151
|
Li X, Garcia J, Lu J, Iriana S, Kalajzic I, Rowe D, Demer LL, Tintut Y. Roles of parathyroid hormone (PTH) receptor and reactive oxygen species in hyperlipidemia-induced PTH resistance in preosteoblasts. J Cell Biochem 2014; 115:179-88. [PMID: 24038594 DOI: 10.1002/jcb.24648] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 08/13/2013] [Indexed: 11/08/2022]
Abstract
Bioactive lipids initiate inflammatory reactions leading to pathogenesis of atherosclerosis. Evidence shows that they also contribute to bone loss by inhibiting parathyroid hormone receptor (PTH1R) expression and differentiation of osteoblasts. We previously demonstrated that bone anabolic effects of PTH(1-34) are blunted in hyperlipidemic mice and that these PTH effects are restored by antioxidants. However, it is not clear which osteoblastic cell developmental stage is targeted by bioactive lipids. To investigate the effects of hyperlipidemia at the cellular level, hyperlipidemic Ldlr(-/-) mice were bred with Col3.6GFPtpz mice, in which preosteoblasts/osteoblasts carry a topaz fluorescent label, and with Col2.3GFPcyan mice, in which more mature osteoblasts/osteocytes carry a cyan fluorescent label. Histological analyses of trabecular bone surfaces in femoral as well as calvarial bones showed that intermittent PTH(1-34) increased fluorescence intensity in WT-Tpz mice, but not in Tpz-Ldlr(-/-) mice. In contrast, PTH(1-34) did not alter fluorescence intensity in femoral cortical envelopes of either WT-Cyan or Ldlr(-/-)-Cyan mice. To test the mechanism of PTH1R downregulation, preosteoblastic MC3T3-E1 cells were treated with bioactive lipids and the antioxidant Trolox. Results showed that inhibitory effects of PTH1R levels by bioactive lipids were rescued by pretreatment with Trolox. The inhibitory effects on expression of PTH1R as well as on PTH-induced osteoblastic genes were mimicked by xanthine/xanthine oxidase, a known generator of reactive oxygen species. These findings suggest an important role of the preosteoblastic development stage as the target and downregulation of PTH receptor expression mediated by intracellular oxidant stress as a mechanism in hyperlipidemia-induced PTH resistance.
Collapse
Affiliation(s)
- Xin Li
- Department of Medicine, University of California, Los Angeles, California
| | | | | | | | | | | | | | | |
Collapse
|
152
|
Alaee F, Hong SH, Dukas AG, Pensak MJ, Rowe DW, Lieberman JR. Evaluation of osteogenic cell differentiation in response to bone morphogenetic protein or demineralized bone matrix in a critical sized defect model using GFP reporter mice. J Orthop Res 2014; 32:1120-8. [PMID: 24888702 DOI: 10.1002/jor.22657] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 05/08/2014] [Indexed: 02/04/2023]
Abstract
We evaluated the osteoprogenitor response to rhBMP-2 and DBM in a transgenic mouse critical sized defect. The mice expressed Col3.6GFPtopaz (a pre-osteoblastic marker), Col2.3GFPemerald (an osteoblastic marker) and α-smooth muscle actin (α-SMA-Cherry, a pericyte/myofibroblast marker). We assessed defect healing at various time points using radiographs, frozen, and conventional histologic analyses. GFP signal in regions of interest corresponding to the areas of new bone formation was quantified using a novel computer assisted algorithm. All defects treated with rhBMP-2 healed. In contrast, the majority of the defects in the DBM (27/30) and control (28/30) groups did not heal. Quantitation of pre-osteoblasts demonstrated a maximal response (% GFP + cells/TV) in the Col3.6GFPtopaz mice at day 7 (7.2% ± 6.0, p < 0.05 compared to days 14, 21, 28, and 56). The maximal response of the Col2.3GFP cells was seen at days 14 (8.04% ± 5.0) and 21 (8.31% ± 4.32), p < 0.05. In contrast, DBM and control groups showed a limited osteogenic response at all time points. In conclusion, we demonstrated that the BMP and DBM induce vastly different osteogenic responses which should influence their clinical application as bone graft substitutes.
Collapse
Affiliation(s)
- Farhang Alaee
- Department of Orthopaedic Surgery, New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, 06030
| | | | | | | | | | | |
Collapse
|
153
|
Xin X, Jiang X, Wang L, Stover ML, Zhan S, Huang J, Goldberg AJ, Liu Y, Kuhn L, Reichenberger EJ, Rowe DW, Lichtler AC. A Site-Specific Integrated Col2.3GFP Reporter Identifies Osteoblasts Within Mineralized Tissue Formed In Vivo by Human Embryonic Stem Cells. Stem Cells Transl Med 2014; 3:1125-37. [PMID: 25122686 DOI: 10.5966/sctm.2013-0128] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The use of human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) for study and treatment of bone diseases or traumatic bone injuries requires efficient protocols to differentiate hESCs/iPSCs into cells with osteogenic potential and the ability to isolate differentiated osteoblasts for analysis. We have used zinc finger nuclease technology to deliver a construct containing the Col2.3 promoter driving GFPemerald to the AAVS1 site (referred to as a "safe harbor" site), in human embryonic stem cells (H9Zn2.3GFP), with the goal of marking the cells that have become differentiated osteoblasts. In teratomas formed using these cells, we identified green fluorescent protein (GFP)-positive cells specifically associated with in vivo bone formation. We also differentiated the cells into a mesenchymal stem cell population with osteogenic potential and implanted them into a mouse calvarial defect model. We observed GFP-positive cells associated with alizarin complexone-labeled newly formed bone surfaces. The cells were alkaline phosphatase-positive, and immunohistochemistry with human specific bone sialoprotein (BSP) antibody indicates that the GFP-positive cells are also associated with the human BSP-containing matrix, demonstrating that the Col2.3GFP construct marks cells in the osteoblast lineage. Single-cell cloning generated a 100% Col2.3GFP-positive cell population, as demonstrated by fluorescence in situ hybridization using a GFP probe. The karyotype was normal, and pluripotency was demonstrated by Tra1-60 immunostaining, pluripotent low density reverse transcription-polymerase chain reaction array and embryoid body formation. These cells will be useful to develop optimal osteogenic differentiation protocols and to isolate osteoblasts from normal and diseased iPSCs for analysis.
Collapse
Affiliation(s)
- Xiaonan Xin
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Xi Jiang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Liping Wang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Mary Louise Stover
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Shuning Zhan
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jianping Huang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - A Jon Goldberg
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Yongxing Liu
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Liisa Kuhn
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Ernst J Reichenberger
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Alexander C Lichtler
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
154
|
James EN, Delany AM, Nair LS. Post-transcriptional regulation in osteoblasts using localized delivery of miR-29a inhibitor from nanofibers to enhance extracellular matrix deposition. Acta Biomater 2014; 10:3571-80. [PMID: 24816265 DOI: 10.1016/j.actbio.2014.04.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 03/31/2014] [Accepted: 04/24/2014] [Indexed: 02/03/2023]
Abstract
MicroRNAs are important post-transcriptional regulators of skeletal biology, and miRNA-based therapeutics have the potential to aid bone repair. However, efficient tools for delivering miRNA mimics or inhibitors to specific target tissues are limited. Polymeric nanofibers closely mimic natural extracellular matrix (ECM) morphology, and are attractive candidates for supporting delivery of cells and bone-anabolic reagents. It is hypothesized that gelatin nanofibers could be used for the localized transient delivery of miRNA-based therapeutics, using miR-29a inhibitor as a prototype to increase ECM deposition. miR-29 family members are negative regulators of ECM synthesis, targeting the mRNAs of selected collagens and osteonectin/SPARC. Inhibiting miR-29 activity may therefore increase ECM production by cells. miR-29a inhibitor-loaded gelatin nanofibers, prepared by electrospinning, demonstrated continuous release of miRNA inhibitor over 72h. Pre-osteoblastic murine MC3T3-E1 cell line seeded on miR-29a inhibitor-loaded nanofibers synthesized more osteonectin, indicating efficient inhibitor delivery. These cells also displayed increased Igf1 and Tgfb1 mRNA. Moreover, primary bone marrow stromal cells from transgenic pOBCol3.6cyan reporter mice, grown on miR-29a inhibitor scaffolds, displayed increased col3.6 cyan expression as well as collagen production. This study demonstrates that ECM mimicking nanostructured scaffolds, in conjunction with bioactive miRNA-based therapeutics, may serve as a novel platform for developing biologically active localized cell delivery systems.
Collapse
|
155
|
McManus A, Utreja A, Chen J, Kalajzic Z, Yang W, Nanda R, Wadhwa S, Uribe F. Evaluation of BSP expression and apoptosis in the periodontal ligament during orthodontic relapse: a preliminary study. Orthod Craniofac Res 2014; 17:239-48. [PMID: 24924469 DOI: 10.1111/ocr.12049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To examine the expression of bone sialoprotein (BSP) and apoptosis in an in vivo orthodontic relapse model. MATERIALS AND METHODS Male mice (10-12 weeks old), either transgenic [green fluorescent protein (GFP) driven by the BSP promoter] or wild type, were used in this study. To achieve orthodontic tooth movement (OTM), maxillary right first molars were moved mesially using closed-coil springs. Animals were divided into an OTM group (14 days continuous orthodontic force - 11 animals) or Relapse group (10 days of force application followed by 4 days of relapse - 8 animals). The control group was comprised of the contralateral maxillary molars. The periodontal ligament (PDL) was analyzed in areas of compression and tension for transgenic expression, osteoclast localization, and the presence of apoptotic cells. RESULTS There was a significant decrease in GFP-labeled cells on the compression and tension sides of the PDL in the OTM group compared with control. In the relapse group, GFP-labeled cells were significantly decreased only on the old compression side. Osteoclasts were localized on the compression side of the OTM group, whereas in the Relapse group, they were present on both sides. PDL apoptosis significantly increased on the compression side in OTM and Relapse groups. CONCLUSION Both OTM and Relapse groups exhibited a decreased number of GFP-labeled cells in areas of compression and tension. There was significant PDL apoptosis in regions under compressive forces following OTM and to a lesser extent following relapse.
