects, including insufficiency fractures and bone loss. Epigenetic regulation plays an important role in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Here, we reported local bone changes after single-dose exposure to 137CS irradiation in rats. Femur bone mineral density (BMD) and trabecular bone volume in the tibia were significantly decreased at 12 weeks after irradiation. Micro-CT results showed that tBMD, Tb.h and Tb.N were also significantly reduced at 12 weeks after irradiation exposure. ALP-positive OB.S/BS was decreased by 42.3% at 2 weeks after irradiation and was decreased by 50.8% at 12 weeks after exposure. In contrast to the decreased expression of Runx2 and BMP2, we found EZH2 expression was significantly increased at 2 weeks after single-dose 137CS irradiation in BMSCs. Together, our results demonstrated that single-dose 137CS irradiation induces BMD loss and the deterioration of bone microarchitecture in the rat skeleton. Furthermore, EZH2 expression increased and osteoblastogenesis decreased after irradiation. The underlying mechanisms warrant further investigation.
Collapse
Affiliation(s)
- Changjun Guo
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Changwei Li
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Kai Yang
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Hui Kang
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Xiaoya Xu
- Department of Bone Metabolism, Institute of Radiation Medicine, Fudan University, Shanghai 200032, China. Address: No. 2094, Xietu Road, Shanghai 200032 China
| | - Xiangyang Xu
- Department of Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Lianfu Deng
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| |
Collapse
|
125
|
Meyer MB, Benkusky NA, Sen B, Rubin J, Pike JW. Epigenetic Plasticity Drives Adipogenic and Osteogenic Differentiation of Marrow-derived Mesenchymal Stem Cells. J Biol Chem 2016; 291:17829-47. [PMID: 27402842 DOI: 10.1074/jbc.m116.736538] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Indexed: 12/11/2022] Open
Abstract
Terminal differentiation of multipotent stem cells is achieved through a coordinated cascade of activated transcription factors and epigenetic modifications that drive gene transcription responsible for unique cell fate. Within the mesenchymal lineage, factors such as RUNX2 and PPARγ are indispensable for osteogenesis and adipogenesis, respectively. We therefore investigated genomic binding of transcription factors and accompanying epigenetic modifications that occur during osteogenic and adipogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (MSCs). As assessed by ChIP-sequencing and RNA-sequencing analyses, we found that genes vital for osteogenic identity were linked to RUNX2, C/EBPβ, retinoid X receptor, and vitamin D receptor binding sites, whereas adipocyte differentiation favored PPARγ, retinoid X receptor, C/EBPα, and C/EBPβ binding sites. Epigenetic marks were clear predictors of active differentiation loci as well as enhancer activities and selective gene expression. These marrow-derived MSCs displayed an epigenetic pattern that suggested a default preference for the osteogenic pathway; however, these patterns were rapidly altered near the Adipoq, Cidec, Fabp4, Lipe, Plin1, Pparg, and Cebpa genes during adipogenic differentiation. Surprisingly, we found that these cells also exhibited an epigenetic plasticity that enabled them to trans-differentiate from adipocytes to osteoblasts (and vice versa) after commitment, as assessed by staining, gene expression, and ChIP-quantitative PCR analysis. The osteogenic default pathway may be subverted during pathological conditions, leading to skeletal fragility and increased marrow adiposity during aging, estrogen deficiency, and skeletal unloading. Taken together, our data provide an increased mechanistic understanding of the epigenetic programs necessary for multipotent differentiation of MSCs that may prove beneficial in the development of therapeutic strategies.
Collapse
Affiliation(s)
- Mark B Meyer
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706 and
| | - Nancy A Benkusky
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706 and
| | - Buer Sen
- the Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27514
| | - Janet Rubin
- the Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27514
| | - J Wesley Pike
- From the Department of Biochemistry, University of Wisconsin, Madison, Wisconsin 53706 and
| |
Collapse
|
126
|
Aguilar R, Bustos FJ, Saez M, Rojas A, Allende ML, van Wijnen AJ, van Zundert B, Montecino M. Polycomb PRC2 complex mediates epigenetic silencing of a critical osteogenic master regulator in the hippocampus. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1043-55. [PMID: 27216774 DOI: 10.1016/j.bbagrm.2016.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022]
Abstract
During hippocampal neuron differentiation, the expression of critical inducers of non-neuronal cell lineages must be efficiently silenced. Runx2 transcription factor is the master regulator of mesenchymal cells responsible for intramembranous osteoblast differentiation and formation of the craniofacial bone tissue that surrounds and protects the central nervous system (CNS) in mammalian embryos. The molecular mechanisms that mediate silencing of the Runx2 gene and its downstream target osteogenic-related genes in neuronal cells have not been explored. Here, we assess the epigenetic mechanisms that mediate silencing of osteoblast-specific genes in CNS neurons. In particular, we address the contribution of histone epigenetic marks and histone modifiers on the silencing of the Runx2/p57 bone-related isoform in rat hippocampal tissues at embryonic to adult stages. Our results indicate enrichment of repressive chromatin histone marks and of the Polycomb PRC2 complex at the Runx2/p57 promoter region. Knockdown of PRC2 H3K27-methyltransferases Ezh2 and Ezh1, or forced expression of the Trithorax/COMPASS subunit Wdr5 activates Runx2/p57 mRNA expression in both immature and mature hippocampal cells. Together these results indicate that complementary epigenetic mechanisms progressively and efficiently silence critical osteoblastic genes during hippocampal neuron differentiation.
Collapse
Affiliation(s)
- Rodrigo Aguilar
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile; FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile
| | - Fernando J Bustos
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile; FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile
| | - Mauricio Saez
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile; FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile
| | - Adriana Rojas
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile; FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago 7800003, Chile
| | | | - Brigitte van Zundert
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile
| | - Martin Montecino
- Center for Biomedical Research, Universidad Andres Bello, Santiago 8370146, Chile; FONDAP Center for Genome Regulation, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago 8370146, Chile.
| |
Collapse
|
127
|
Lewallen EA, Jones DL, Dudakovic A, Thaler R, Paradise CR, Kremers HM, Abdel MP, Kakar S, Dietz AB, Cohen RC, Lewallen DG, van Wijnen AJ. Osteogenic potential of human adipose-tissue-derived mesenchymal stromal cells cultured on 3D-printed porous structured titanium. Gene 2016; 581:95-106. [PMID: 26774799 PMCID: PMC5054723 DOI: 10.1016/j.gene.2016.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 01/12/2016] [Indexed: 01/08/2023]
Abstract
Integration of porous metal prosthetics, which restore form and function of irreversibly damaged joints, into remaining healthy bone is critical for implant success. We investigated the biological properties of adipose-tissue-derived mesenchymal stromal/stem cells (AMSCs) and addressed their potential to alter the in vitro microenvironment of implants. We employed human AMSCs as a practical source for musculoskeletal applications because these cells can be obtained in large quantities, are multipotent, and have trophic paracrine functions. AMSCs were cultured on surgical-grade porous titanium disks as a model for orthopedic implants. We monitored cell/substrate attachment, cell proliferation, multipotency, and differentiation phenotypes of AMSCs upon osteogenic induction. High-resolution scanning electron microscopy and histology revealed that AMSCs adhere to the porous metallic surface. Compared to standard tissue culture plastic, AMSCs grown in the porous titanium microenvironment showed differences in temporal expression for genes involved in cell cycle progression (CCNB2, HIST2H4), extracellular matrix production (COL1A1, COL3A1), mesenchymal lineage identity (ACTA2, CD248, CD44), osteoblastic transcription factors (DLX3, DLX5, ID3), and epigenetic regulators (EZH1, EZH2). We conclude that metal orthopedic implants can be effectively seeded with clinical-grade stem/stromal cells to create a pre-conditioned implant.
Collapse
Affiliation(s)
- Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Dakota L Jones
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA; Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Christopher R Paradise
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Hilal M Kremers
- Department of Health Sciences Research, College of Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Sanjeev Kakar
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First St. SW Rochester, MN 55905, USA
| | - Robert C Cohen
- Stryker Orthopedics, 325 Corporate Drive, Mahwah, NJ 07430, USA
| | - David G Lewallen
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA.