Collapse
Affiliation(s)
- A McManus
- Private Practice, Shrewsbury, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
156
|
Caselli A, Olson TS, Otsuru S, Chen X, Hofmann TJ, Nah HD, Grisendi G, Paolucci P, Dominici M, Horwitz EM. IGF-1-mediated osteoblastic niche expansion enhances long-term hematopoietic stem cell engraftment after murine bone marrow transplantation. Stem Cells 2014; 31:2193-204. [PMID: 23818291 DOI: 10.1002/stem.1463] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 05/14/2013] [Accepted: 05/29/2013] [Indexed: 01/22/2023]
Abstract
The efficiency of hematopoietic stem cell (HSC) engraftment after bone marrow (BM) transplantation depends largely on the capacity of the marrow microenvironment to accept the transplanted cells. While radioablation of BM damages osteoblastic stem cell niches, little is known about their restoration and mechanisms governing their receptivity to engraft transplanted HSCs. We previously reported rapid restoration and profound expansion of the marrow endosteal microenvironment in response to marrow radioablation. Here, we show that this reorganization represents proliferation of mature endosteal osteoblasts which seem to arise from a small subset of high-proliferative, relatively radio-resistant endosteal cells. Multiple layers of osteoblasts form along the endosteal surface within 48 hours after total body irradiation, concomitant with a peak in marrow cytokine expression. This niche reorganization fosters homing of the transplanted hematopoietic cells to the host marrow space and engraftment of long-term-HSC. Inhibition of insulin-like growth factor (IGF)-1-receptor tyrosine kinase signaling abrogates endosteal osteoblast proliferation and donor HSC engraftment, suggesting that the cytokine IGF-1 is a crucial mediator of endosteal niche reorganization and consequently donor HSC engraftment. Further understanding of this novel mechanism of IGF-1-dependent osteoblastic niche expansion and HSC engraftment may yield clinical applications for improving engraftment efficiency after clinical HSC transplantation.
Collapse
Affiliation(s)
- Anna Caselli
- Department of Medical and Surgical Sciences of Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy; Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Villa MM, Wang L, Huang J, Rowe DW, Wei M. Bone tissue engineering with a collagen-hydroxyapatite scaffold and culture expanded bone marrow stromal cells. J Biomed Mater Res B Appl Biomater 2014; 103:243-53. [PMID: 24909953 DOI: 10.1002/jbm.b.33225] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 05/06/2014] [Accepted: 05/17/2014] [Indexed: 01/18/2023]
Abstract
Osteoprogenitor cells combined with supportive biomaterials represent a promising approach to advance the standard of care for bone grafting procedures. However, this approach faces challenges, including inconsistent bone formation, cell survival in the implant, and appropriate biomaterial degradation. We have developed a collagen-hydroxyapatite (HA) scaffold that supports consistent osteogenesis by donor-derived osteoprogenitors, and is more easily degraded than a pure ceramic scaffold. Herein, the material properties are characterized as well as cell attachment, viability, and progenitor distribution in vitro. Furthermore, we examined the biological performance in vivo in a critical-size mouse calvarial defect. To aid in the evaluation of the in-house collagen-HA scaffold, the in vivo performance was compared with a commercial collagen-HA scaffold (Healos(®) , Depuy). The in-house collagen-HA scaffold supported consistent bone formation by predominantly donor-derived osteoblasts, nearly completely filling a 3.5 mm calvarial defect with bone in all samples (n = 5) after 3 weeks of implantation. In terms of bone formation and donor cell retention at 3 weeks postimplantation, no statistical difference was found between the in-house and commercial scaffold following quantitative histomorphometry. The collagen-HA scaffold presented here is an open and well-defined platform that supports robust bone formation and should facilitate the further development of collagen-hydroxyapatite biomaterials for bone tissue engineering.
Collapse
Affiliation(s)
- Max M Villa
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, 06269-3136
| | | | | | | | | |
Collapse
|
158
|
Kemp JP, Medina-Gomez C, Estrada K, St Pourcain B, Heppe DHM, Warrington NM, Oei L, Ring SM, Kruithof CJ, Timpson NJ, Wolber LE, Reppe S, Gautvik K, Grundberg E, Ge B, van der Eerden B, van de Peppel J, Hibbs MA, Ackert-Bicknell CL, Choi K, Koller DL, Econs MJ, Williams FMK, Foroud T, Carola Zillikens M, Ohlsson C, Hofman A, Uitterlinden AG, Davey Smith G, Jaddoe VWV, Tobias JH, Rivadeneira F, Evans DM. Phenotypic dissection of bone mineral density reveals skeletal site specificity and facilitates the identification of novel loci in the genetic regulation of bone mass attainment. PLoS Genet 2014; 10:e1004423. [PMID: 24945404 PMCID: PMC4063697 DOI: 10.1371/journal.pgen.1004423] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/14/2014] [Indexed: 11/19/2022] Open
Abstract
Heritability of bone mineral density (BMD) varies across skeletal sites, reflecting different relative contributions of genetic and environmental influences. To quantify the degree to which common genetic variants tag and environmental factors influence BMD, at different sites, we estimated the genetic (rg) and residual (re) correlations between BMD measured at the upper limbs (UL-BMD), lower limbs (LL-BMD) and skull (SK-BMD), using total-body DXA scans of ∼ 4,890 participants recruited by the Avon Longitudinal Study of Parents and their Children (ALSPAC). Point estimates of rg indicated that appendicular sites have a greater proportion of shared genetic architecture (LL-/UL-BMD rg = 0.78) between them, than with the skull (UL-/SK-BMD rg = 0.58 and LL-/SK-BMD rg = 0.43). Likewise, the residual correlation between BMD at appendicular sites (r(e) = 0.55) was higher than the residual correlation between SK-BMD and BMD at appendicular sites (r(e) = 0.20-0.24). To explore the basis for the observed differences in rg and re, genome-wide association meta-analyses were performed (n ∼ 9,395), combining data from ALSPAC and the Generation R Study identifying 15 independent signals from 13 loci associated at genome-wide significant level across different skeletal regions. Results suggested that previously identified BMD-associated variants may exert site-specific effects (i.e. differ in the strength of their association and magnitude of effect across different skeletal sites). In particular, variants at CPED1 exerted a larger influence on SK-BMD and UL-BMD when compared to LL-BMD (P = 2.01 × 10(-37)), whilst variants at WNT16 influenced UL-BMD to a greater degree when compared to SK- and LL-BMD (P = 2.31 × 10(-14)). In addition, we report a novel association between RIN3 (previously associated with Paget's disease) and LL-BMD (rs754388: β = 0.13, SE = 0.02, P = 1.4 × 10(-10)). Our results suggest that BMD at different skeletal sites is under a mixture of shared and specific genetic and environmental influences. Allowing for these differences by performing genome-wide association at different skeletal sites may help uncover new genetic influences on BMD.
Collapse
Affiliation(s)
- John P. Kemp
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - Karol Estrada
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- School of Oral and Dental Sciences, University of Bristol, Bristol, United Kingdom
- School of Experimental Psychology, University of Bristol, Bristol, United Kingdom
| | - Denise H. M. Heppe
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole M. Warrington
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Ling Oei
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - Susan M. Ring
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
| | - Claudia J. Kruithof
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicholas J. Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
| | - Lisa E. Wolber
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Sjur Reppe
- Department of Medical Biochemistry, Oslo University Hospital, Ullevaal, Oslo, Norway
| | - Kaare Gautvik
- Department of Medical Biochemistry, Oslo University Hospital, Ullevaal, Oslo, Norway
- Department of Medical Biochemistry, Oslo Deacon Hospital, Oslo, Norway
| | - Elin Grundberg
- Department of Human Genetics, McGill University, Montréal, Canada
- McGill University and Genome Québec Innovation Centre, Montréal, Canada
| | - Bing Ge
- McGill University and Genome Québec Innovation Centre, Montréal, Canada
| | - Bram van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Matthew A. Hibbs
- Department of Computer Science, Trinity University, San Antonio, Texas, United States of America
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | - Kwangbom Choi
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Daniel L. Koller
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Michael J. Econs
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Albert Hofman
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - George Davey Smith
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
| | - Vincent W. V. Jaddoe
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Paediatrics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jonathan H. Tobias
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA), The Netherlands
| | - David M. Evans
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
159
|
Abstract
Bone and muscle mass are highly correlated. In part, this is a consequence of both tissues sharing common genetic determinants. In addition, both tissues are responsive to their mechanical environments. New genetic tools in mice will allow genes of interest to be inactivated in experimentally defined contexts, thus allowing investigators to distinguish direct effects on each tissue from physiological responses to a primary phenotype in the other.
Collapse
|
160
|
Gomillion CT, Lakhman RK, Kasi RM, Weiss RA, Kuhn LT, Goldberg AJ. Lithium-end-capped polylactide thin films influence osteoblast progenitor cell differentiation and mineralization. J Biomed Mater Res A 2014; 103:500-10. [PMID: 24733780 DOI: 10.1002/jbm.a.35190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 03/18/2014] [Accepted: 03/26/2014] [Indexed: 11/08/2022]
Abstract
End-capping by covalently binding functional groups to the ends of polymer chains offers potential advantages for tissue engineering scaffolds, but the ability of such polymers to influence cell behavior has not been studied. As a demonstration, polylactide (PLA) was end-capped with lithium carboxylate ionic groups (hPLA13kLi) and evaluated. Thin films of the hPLA13kLi and PLA homopolymer were prepared with and without surface texturing. Murine osteoblast progenitor cells from collagen 1α1 transgenic reporter mice were used to assess cell attachment, proliferation, differentiation, and mineralization. Measurement of green fluorescent protein expressed by these cells and xylenol orange staining for mineral allowed quantitative analysis. The hPLA13kLi was biologically active, increasing initial cell attachment and enhancing differentiation, while reducing proliferation and strongly suppressing mineralization, relative to PLA. These effects of bound lithium ions (Li(+) ) had not been previously reported, and were generally consistent with the literature on soluble additions of lithium. The surface texturing generated here did not influence cell behavior. These results demonstrate that end-capping could be a useful approach in scaffold design, where a wide range of biologically active groups could be employed, while likely retaining the desirable characteristics associated with the unaltered homopolymer backbone.
Collapse
Affiliation(s)
- Cheryl T Gomillion
- Department of Reconstructive Sciences, Center for Biomaterials, University of Connecticut Health Center, Farmington, Connecticut, 06030
| | | | | | | | | | | |
Collapse
|
161
|
Chan LLY, Huang J, Hagiwara Y, Aguila L, Rowe D. Discriminating multiplexed GFP reporters in primary articular chondrocyte cultures using image cytometry. J Fluoresc 2014; 24:1041-53. [PMID: 24728974 DOI: 10.1007/s10895-014-1383-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 03/20/2014] [Indexed: 11/25/2022]
Abstract
Flow cytometry has become a standard tool for defining a heterogeneous cell population based on surface expressed epitopes or GFP reporters that reflect cell types or cellular differentiation. The introduction of image cytometry raised the possibility of adaptation to discriminate GFP reporters used to appreciate cell heterogeneity within the skeletal lineages. The optical filters and LEDs were optimized for the reporters used in transgenic mice expressing various fluorescent proteins. In addition, the need for compensation between eGFP and surrounding reporters due to optical cross-talk was eliminated by selecting the appropriate excitation and emission filters. Bone marrow or articular cartilage cell cultures from GFP and RFP reporter mouse lines were established to demonstrate the equivalency in functionalities of image to flow cytometry analysis. To examine the ability for monitoring primary cell differentiation, articular chondrocyte cell cultures were established from mice that were single or doubly transgenic (Dkk3eGFP and Col2A1GFPcyan), which identify the progression of superficial small articular cell to a mature chondrocyte. The instrument was able to rapidly and accurately discriminate cells that were Dkk3eGFP only, Dkk3eGFP/Col2A1GFPcyan, and Col2A1GFP, which provides a useful tool for studying the impact of culture conditions on lineage expansion and differentiation.
Collapse
Affiliation(s)
- Leo Li-Ying Chan
- Department of Technology R&D, Nexcelom Bioscience LLC, 360 Merrimack St. Building 9, Lawrence, MA, 01843, USA,
| | | | | | | | | |
Collapse
|
162
|
Gohil SV, Adams DJ, Maye P, Rowe DW, Nair LS. Evaluation of rhBMP-2 and bone marrow derived stromal cell mediated bone regeneration using transgenic fluorescent protein reporter mice. J Biomed Mater Res A 2014; 102:4568-80. [PMID: 24677665 DOI: 10.1002/jbm.a.35122] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/17/2014] [Accepted: 02/10/2014] [Indexed: 01/08/2023]
Abstract
The aim of the study is use of transgenic fluorescent protein reporter mouse models to understand the cellular processes in recombinant human bone morphogenetic protein-2 (rhBMP-2) mediated bone formation. Bilateral parietal calvarial bone defects in Col3.6Topaz transgenic fluorescent osteoblast reporter mouse were used to understand the bone formation in the presence and absence of rhBMP2 and/or Col3.6Cyan bone marrow derived stromal cells (BMSCs), using collagen-hydroxyapatite matrix (Healos) as a biomaterial. The bone regeneration was not confined to the site of BMP-2 implantation and significant bone formation was observed in the neighboring defect site. Osteogenic cellular activity with overlying alizarin complexone staining was observed in both the defects indicating host cell induced mineralization. However, implantation of BMSCs along with rhBMP-2 demonstrated a donor cell derived bone formation. The presence of rhBMP-2 did not support host cell recruitment in the presence of donor cells. This study demonstrates the potential of multiple fluorescent reporters to understand the cellular processes involved in the bone regeneration process using biomaterials, growth factors, and/or stem cells.
Collapse
Affiliation(s)
- Shalini V Gohil
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, 06032; Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, Connecticut, 06032
| | | | | | | | | |
Collapse
|
163
|
Yu X, Wang L, Xia Z, Chen L, Jiang X, Rowe D, Wei M. Modulation of Host Osseointegration during Bone Regeneration by Controlling Exogenous Stem Cells Differentiation Using a Material Approach. Biomater Sci 2014; 2:242-251. [PMID: 24999385 PMCID: PMC4078879 DOI: 10.1039/c3bm60173k] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cell-based tissue engineering for large bone defect healing has attracted enormous attention in regenerative medicine. However, sufficient osseointegration of the grafts combined with exogenous stem cells still remains a major challenge. Here we developed a material approach to modulate the integration of the grafts to the host tissue when exogenous bone marrow stromal cells (BMSCs) were used as donor cells. Distinctive osseointegration of bone grafts was observed as we varied the content of hydroxyapatite (HA) in the tissue scaffolds implanted in a mouse femur model. More than 80% of new bone was formed in the first two weeks of implantation in high HA content scaffold but lack of host integration while only less than 5% of the new bone was formed during this time period in the no HA group but with much stronger host integration. Cell origin analysis leveraging GFP reporter indicates new bone in HA containing groups was mainly derived from donor BMSCs. In comparison, both host and donor cells were found on new bone surface in the no HA groups which led to seamless bridging between host tissue and the scaffold. Most importantly, host integration during bone formation is closely dictated to the content of HA present in the scaffolds. Taken together, we demonstrate a material approach to modulate the osseointegration of bone grafts in the context of exogenous stem cell-based bone healing strategy which might lead to fully functional bone tissue regeneration.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - Liping Wang
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - Zengmin Xia
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - Li Chen
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - Xi Jiang
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - David Rowe
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| | - Mei Wei
- Department of Materials Science and Engineering, Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
164
|
Dela Cruz A, Mattocks M, Sugamori KS, Grynpas MD, Mitchell J. Reduced trabecular bone mass and strength in mice overexpressing Gα11 protein in cells of the osteoblast lineage. Bone 2014; 59:211-22. [PMID: 24308950 DOI: 10.1016/j.bone.2013.11.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 01/10/2023]
Abstract
G protein-coupled receptors (GPCRs) require G proteins for intracellular signaling to regulate a variety of growth and maintenance processes, including osteogenesis and bone turnover. Bone maintenance events may be altered by changes in the activity or level of G proteins, which then modify signaling in bone cells such as osteoblasts. We have previously reported increased levels of Gα11 protein and signaling to phospholipase C/protein kinase C pathways in response to dexamethasone in osteoblastic UMR 106-01 cells. Here we generated pOBCol3.6-GNA11 transgenic mice that overexpress Gα11 protein in cells of the osteoblast lineage (G11-Tg mice). G11-Tg mice exhibit an osteopenic phenotype characterized by significant reductions in trabecular bone mineral density, thickness, number and strength. The numbers of osteoblasts and osteocytes were unchanged in G11-Tg bone, but early markers of osteoblast differentiation, Alp and Bsp, were increased while the late stage differentiation marker Ocn was not changed suggesting reduced osteoblast maturation in G11-Tg trabecular bone which was accompanied by a decreased bone formation rate. Furthermore, in vitro cultures of G11-Tg primary osteoblasts show delayed osteoblast differentiation and mineralization. Histological analyses also revealed increased osteoclast parameters, accompanied by elevated mRNA expression of Trap and Ctsk. mRNA levels of Rankl and M-csf were elevated in vitro in bone marrow stromal cells undergoing osteogenesis and in trabecular bone in vivo. Together, these findings demonstrate that increasing Gα11 protein expression in osteoblasts can alter gene expression and result in a dual mechanism of trabecular bone loss.
Collapse
Affiliation(s)
- Ariana Dela Cruz
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Michael Mattocks
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Kim S Sugamori
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Marc D Grynpas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Jane Mitchell
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
165
|
Local transplantation is an effective method for cell delivery in the osteogenesis imperfecta murine model. INTERNATIONAL ORTHOPAEDICS 2014; 38:1955-62. [PMID: 24384938 DOI: 10.1007/s00264-013-2249-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 12/03/2013] [Indexed: 12/11/2022]
Abstract
PURPOSE Osteogenesis imperfecta is a serious genetic disorder that results from improper type I collagen production. We aimed to evaluate whether bone marrow stromal cells (BMSC) delivered locally into femurs were able to engraft, differentiate into osteoblasts, and contribute to formation of normal bone matrix in the osteogenesis imperfect murine (oim) model. METHODS Donor BMSCs from bone-specific reporter mice (Col2.3GFP) were expanded in vitro and transplanted into the femoral intramedullary cavity of oim mice. Engraftment was evaluated after four weeks. RESULTS We detected differentiation of donor BMSCs into Col2.3GFP+ osteoblasts and osteocytes in cortical and trabecular bone of transplanted oim femurs. New bone formation was detected by deposition of dynamic label in the proximity to the Col2.3GFP+ osteoblasts, and new bone showed more organized collagen structure and expression of type I α2 collagen. Col2.3GFP cells were not found in the contralateral femur indicating that transplanted osteogenic cells did not disseminate by circulation. No osteogenic engraftment was observed following intravenous transplantation of BMSCs. BMSC cultures derived from transplanted femurs showed numerous Col2.3GFP+ colonies, indicating the presence of donor progenitor cells. Secondary transplantation of cells recovered from recipient femurs and expanded in vitro also showed Col2.3GFP+ osteoblasts and osteocytes confirming the persistence of donor stem/progenitor cells. CONCLUSION We show that BMSCs delivered locally in oim femurs are able to engraft, differentiate into osteoblasts and osteocytes and maintain their progenitor potential in vivo. This suggests that local delivery is a promising approach for introduction of autologous MSC in which mutations have been corrected.