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA; Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905, USA.
| |
Collapse
|
128
|
Fang C, Qiao Y, Mun SH, Lee MJ, Murata K, Bae S, Zhao B, Park-Min KH, Ivashkiv LB. Cutting Edge: EZH2 Promotes Osteoclastogenesis by Epigenetic Silencing of the Negative Regulator IRF8. THE JOURNAL OF IMMUNOLOGY 2016; 196:4452-4456. [PMID: 27183582 DOI: 10.4049/jimmunol.1501466] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 04/04/2016] [Indexed: 11/19/2022]
Abstract
Osteoclasts are resorptive cells that are important for homeostatic bone remodeling and pathological bone resorption. Emerging evidence suggests an important role for epigenetic mechanisms in osteoclastogenesis. A recent study showed that epigenetic silencing of the negative regulator of osteoclastogenesis Irf8 by DNA methylation is required for osteoclast differentiation. In this study, we investigated the role of EZH2, which epigenetically silences gene expression by histone methylation, in osteoclastogenesis. Inhibition of EZH2 by the small molecule GSK126, or decreasing its expression using antisense oligonucleotides, impeded osteoclast differentiation. Mechanistically, EZH2 was recruited to the IRF8 promoter after RANKL stimulation to deposit the negative histone mark H3K27me3 and downregulate IRF8 expression. GSK126 attenuated bone loss in the ovariectomy mouse model of postmenopausal osteoporosis. Our findings provide evidence for an additional mechanism of epigenetic IRF8 silencing during osteoclastogenesis that likely works cooperatively with DNA methylation, further emphasizing the importance of IRF8 as a negative regulator of osteoclastogenesis.
Collapse
Affiliation(s)
- Celestia Fang
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Yu Qiao
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Se Hwan Mun
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Min Joon Lee
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Koichi Murata
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Seyeon Bae
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery.,Department of Medicine Weill Cornell Medical College
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery.,Department of Medicine Weill Cornell Medical College
| | - Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program David Z. Rosensweig Genomics Research Center Hospital for Special Surgery.,Department of Medicine Weill Cornell Medical College.,Graduate Program in Immunology and Microbial Pathogenesis Weill Cornell Graduate School of Medical Sciences
| |
Collapse
|
129
|
Thaler R, Maurizi A, Roschger P, Sturmlechner I, Khani F, Spitzer S, Rumpler M, Zwerina J, Karlic H, Dudakovic A, Klaushofer K, Teti A, Rucci N, Varga F, van Wijnen AJ. Anabolic and Antiresorptive Modulation of Bone Homeostasis by the Epigenetic Modulator Sulforaphane, a Naturally Occurring Isothiocyanate. J Biol Chem 2016; 291:6754-71. [PMID: 26757819 DOI: 10.1074/jbc.m115.678235] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Indexed: 11/06/2022] Open
Abstract
Bone degenerative pathologies like osteoporosis may be initiated by age-related shifts in anabolic and catabolic responses that control bone homeostasis. Here we show that sulforaphane (SFN), a naturally occurring isothiocyanate, promotes osteoblast differentiation by epigenetic mechanisms. SFN enhances active DNA demethylation viaTet1andTet2and promotes preosteoblast differentiation by enhancing extracellular matrix mineralization and the expression of osteoblastic markers (Runx2,Col1a1,Bglap2,Sp7,Atf4, andAlpl). SFN decreases the expression of the osteoclast activator receptor activator of nuclear factor-κB ligand (RANKL) in osteocytes and mouse calvarial explants and preferentially induces apoptosis in preosteoclastic cells via up-regulation of theTet1/Fas/Caspase 8 and Caspase 3/7 pathway. These mechanistic effects correlate with higher bone volume (∼20%) in both normal and ovariectomized mice treated with SFN for 5 weeks compared with untreated mice as determined by microcomputed tomography. This effect is due to a higher trabecular number in these mice. Importantly, no shifts in mineral density distribution are observed upon SFN treatment as measured by quantitative backscattered electron imaging. Our data indicate that the food-derived compound SFN epigenetically stimulates osteoblast activity and diminishes osteoclast bone resorption, shifting the balance of bone homeostasis and favoring bone acquisition and/or mitigation of bone resorptionin vivo Thus, SFN is a member of a new class of epigenetic compounds that could be considered for novel strategies to counteract osteoporosis.
Collapse
Affiliation(s)
- Roman Thaler
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria, Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Antonio Maurizi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Paul Roschger
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Ines Sturmlechner
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Farzaneh Khani
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Silvia Spitzer
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Monika Rumpler
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Jochen Zwerina
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Heidrun Karlic
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Amel Dudakovic
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Klaus Klaushofer
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Franz Varga
- From the Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of Social Health Insurance Vienna (WGKK) and Austrian Social Insurance for Occupational Risks (AUVA) Trauma Center Meidling, First Medical Department, Hanusch Hospital, 1140 Vienna, Austria,
| | - Andre J van Wijnen
- Department of Orthopedic Surgery and Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, and
| |
Collapse
|