Collapse
|
166
|
Ouyang Z, Chen Z, Ishikawa M, Yue X, Kawanami A, Leahy P, Greenfield EM, Murakami S. Prx1 and 3.2kb Col1a1 promoters target distinct bone cell populations in transgenic mice. Bone 2014; 58:136-145. [PMID: 24513582 PMCID: PMC4000289 DOI: 10.1016/j.bone.2013.10.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 10/11/2013] [Accepted: 10/19/2013] [Indexed: 02/07/2023]
Abstract
Bones consist of a number of cell types including osteoblasts and their precursor cells at various stages of differentiation. To analyze cellular organization within the bone, we generated Col1a1CreER-DsRed transgenic mice that express, in osteoblasts, CreER and DsRed under the control of a mouse 3.2kb Col1a1 promoter. We further crossed Col1a1CreER-DsRed mice with Prx1CreER-GFP mice that express CreER and GFP in osteochondro progenitor cells under the control of a 2.4kb Prx1 promoter. Since the 3.2kb Col1a1 promoter becomes active in osteoblasts at early stages of differentiation, and Prx1CreER-GFP-expressing periosteal cells show endogenous Col1a1 expression, we expected to find a cell population in which both the 2.4kb Prx1 promoter and the 3.2kb Col1a1 promoter are active. However, our histological and flow cytometric analyses demonstrated that these transgenes are expressed in distinct cell populations. In the periosteum of long bones, Col1a1CreER-DsRed is expressed in the innermost layer directly lining the bone surface, while Prx1CreER-GFP-expressing cells are localized immediately outside of the Col1a1CreER-DsRed-expressing osteoblasts. In the calvaria, Prx1CreER-GFP-expressing cells are also localized in the cranial suture mesenchyme. Our experiments further showed that Col1a1CreER-DsRed-expressing cells lack chondrogenic potential, while the Prx1CreER-GFP-expressing cells show both chondrogenic and osteogenic potential. Our results indicate that Col1a1CreER-DsRed-expressing cells are committed osteoblasts, while Prx1CreER-GFP-expressing cells are osteochondro progenitor cells. The Prx1CreER-GFP and Col1a1CreER-DsRed transgenes will offer novel approaches for analyzing lineage commitment and early stages of osteoblast differentiation under physiologic and pathologic conditions.
Collapse
Affiliation(s)
- Zhufeng Ouyang
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zhijun Chen
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Masakazu Ishikawa
- Department of Medicine, Cardiovascular Institute, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Xiuzhen Yue
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Aya Kawanami
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Patrick Leahy
- Division of General Medical Sciences, Oncology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Edward M Greenfield
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Division of General Medical Sciences, National Center for Regenerative Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Shunichi Murakami
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Division of General Medical Sciences, National Center for Regenerative Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Department of Genetics and Genomic Sciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| |
Collapse
|
167
|
Abstract
During the last decade a considerable amount of data have been accumulated regarding the role of intracellular signaling pathways in the pathogenesis of human diseases. One of these, Notch signaling, well known for its significance in cellular development and tissue morphogenesis, has been increasingly recognized as a crucial participant in the pathogenetic mechanisms underlying certain skeletal disorders. A better understanding of the biology and regulation of this multifaceted pathway is considered an important step towards clarification of the pathogenesis of various skeletal diseases and the development of novel targets for therapeutic purposes.
Collapse
Affiliation(s)
- Maria P Yavropoulou
- Division of Endocrinology and Metabolism, Laboratory of Molecular Endocrinology, 1st Department of Medicine, ΑHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - John G Yovos
- Division of Endocrinology and Metabolism, Laboratory of Molecular Endocrinology, 1st Department of Medicine, ΑHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
168
|
Matthews BG, Grcevic D, Wang L, Hagiwara Y, Roguljic H, Joshi P, Shin DG, Adams DJ, Kalajzic I. Analysis of αSMA-labeled progenitor cell commitment identifies notch signaling as an important pathway in fracture healing. J Bone Miner Res 2014; 29:1283-94. [PMID: 24190076 PMCID: PMC4864015 DOI: 10.1002/jbmr.2140] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 10/14/2013] [Accepted: 10/27/2013] [Indexed: 11/11/2022]
Abstract
Fracture healing is a regenerative process that involves coordinated responses of many cell types, but characterization of the roles of specific cell populations in this process has been limited. We have identified alpha smooth muscle actin (αSMA) as a marker of a population of mesenchymal progenitor cells in the periosteum that contributes to osteochondral elements during fracture healing. Using a lineage tracing approach, we labeled αSMA-expressing cells, and characterized changes in the periosteal population during the early stages of fracture healing by histology, flow cytometry, and gene expression profiling. In response to fracture, the αSMA-labeled population expanded and began to differentiate toward the osteogenic and chondrogenic lineages. The frequency of mesenchymal progenitor cell markers such as Sca1 and PDGFRα increased after fracture. By 6 days after fracture, genes involved in matrix production and remodeling were elevated. In contrast, genes associated with muscle contraction and Notch signaling were downregulated after fracture. We confirmed that activating Notch signaling in αSMA-labeled cells inhibited differentiation into osteogenic and adipogenic lineages in vitro and ectopic bone formation in vivo. By characterizing changes in a selected αSMA-labeled progenitor cell population during fracture callus formation, we have shown that modulation of Notch signaling may determine osteogenic potential of αSMA-expressing progenitor cells during bone healing.
Collapse
Affiliation(s)
- Brya G Matthews
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Inactivation of the androgen receptor in bone-forming cells leads to trabecular bone loss in adult female mice. BONEKEY REPORTS 2013; 2:440. [PMID: 24422138 DOI: 10.1038/bonekey.2013.174] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 09/09/2013] [Indexed: 12/28/2022]
Abstract
Removal of the androgen receptor (AR) from bone-forming cells has been shown to reduce trabecular bone volume in male mice. In female mice, the role of AR in the regulation of bone homeostasis has been poorly understood. We generated a mouse strain in which the AR is completely inactivated only in mineralizing osteoblasts and osteocytes by breeding mice carrying osteocalcin promoter-regulated Cre-recombinase with mice possessing loxP recombination sites flanking exon 2 of the AR gene (AR(ΔOB/ΔOB) mice). In female AR(ΔOB/ΔOB) mice, the trabecular bone volume was reduced owing to a smaller number of trabeculae at 6 months of age compared with the control AR(fl/fl) animals. In male AR(ΔOB/ΔOB) mice, an increase in trabecular bone separation could already be detected at 3.5 months of age, and at 6 months, the trabecular bone volume was significantly reduced compared with that of male AR(fl/fl) mice. No AR-dependent changes were observed in the cortical bone of either sex. On the basis of micro-computed tomography and histomorphometry, we conclude that in male mice, the AR is involved in the regulation of osteoclast number by osteoblasts, whereas in female mice, the lack of the AR in the bone-forming cells leads to a decreased number of trabeculae upon aging.
Collapse
|
170
|
Abstract
Few investigators think of bone as an endocrine gland, even after the discovery that osteocytes produce circulating fibroblast growth factor 23 that targets the kidney and potentially other organs. In fact, until the last few years, osteocytes were perceived by many as passive, metabolically inactive cells. However, exciting recent discoveries have shown that osteocytes encased within mineralized bone matrix are actually multifunctional cells with many key regulatory roles in bone and mineral homeostasis. In addition to serving as endocrine cells and regulators of phosphate homeostasis, these cells control bone remodeling through regulation of both osteoclasts and osteoblasts, are mechanosensory cells that coordinate adaptive responses of the skeleton to mechanical loading, and also serve as a manager of the bone's reservoir of calcium. Osteocytes must survive for decades within the bone matrix, making them one of the longest lived cells in the body. Viability and survival are therefore extremely important to ensure optimal function of the osteocyte network. As we continue to search for new therapeutics, in addition to the osteoclast and the osteoblast, the osteocyte should be considered in new strategies to prevent and treat bone disease.
Collapse
Affiliation(s)
- Sarah L Dallas
- PhD, Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 650 East 25th Street, Kansas City, Missouri 64108.
| | | | | |
Collapse
|
171
|
Preosteocytes/osteocytes have the potential to dedifferentiate becoming a source of osteoblasts. PLoS One 2013; 8:e75204. [PMID: 24040401 PMCID: PMC3765403 DOI: 10.1371/journal.pone.0075204] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/11/2013] [Indexed: 01/23/2023] Open
Abstract
Presently there is no clear evidence for the ability of mature osteogenic lineage cells to dedifferentiate. In order to identify and trace mature osteogenic lineage cells, we have utilized transgenic mouse models in which the dentin matrix protein 1 (Dmp1) promoter drives expression of GFP (active marker) or Cre recombinase (historic label) in preosteocytes/osteocytes. In long bone chip outgrowth cultures, in which cells on the bone surface were enzymatically removed, cells with previous activity of the Dmp1 promoter migrated onto plastic and down-regulated Dmp1-GFP expression. Dmp1Cre-labeled cells from these cultures had the potential to re-differentiate into the osteogenic lineage, while the negative population showed evidence of adipogenesis. We observed numerous Dmp1Cre-labeled osteoblasts on the surface of bone chips following their in vivo transplantation. Our data indicate that cells embedded in bone matrix are motile, and once given access to the extra bony milieu will migrate out of their lacunae. This population of cells is phenotypically and functionally heterogeneous in vitro. Once the preosteocytes/osteocytes leave lacunae, they can dedifferentiate, potentially providing an additional source of functional osteoblasts.
Collapse
|
172
|
Roguljic H, Matthews BG, Yang W, Cvija H, Mina M, Kalajzic I. In vivo identification of periodontal progenitor cells. J Dent Res 2013; 92:709-15. [PMID: 23735585 PMCID: PMC3711570 DOI: 10.1177/0022034513493434] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The periodontal ligament contains progenitor cells; however, their identity and differentiation potential in vivo remain poorly characterized. Previous results have suggested that periodontal tissue progenitors reside in perivascular areas. Therefore, we utilized a lineage-tracing approach to identify and track periodontal progenitor cells from the perivascular region in vivo. We used an alpha-smooth muscle actin (αSMA) promoter-driven and tamoxifen-inducible Cre system (αSMACreERT2) that, in combination with a reporter mouse line (Ai9), permanently labels a cell population, termed 'SMA9'. To trace the differentiation of SMA9-labeled cells into osteoblasts/cementoblasts, we utilized a Col2.3GFP transgene, while expression of Scleraxis-GFP was used to follow differentiation into periodontal ligament fibroblasts during normal tissue formation and remodeling following injury. In uninjured three-week-old SMA9 mice, tamoxifen labeled a small population of cells in the periodontal ligament that expanded over time, particularly in the apical region of the root. By 17 days and 7 weeks after labeling, some SMA9-labeled cells expressed markers indicating differentiation into mature lineages, including cementocytes. Following injury, SMA9 cells expanded, and differentiated into cementoblasts, osteoblasts, and periodontal ligament fibroblasts. SMA9-labeled cells represent a source of progenitors that can give rise to mature osteoblasts, cementoblasts, and fibroblasts within the periodontium.
Collapse
Affiliation(s)
- H Roguljic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | | | | | | | | | | |
Collapse
|
173
|
Cheng SL, Shao JS, Behrmann A, Krchma K, Towler DA. Dkk1 and MSX2-Wnt7b signaling reciprocally regulate the endothelial-mesenchymal transition in aortic endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33:1679-89. [PMID: 23685555 PMCID: PMC3837473 DOI: 10.1161/atvbaha.113.300647] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 04/25/2013] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Endothelial cells (ECs) can undergo an endothelial-mesenchymal transition with tissue fibrosis. Wnt- and Msx2-regulated signals participate in arteriosclerotic fibrosis and calcification. We studied the impact of Wnt7, Msx2, and Dkk1, a Wnt7 antagonist, on endothelial-mesenchymal transition in primary aortic ECs. APPROACH AND RESULTS Transduction of aortic ECs with vectors expressing Dkk1 suppressed EC differentiation and induced a mineralizing myofibroblast phenotype. Dkk1 suppressed claudin 5, PECAM, cadherin 5 (Cdh5), Tie1, and Tie2. Dkk1 converted the cuboidal cell monolayer into a spindle-shaped multilayer and inhibited EC cord formation. Myofibroblast and osteogenic markers, SM22, type I collagen, Osx, Runx2, and alkaline phosphatase, were upregulated by Dkk1 via activin-like kinase/Smad pathways. Dkk1 increased fibrotic mineralization of aortic ECs cultured under osteogenic conditions--the opposite of mesenchymal cell responses. Msx2 and Wnt7b maintained morphology and upregulated markers of differentiated ECs. Deleting EC Wnt7b with the Cdh5-Cre transgene in Wnt7b(fl/fl);LDLR(-/-) mice upregulated aortic osteogenic genes (Osx, Sox9, Runx2, and Msx2) and nuclear phospho-Smad1/5, and increased collagen and calcium accumulation. CONCLUSIONS Dkk1 enhances endothelial-mesenchymal transition in aortic ECs, whereas Wnt7b and Msx2 signals preserve EC phenotype. EC responses to Dkk1, Wnt7b, and Msx2 are the opposite of mesenchymal responses, coupling EC phenotypic stability with osteofibrogenic predilection during arteriosclerosis.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Arteriosclerosis/genetics
- Arteriosclerosis/metabolism
- Arteriosclerosis/pathology
- Biomarkers/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cattle
- Cell Differentiation
- Cell Shape
- Cells, Cultured
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Epithelial-Mesenchymal Transition
- Fibrosis
- Gene Expression Regulation
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Male
- Mice
- Mice, Knockout
- Myofibroblasts/metabolism
- Myofibroblasts/pathology
- Neovascularization, Physiologic
- Ossification, Heterotopic/metabolism
- Phenotype
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Transduction, Genetic
- Transfection
- Wnt Proteins/deficiency
- Wnt Proteins/genetics
- Wnt Proteins/metabolism
- Wnt Signaling Pathway
Collapse
Affiliation(s)
- Su-Li Cheng
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Jian-Su Shao
- Department of Internal Medicine, Washington University, St. Louis, Missouri 63110
| | - Abraham Behrmann
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Karen Krchma
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Dwight A. Towler
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| |
Collapse
|
174
|
Murao H, Yamamoto K, Matsuda S, Akiyama H. Periosteal cells are a major source of soft callus in bone fracture. J Bone Miner Metab 2013; 31:390-8. [PMID: 23475152 DOI: 10.1007/s00774-013-0429-x] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 01/24/2013] [Indexed: 12/26/2022]
Abstract
During the healing process after bone fracture, soft callus forms adjacent to the fracture site, is replaced by hard callus, and is finally remodeled to the original bone configuration. Although the cambium layer of the periosteum is reported to play an essential role in callus formation, we still lack direct in vivo evidence of this. To investigate the cell lineage of the soft callus, we analyzed the process of fracture healing in Prx1-Cre;ROSA26 reporter (R26R), Col1a1(3.6 kb)-Cre;R26R, Col1a1(2.3 kb)-Cre;R26R, Sox9-CreERT2;R26R, and Sox9-LacZ mice with X-gal staining. In the Prx1-Cre;R26R, in which the cells of the periosteum stained for X-gal before fracture, all cells in the soft callus were X-gal positive, whereas in the Col1a1(3.6 kb)-Cre;R26R mice, the cells in the periosteum before fracture stained for X-gal and the soft callus was partly composed of X-gal-positive cells. In contrast, in the Col1a1(2.3 kb)-Cre;R26R mice, in which the mature osteoblasts in the cambium layer of the periosteum were marked before fracture, no cells in the soft callus at the fracture site were X-gal positive. These results suggest that most of the cells in the soft callus are derived from the mesenchymal progenitors in the periosteum, and not from mature osteoblastic cells. Interestingly, in the Sox9-LacZ mice, Sox9-expressing X-gal-positive cells emerged in the periosteum adjacent to the fracture site 3 days after fracture. We demonstrated this by injecting tamoxifen into the Sox9-CreERT2;R26R mice for 3 days after fracture, so that these Sox9-expressing periosteal cells gave rise to cells in the soft and hard calli. Our findings show that the periosteal cells in which Sox9 expression is induced just after fracture are the major source of the chondrocytes and osteoblasts in the fracture callus.
Collapse
Affiliation(s)
- Hiroki Murao
- Department of Orthopaedics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo, Kyoto, Japan
| | | | | | | |
Collapse
|
175
|
Zhou H, Cooper MS, Seibel MJ. Endogenous Glucocorticoids and Bone. Bone Res 2013; 1:107-19. [PMID: 26273496 DOI: 10.4248/br201302001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/20/2013] [Indexed: 01/28/2023] Open
Abstract
While the adverse effects of glucocorticoids on bone are well described, positive effects of glucocorticoids on the differentiation of osteoblasts are also observed. These paradoxical effects of glucocorticoids are dose dependent. At both physiologicaland supraphysiological levels of glucocorticoids, osteoblasts and osteocytes are the major glucocorticoid target cells. However, the response of the osteoblasts to each of these is quite distinct. At physiology levels, glucocorticoids direct mesenchymal progenitor cells to differentiate towards osteoblasts and thus increase bone formation in a positive way. In contrast with ageing, the excess production of glucocorticoids, at both systemic and intracellular levels, appear to impact on osteoblast and osteocytes in a negative way in a similar fashion to that seen with therapeutic glucocorticoids. This review will focus on therole of glucocorticoids in normal bone physiology, with particular emphasis on the mechanism by which endogenous glucocorticoids impact on bone and its constituent cells.
Collapse
Affiliation(s)
- Hong Zhou
- Bone Research Program, ANZAC Research Institute , Sydney, Australia ; Concord Clinical School, The University of Sydney , Sydney, Australia
| | - Mark S Cooper
- Concord Clinical School, The University of Sydney , Sydney, Australia ; Department of Endocrinology & Metabolism, Concord Hospital , Sydney, Australia
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute , Sydney, Australia ; Concord Clinical School, The University of Sydney , Sydney, Australia ; Department of Endocrinology & Metabolism, Concord Hospital , Sydney, Australia
| |
Collapse
|
176
|
Zanotti S, Canalis E. Hairy and Enhancer of Split-related with YRPW motif (HEY)2 regulates bone remodeling in mice. J Biol Chem 2013; 288:21547-57. [PMID: 23782701 DOI: 10.1074/jbc.m113.489435] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Notch induces Hairy and Enhancer of Split-related with YRPW motif (Hey)1, Hey2, and HeyL expression in osteoblasts, but the contributions of these genes to the skeletal effects of Notch are not fully understood. HEY1 misexpression has limited skeletal impact, female HeyL null mice display increased bone mass, and Hey2 inactivation is developmentally lethal. To inactivate Hey2 in immature or mature osteoblasts, Hey2(loxP/loxP) mice were crossed with transgenics expressing CRE under the control of the osterix (Osx-Cre) or osteocalcin (Oc-Cre) promoters to generate Osx-Cre(+/-);Hey2(Δ/Δ) or Oc-Cre(+/-);Hey2(Δ/Δ) mice. Trabecular bone volume increased in 3-month-old Osx-Cre(+/-);Hey2(Δ/Δ) and Oc-Cre(+/-);Hey2(Δ/Δ) male mice and in 1-month-old Oc-Cre(+/-);Hey2(Δ/Δ) female mice, although 3-month-old Oc-Cre(+/-);Hey2(Δ/Δ) females developed osteopenia. Alkaline phosphatase liver/bone/kidney (ALPL) expression and activity were suppressed in osteoblasts from Oc-Cre(+/-);Hey2(Δ/Δ) mice of both sexes. To overexpress HEY2 in osteoblasts, transgenic mice where a 3.6-kb fragment of the rat collagen type-I α1 promoter directs HEY2 expression were created. Three-month-old Hey2 transgenic males exhibited decreased osteoblast activity and increased bone resorption and developed osteopenia at 6 months of age. Hey2 transgenic females exhibited reduced osteoblast number and function, but no changes in bone resorption. HEY2 overexpression in osteoblasts from mice of both sexes inhibited ALPL expression and activity and suppressed osteocalcin transcripts in cells from male mice only. HEY2 overexpression in osteoblasts from male mice enhanced bone resorption by co-cultured splenocytes and induced interleukin-6, a molecule that promotes osteoclastogenesis. In conclusion, HEY2 decreases skeletal mass and regulates bone remodeling in male mice.
Collapse
Affiliation(s)
- Stefano Zanotti
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut 06105-1299, USA
| | | |
Collapse
|
177
|
Villa MM, Wang L, Huang J, Rowe DW, Wei M. Visualizing osteogenesis in vivo within a cell-scaffold construct for bone tissue engineering using two-photon microscopy. Tissue Eng Part C Methods 2013; 19:839-49. [PMID: 23641794 DOI: 10.1089/ten.tec.2012.0490] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Tissue-engineering therapies have shown early success in the clinic, however, the cell-biomaterial interactions that result in successful outcomes are not yet well understood and are difficult to observe. Here we describe a method for visualizing bone formation within a tissue-engineered construct in vivo, at a single-cell resolution, and in situ in three dimensions using two-photon microscopy. First, two-photon microscopy and histological perspectives were spatially linked using fluorescent reporters for cells in the skeletal lineage. In the process, the tissue microenvironment that precedes a repair-focused study was described. The distribution and organization of type I collagen in the calvarial microenvironment was also described using its second harmonic signal. Second, this platform was used to observe in vivo, for the first time, host cells, donor cells, scaffold, and new bone formation within cell-seeded constructs in a bone defect. We examined constructs during bone repair 4 and 6 weeks after implantation. New bone formed on scaffolds, primarily by donor cells. Host cells formed a new periosteal layer that covered the implant. Scaffold resorption appeared to be site specific, where areas near the top were removed and deeper areas were completely embedded in new mineral. Visualizing the in vivo progression of the cell and scaffold microenvironment will contribute to our understanding of tissue-engineered regeneration and should lead to the development of more streamlined and therapeutically powerful approaches.
Collapse
Affiliation(s)
- Max M Villa
- 1 Department of Materials Science and Engineering, University of Connecticut , Storrs, Connecticut
| | | | | | | | | |
Collapse
|
178
|
Wattanachanya L, Lu WD, Kundu RK, Wang L, Abbott MJ, O'Carroll D, Quertermous T, Nissenson RA. Increased bone mass in mice lacking the adipokine apelin. Endocrinology 2013; 154:2069-80. [PMID: 23584856 PMCID: PMC3740482 DOI: 10.1210/en.2012-2034] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adipose tissue plays an important role in skeletal homeostasis, and there is interest in identifying adipokines that influence bone mass. One such adipokine may be apelin, a ligand for the Gi-G protein-coupled receptor APJ, which has been reported to enhance mitogenesis and suppress apoptosis in MC3T3-E1 cells and primary human osteoblasts (OBs). However, it is unclear whether apelin plays a physiological role in regulating skeletal homeostasis in vivo. In this study, we compared the skeletal phenotypes of apelin knockout (APKO) and wild-type mice and investigated the direct effects of apelin on bone cells in vitro. The increased fractional cancellous bone volume at the distal femur was observed in APKO mice of both genders at 12 weeks of age and persisted until the age of 20. Cortical bone perimeter at the femoral midshaft was significantly increased in males and females at both time points. Dynamic histomorphometry revealed that APKO mice had increased rates of bone formation and mineral apposition, with evidences of accelerated OB proliferation and differentiation, without significant alteration in osteoclast activity. An in vitro study showed that apelin increased proliferation of primary mouse OBs as well as suppressed apoptosis in a dose-dependent manner with the maximum effect at 5nM. However, it had no effect on the formation of mineralized nodules. We did not observed significantly altered in osteoclast parameters in vitro. Taken together, the increased bone mass in mice lacking apelin suggested complex direct and paracrine/endocrine effects of apelin on bone, possibly via modulating insulin sensitivity. These results indicate that apelin functions as a physiologically significant antianabolic factor in bone in vivo.
Collapse
Affiliation(s)
- Lalita Wattanachanya
- Veterans Affairs Medical Center (111 N), 4150 Clement Street, San Francisco, California 94121, USA
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Kalajzic I, Matthews BG, Torreggiani E, Harris MA, Divieti Pajevic P, Harris SE. In vitro and in vivo approaches to study osteocyte biology. Bone 2013; 54:296-306. [PMID: 23072918 PMCID: PMC3566324 DOI: 10.1016/j.bone.2012.09.040] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/27/2012] [Accepted: 09/27/2012] [Indexed: 11/26/2022]
Abstract
Osteocytes, the most abundant cell population of the bone lineage, have been a major focus in the bone research field in recent years. This population of cells that resides within mineralized matrix is now thought to be the mechanosensory cell in bone and plays major roles in the regulation of bone formation and resorption. Studies of osteocytes had been impaired by their location, resulting in numerous attempts to isolate primary osteocytes and to generate cell lines representative of the osteocytic phenotype. Progress has been achieved in recent years by utilizing in vivo genetic technology and generation of osteocyte directed transgenic and gene deficiency mouse models. We will provide an overview of the current in vitro and in vivo models utilized to study osteocyte biology. We discuss generation of osteocyte-like cell lines and isolation of primary osteocytes and summarize studies that have utilized these cellular models to understand the functional role of osteocytes. Approaches that attempt to selectively identify and isolate osteocytes using fluorescent protein reporters driven by regulatory elements of genes that are highly expressed in osteocytes will be discussed. In addition, recent in vivo studies utilizing overexpression or conditional deletion of various genes using dentin matrix protein (Dmp1) directed Cre recombinase are outlined. In conclusion, evaluation of the benefits and deficiencies of currently used cell lines/genetic models in understanding osteocyte biology underlines the current progress in this field. The future efforts will be directed towards developing novel in vitro and in vivo models that would additionally facilitate in understanding the multiple roles of osteocytes.
Collapse
Affiliation(s)
- Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut 06032, USA.
| | | | | | | | | | | |
Collapse
|
180
|
Matic I, Matthews BG, Kizivat T, Igwe JC, Marijanovic I, Ruohonen ST, Savontaus E, Adams DJ, Kalajzic I. Bone-specific overexpression of NPY modulates osteogenesis. KLIN NEUROPHYSIOL 2013. [PMID: 23196263 DOI: 10.1055/s-0032-1305278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Neuropeptide Y (NPY) is a peptide involved in the regulation of appetite and energy homeostasis. Genetic data indicates that NPY decreases bone formation via central and peripheral activities. NPY is produced by various cell types including osteocytes and osteoblasts and there is evidence suggesting that peripheral NPY is important for regulation of bone formation. We sought to investigate the role of bone-derived NPY in bone metabolism. METHODS We generated a mouse where NPY was over-expressed specifically in mature osteoblasts and osteocytes (Col2.3NPY) and characterized the bone phenotype of these mice in vivo and in vitro. RESULTS Trabecular and cortical bone volume was reduced in 3-month-old animals, however bone formation rate and osteoclast activity were not significantly changed. Calvarial osteoblast cultures from Col2.3NPY mice also showed reduced mineralization and expression of osteogenic marker genes. CONCLUSIONS Our data suggest that osteoblast/osteocyte-derived NPY is capable of altering osteogenesis in vivo and in vitro and may represent an important source of NPY for regulation of bone formation. However, it is possible that other peripheral sources of NPY such as the sympathetic nervous system and vasculature also contribute to peripheral regulation of bone turnover.
Collapse
Affiliation(s)
- I Matic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Gencheva M, Hare I, Kurian S, Fortney J, Piktel D, Wysolmerski R, Gibson LF. Bone marrow osteoblast vulnerability to chemotherapy. Eur J Haematol 2013; 90:469-78. [PMID: 23551534 DOI: 10.1111/ejh.12109] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2013] [Indexed: 12/21/2022]
Abstract
Osteoblasts are a major component of the bone marrow microenvironment, which provide support for hematopoietic cell development. Functional disruption of any element of the bone marrow niche, including osteoblasts, can potentially impair hematopoiesis. We have studied the effect of two widely used drugs with different mechanisms of action, etoposide (VP16) and melphalan, on murine osteoblasts at distinct stages of maturation. VP16 and melphalan delayed maturation of preosteoblasts and altered CXCL12 protein levels, a key regulator of hematopoietic cell homing to the bone marrow. Sublethal concentrations of VP16 and melphalan also decreased the levels of several transcripts which contribute to the composition of the extracellular matrix (ECM) including osteopontin (OPN), osteocalcin (OCN), and collagen 1A1 (Col1a1). The impact of chemotherapy on message and protein levels for some targets was not always aligned, suggesting differential responses at the transcription and translation or protein stability levels. As one of the main functions of a mature osteoblast is to synthesize ECM of a defined composition, disruption of the ratio of its components may be one mechanism by which chemotherapy affects the ability of osteoblasts to support hematopoietic recovery coincident with altered marrow architecture. Collectively, these observations suggest that the osteoblast compartment of the marrow hematopoietic niche is vulnerable to functional dysregulation by damage imposed by agents frequently used in clinical settings. Understanding the mechanistic underpinning of chemotherapy-induced changes on the hematopoietic support capacity of the marrow microenvironment may contribute to improved strategies to optimize patient recovery post-transplantation.
Collapse
Affiliation(s)
- Marieta Gencheva
- Alexander B. Osborn Hematopoietic Malignancy and Transplantation Program of the Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, WV 26506-9300, USA
| | | | | | | | | | | | | |
Collapse
|
182
|
Strecker S, Fu Y, Liu Y, Maye P. Generation and characterization of Osterix-Cherry reporter mice. Genesis 2013; 51:246-58. [PMID: 23180553 PMCID: PMC3602345 DOI: 10.1002/dvg.22360] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/15/2012] [Accepted: 11/17/2012] [Indexed: 01/22/2023]
Abstract
Osterix is a zinc finger containing transcription factor, which functions as a key regulator of osteoblast differentiation. To better understand the temporal and spatial expression of Osterix during embryonic development and in the adult skeleton, we generated Osterix-Cherry reporter mice. Bacterial recombination techniques were employed to engineer a transgenic construct, which consisted of a ∼39 kb DNA fragment encompassing the Osterix/Sp7 gene, but excluding adjacent gene sequences. Osterix reporter expression was characterized at embryonic, neonatal, and adult ages both by itself and in the context of a cross with Bone Sialoprotein (BSP)-Topaz reporter mice. Relative to Osterix, BSP is a more mature marker of osteoblast differentiation. In agreement with osteoblast lineage maturation, Osterix reporter expression preceded BSP reporter expression during embryonic development and spatially appeared in a much broader cell population. Strong Osterix reporter expression was observed in mature osteoblasts and osteocytes. However, weaker Osterix-Cherry positive cells were also observed in the bone marrow, possibly identifying an early osteoprogenitor cell population. Evaluation of Osterix reporter expression in male femur tissue sections from 10 days to 12 weeks of age revealed persistent expression in cells of the osteoblast lineage and a surprising increase in maturing chondrocytes of the growth plate. Also, Osterix reporter expression was transiently detected in the kidney after birth.
Collapse
Affiliation(s)
- Sara Strecker
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| | - Yu Fu
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| | - Yaling Liu
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| | - Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center
| |
Collapse
|
183
|
Siclari VA, Zhu J, Akiyama K, Liu F, Zhang X, Chandra A, Nah-Cederquist HD, Shi S, Qin L. Mesenchymal progenitors residing close to the bone surface are functionally distinct from those in the central bone marrow. Bone 2013; 53:575-86. [PMID: 23274348 PMCID: PMC3674849 DOI: 10.1016/j.bone.2012.12.013] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/12/2012] [Accepted: 12/15/2012] [Indexed: 12/15/2022]
Abstract
Long bone is an anatomically complicated tissue with trabecular-rich metaphyses at two ends and cortical-rich diaphysis at the center. The traditional flushing method isolates only mesenchymal progenitor cells from the central region of long bones and these cells are distant from the bone surface. We propose that mesenchymal progenitors residing in endosteal bone marrow that is close to the sites of bone formation, such as trabecular bone and endosteum, behave differently from those in the central bone marrow. In this report, we separately isolated endosteal bone marrow using a unique enzymatic digestion approach and demonstrated that it contained a much higher frequency of mesenchymal progenitors than the central bone marrow. Endosteal mesenchymal progenitors express common mesenchymal stem cell markers and are capable of multi-lineage differentiation. However, we found that mesenchymal progenitors isolated from different anatomical regions of the marrow did exhibit important functional differences. Compared with their central marrow counterparts, endosteal mesenchymal progenitors have superior proliferative ability with reduced expression of cell cycle inhibitors. They showed greater immunosuppressive activity in culture and in a mouse model of inflammatory bowel disease. Aging is a major contributing factor for trabecular bone loss. We found that old mice have a dramatically decreased number of endosteal mesenchymal progenitors compared with young mice. Parathyroid hormone (PTH) treatment potently stimulates bone formation. A single PTH injection greatly increased the number of endosteal mesenchymal progenitors, particularly those located at the metaphyseal bone, but had no effect on their central counterparts. In summary, endosteal mesenchymal progenitors are more metabolically active and relevant to physiological bone formation than central mesenchymal progenitors. Hence, they represent a biologically important target for future mesenchymal stem cell studies.
Collapse
Affiliation(s)
- Valerie A. Siclari
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | - Ji Zhu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | - Kentaro Akiyama
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Fei Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | - Xianrong Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | - Abhishek Chandra
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| | - Hyun-Duck Nah-Cederquist
- Department of Plastic and Reconstructive Surgery, The Children’s Hospital of Philadelphia, 1116G Abramson, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Songtao Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 424 Stemmler Hall, 36St and Hamilton Walk, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
184
|
Kuhn LT, Ou G, Charles L, Hurley MM, Rodner CM, Gronowicz G. Fibroblast growth factor-2 and bone morphogenetic protein-2 have a synergistic stimulatory effect on bone formation in cell cultures from elderly mouse and human bone. J Gerontol A Biol Sci Med Sci 2013; 68:1170-80. [PMID: 23531867 DOI: 10.1093/gerona/glt018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Combined regimens of fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) were investigated to stimulate osteogenic differentiation. In young mouse calvaria-derived cells, FGF-2 (0.16ng/mL) in combination with BMP-2 (50ng/mL) did not enhance mineralization, but in old mouse cells it resulted in more mineralization than BMP-2 alone. In young long bone mouse cultures, FGF-2 enhanced mineralization relative to BMP-2 alone, but in old cultures, lower dose of FGF-2 (0.016ng/mL) was necessary. In neonatal mouse calvarial cells, sequential delivery of low-dose FGF-2 and low-dose BMP-2 (5ng/mL) was more stimulatory than co-delivery. In young human cultures, 0.016ng/mL of FGF-2 did not enhance mineralization, in combination with 5ng/mL of BMP-2, but in older cultures, codelivery of FGF-2 and BMP-2 was superior to BMP-2 alone. In conclusion, BMP-2 treatment alone was sufficient for maximal mineralization in young osteoblast cultures. However, coadministration of FGF-2 and BMP-2 increases mineralization more than BMP-2 alone in cultures from old and young mouse long bones and old humans but not in young mouse calvarial cultures.
Collapse
Affiliation(s)
- Liisa T Kuhn
- Department of Surgery MC-3105, University of Connecticut Health Center, Farmington, CT 06030-3105.
| | | | | | | | | | | |
Collapse
|
185
|
Su N, Chen M, Chen S, Li C, Xie Y, Zhu Y, Zhang Y, Zhao L, He Q, Du X, Chen D, Chen L. Overexpression of H1 calponin in osteoblast lineage cells leads to a decrease in bone mass by disrupting osteoblast function and promoting osteoclast formation. J Bone Miner Res 2013; 28:660-71. [PMID: 23044709 PMCID: PMC3716280 DOI: 10.1002/jbmr.1778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 08/24/2012] [Accepted: 09/07/2012] [Indexed: 02/03/2023]
Abstract
H1 calponin (CNN1) is known as a smooth muscle-specific, actin-binding protein which regulates smooth muscle contractive activity. Although previous studies have shown that CNN1 has effect on bone, the mechanism is not well defined. To investigate the role of CNN1 in maintaining bone homeostasis, we generated transgenic mice overexpressing Cnn1 under the control of the osteoblast-specific 3.6-kb Col1a1 promoter. Col1a1-Cnn1 transgenic mice showed delayed bone formation at embryonic stage and decreased bone mass at adult stage. Morphology analyses showed reduced trabecular number, thickness and defects in bone formation. The proliferation and migration of osteoblasts were decreased in Col1a1-Cnn1 mice due to alterations in cytoskeleton. The early osteoblast differentiation of Col1a1-Cnn1 mice was increased, but the late stage differentiation and mineralization of osteoblasts derived from Col1a1-Cnn1 mice were significantly decreased. In addition to impaired bone formation, the decreased bone mass was also associated with enhanced osteoclastogenesis. Tartrate-resistant acid phosphatase (TRAP) staining revealed increased osteoclast numbers in tibias of 2-month-old Col1a1-Cnn1 mice, and increased numbers of osteoclasts co-cultured with Col1a1-Cnn1 osteoblasts. The ratio of RANKL to OPG was significantly increased in Col1a1-Cnn1 osteoblasts. These findings reveal a novel function of CNN1 in maintaining bone homeostasis by coupling bone formation to bone resorption.
Collapse
Affiliation(s)
- Nan Su
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Maomao Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Siyu Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Can Li
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ying Zhu
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yaozong Zhang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Ling Zhao
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Qifen He
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
186
|
Canalis E, Parker K, Feng JQ, Zanotti S. Osteoblast lineage-specific effects of notch activation in the skeleton. Endocrinology 2013; 154:623-34. [PMID: 23275471 PMCID: PMC3548181 DOI: 10.1210/en.2012-1732] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Transgenic overexpression of the Notch1 intracellular domain inhibits osteoblast differentiation and causes osteopenia, and inactivation of Notch1 and Notch2 increases bone volume transiently and induces osteoblastic differentiation. However, the biology of Notch is cell-context-dependent, and consequences of Notch activation in cells of the osteoblastic lineage at various stages of differentiation and in osteocytes have not been defined. For this purpose, Rosa(Notch) mice, where a loxP-flanked STOP cassette placed between the Rosa26 promoter and the NICD coding sequence, were crossed with transgenics expressing the Cre recombinase under the control of the Osterix (Osx), Osteocalcin (Oc), Collagen 1a1 (Col2.3), or Dentin matrix protein1 (Dmp1) promoters. At 1 month, Osx-Cre;Rosa(Notch) and Oc-Cre;Rosa(Notch) mice exhibited osteopenia due to impaired bone formation. In contrast, Col2.3-Cre;Rosa(Notch) and Dmp1-Cre;Rosa(Notch) exhibited increased femoral trabecular bone volume due to a decrease in osteoclast number and eroded surface. In the four lines studied, cortical bone was either not present, was porous, or had the appearance of trabecular bone. Oc-Cre;Rosa(Notch) and Col2.3-Cre;Rosa(Notch) mice exhibited early lethality so that their adult phenotype was not established. At 3 months, Osx-Cre;Rosa(Notch) and Dmp1-Cre;Rosa(Notch) mice displayed increased bone volume, and increased osteoblasts although calcein-demeclocycline labels were diffuse and fragmented, indicating abnormal bone formation. In conclusion, Notch effects in the skeleton are cell-context-dependent. When expressed in immature osteoblasts, Notch arrests their differentiation, causing osteopenia, and when expressed in osteocytes, it causes an initial suppression of bone resorption and increased bone volume, a phenotype that evolves as the mice mature.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, 114 Woodland Street, Hartford, CT 06105-1299, USA.
| | | | | | | |
Collapse
|
187
|
Yang B, Tang Q, Post J, Zhou H, Huang XB, Zhang XD, Wang Q, Sun YM, Fan FY. Effect of radiation on the Notch signaling pathway in osteoblasts. Int J Mol Med 2013; 31:698-706. [PMID: 23340672 DOI: 10.3892/ijmm.2013.1255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 12/28/2012] [Indexed: 11/06/2022] Open
Abstract
Notch signaling has been shown to be important in osteoblast differentiation. Therapeutic radiation has been shown to alter the skeletal system, yet little information is available on the changes in Notch signaling in irradiated osteoblasts. The purpose of this study was to analyze the effect of radiation therapy with 2 and 4 Gy on Notch signaling in osteoblasts. In order to assess the radiation damage on osteoblast differentiation, total RNA and protein were collected three days after exposure to radiation. The effects of radiation on Notch signaling at the early and terminal stages of osteoblastic MC3T3-E1 cell differentiation was analyzed by qRT-PCR and western blot analysis. Our study applied a previously established method to induce MC3T3-E1 cell differentiation into osteoblasts and osteoblast precursors. Our results showed that the expression of Notch receptors (Notch1-4), ligands (Jagged1, Jagged2 and Delta1), target of Notch signaling (Hes1) and markers (ALP, M-CSF, RANKL and OPG) were altered following 2 and 4 Gy of irradiation. The present research did not indicate a strong relationship between Notch1 regulation and suppression of osteoblast differentiation. We found Hes1 may play a role in the radiation effect on osteoblast differentiation. Our results indicate that radiated osteoblast precursors and osteoblasts promoted osteoclast differentiation and proliferation.
Collapse
Affiliation(s)
- Bing Yang
- Tianjin Key Laboratory of Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Repic D, Torreggiani E, Franceschetti T, Matthews BG, Ivcevic S, Lichtler AC, Grcevic D, Kalajzic I. Utilization of transgenic models in the evaluation of osteogenic differentiation of embryonic stem cells. Connect Tissue Res 2013; 54:296-304. [PMID: 23782451 PMCID: PMC3893759 DOI: 10.3109/03008207.2013.814646] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Previous studies reported that embryonic stem cells (ESCs) can be induced to differentiate into cells showing a mature osteoblastic phenotype by culturing them under osteo-inductive conditions. It is probable that osteogenic differentiation requires that ESCs undergo differentiation through an intermediary step involving a mesenchymal lineage precursor. Based on our previous studies indicating that adult mesenchymal progenitor cells express α-smooth muscle actin (αSMA), we have generated ESCs from transgenic mice in which an αSMA promoter directs the expression of red fluorescent protein (RFP) to mesenchymal progenitor cells. To track the transition of ESC-derived MSCs into mature osteoblasts, we have utilized a bone-specific fragment of rat type I collagen promoter driving green fluorescent protein (Col2.3GFP). Following osteogenic induction in ESCs, we have observed expression of alkaline phosphatase (ALP) and subsequent mineralization as detected by von Kossa staining. After 1 week of osteogenic induction, ESCs begin to express αSMARFP. This expression was localized to the peripheral area encircling a typical ESC colony. Nevertheless, these αSMARFP positive cells did not show activation of the Col2.3GFP promoter, even after 7 weeks of osteogenic differentiation in vitro. In contrast, Col2.3GFP expression was detected in vivo, in mineralized areas following teratoma formation. Our results indicate that detection of ALP activity and mineralization of ESCs cultured under osteogenic conditions is not sufficient to demonstrate osteogenic maturation. Our study indicates the utility of the promoter-visual transgene approach to assess the commitment and differentiation of ESCs into the osteoblast lineage.
Collapse
Affiliation(s)
- Dario Repic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA,University of Split, School of Dental Medicine, Split Croatia
| | - Elena Torreggiani
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Tiziana Franceschetti
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Brya G. Matthews
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Sanja Ivcevic
- Department of Physiology and Immunology, University School of Medicine, Zagreb, Croatia
| | - Alexander C. Lichtler
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Danka Grcevic
- Department of Physiology and Immunology, University School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
189
|
Zhu J, Siclari VA, Liu F, Spatz JM, Chandra A, Divieti Pajevic P, Qin L. Amphiregulin-EGFR signaling mediates the migration of bone marrow mesenchymal progenitors toward PTH-stimulated osteoblasts and osteocytes. PLoS One 2012; 7:e50099. [PMID: 23300521 PMCID: PMC3534030 DOI: 10.1371/journal.pone.0050099] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/16/2012] [Indexed: 12/31/2022] Open
Abstract
Intermittent administration of parathyroid hormone (PTH) dramatically increases bone mass and currently is one of the most effective treatments for osteoporosis. However, the detailed mechanisms are still largely unknown. Here we demonstrate that conditioned media from PTH-treated osteoblastic and osteocytic cells contain soluble chemotactic factors for bone marrow mesenchymal progenitors, which express a low amount of PTH receptor (PTH1R) and do not respond to PTH stimulation by increasing cAMP production or migrating toward PTH alone. Conditioned media from PTH-treated osteoblasts elevated phosphorylated Akt and p38MAPK amounts in mesenchymal progenitors and inhibition of these pathways blocked the migration of these progenitors toward conditioned media. Our previous and current studies revealed that PTH stimulates the expression of amphiregulin, an epidermal growth factor (EGF)-like ligand that signals through the EGF receptor (EGFR), in both osteoblasts and osteocytes. Interestingly, conditioned media from PTH-treated osteoblasts increased EGFR phosphorylation in mesenchymal progenitors. Using several different approaches, including inhibitor, neutralizing antibody, and siRNA, we demonstrate that PTH increases the release of amphiregulin from osteoblastic cells, which acts on the EGFRs expressed on mesenchymal progenitors to stimulate the Akt and p38MAPK pathways and subsequently promote their migration in vitro. Furthermore, inactivation of EGFR signaling specifically in osteoprogenitors/osteoblasts attenuated the anabolic actions of PTH on bone formation. Taken together, these results suggest a novel mechanism for the therapeutic effect of PTH on osteoporosis and an important role of EGFR signaling in mediating PTH's anabolic actions on bone.
Collapse
Affiliation(s)
- Ji Zhu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Valerie A. Siclari
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Fei Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai, China
| | - Jordan M. Spatz
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Abhishek Chandra
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Paola Divieti Pajevic
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ling Qin
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
190
|
Pivonka P, Dunstan CR. Role of mathematical modeling in bone fracture healing. BONEKEY REPORTS 2012; 1:221. [PMID: 24228159 PMCID: PMC3727792 DOI: 10.1038/bonekey.2012.221] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 10/11/2012] [Indexed: 01/05/2023]
Abstract
Bone fracture healing is a complex physiological process commonly described by a four-phase model consisting of an inflammatory phase, two repair phases with soft callus formation followed by hard callus formation, and a remodeling phase, or more recently by an anabolic/catabolic model. Data from humans and animal models have demonstrated crucial environmental conditions for optimal fracture healing, including the mechanical environment, blood supply and availability of mesenchymal stem cells. Fracture healing spans multiple length and time scales, making it difficult to know precisely which factors and/or phases to manipulate in order to obtain optimal fracture-repair outcomes. Deformations resulting from physiological loading or fracture fixation at the organ scale are sensed at the cellular scale by cells inside the fracture callus. These deformations together with autocrine and paracrine signals determine cellular differentiation, proliferation and migration. The local repair activities lead to new bone formation and stabilization of the fracture. Although experimental data are available at different spatial and temporal scales, it is not clear how these data can be linked to provide a holistic view of fracture healing. Mathematical modeling is a powerful tool to quantify conceptual models and to establish the missing links between experimental data obtained at different scales. The objective of this review is to introduce mathematical modeling to readers who are not familiar with this methodology and to demonstrate that once validated, such models can be used for hypothesis testing and to assist in clinical treatment as will be shown for the example of atrophic nonunions.
Collapse
Affiliation(s)
- Peter Pivonka
- Faculty of Engineering, Computing and Mathematics, University of Western Australia, WA, Australia
| | - Colin R Dunstan
- Biomedical Engineering, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
191
|
Sugimura R, He XC, Venkatraman A, Arai F, Box A, Semerad C, Haug JS, Peng L, Zhong XB, Suda T, Li L. Noncanonical Wnt signaling maintains hematopoietic stem cells in the niche. Cell 2012; 150:351-65. [PMID: 22817897 DOI: 10.1016/j.cell.2012.05.041] [Citation(s) in RCA: 222] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/18/2012] [Accepted: 05/01/2012] [Indexed: 12/17/2022]
Abstract
Wnt signaling is involved in self-renewal and maintenance of hematopoietic stem cells (HSCs); however, the particular role of noncanonical Wnt signaling in regulating HSCs in vivo is largely unknown. Here, we show Flamingo (Fmi) and Frizzled (Fz) 8, members of noncanonical Wnt signaling, both express in and functionally maintain quiescent long-term HSCs. Fmi regulates Fz8 distribution at the interface between HSCs and N-cadherin(+) osteoblasts (N-cad(+)OBs that enrich osteoprogenitors) in the niche. We further found that N-cad(+)OBs predominantly express noncanonical Wnt ligands and inhibitors of canonical Wnt signaling under homeostasis. Under stress, noncanonical Wnt signaling is attenuated and canonical Wnt signaling is enhanced in activation of HSCs. Mechanistically, noncanonical Wnt signaling mediated by Fz8 suppresses the Ca(2+)-NFAT- IFNγ pathway, directly or indirectly through the CDC42-CK1α complex and also antagonizes canonical Wnt signaling in HSCs. Taken together, our findings demonstrate that noncanonical Wnt signaling maintains quiescent long-term HSCs through Fmi and Fz8 interaction in the niche.
Collapse
Affiliation(s)
- Ryohichi Sugimura
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Olson C, Uribe F, Kalajzic Z, Utreja A, Nanda R, Rowe D, Wadhwa S. Orthodontic tooth movement causes decreased promoter expression of collagen type 1, bone sialoprotein and alpha-smooth muscle actin in the periodontal ligament. Orthod Craniofac Res 2012; 15:52-61. [PMID: 22264327 DOI: 10.1111/j.1601-6343.2011.01536.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To evaluate the effects of orthodontic tooth movement on the promoter expression of collagen type 1 (3.6Col1), bone sialoprotein (BSP) and alpha-smooth muscle actin (αSMA) in the periodontal ligament (PDL) using transgenic mice containing transgenes of these promoters fused to green fluorescent proteins (GFP). MATERIALS AND METHODS The maxillary first molars of 10-12 week-old transgenic mice were loaded with 10-12 g of force for 12, 48 h, or 7 days. Mice were transgenic for one of the following GFP-tagged bone markers of osteoblast lineage cells: 3.6-kb fragment of the rat collagen type 1 promoter (3.6Col1), BSP or α-smooth muscle actin (αSMA). Loaded molars under compression and tension were compared with contra-lateral unloaded controls. RESULTS On the compression side of the PDL, orthodontic tooth movement caused a significant decrease in GFP expression of all the promoters at each time point. On the tension side, there was a significant increase in BSP-GFP expression, 12 h following loading compared to the contralateral unloaded controls. CONCLUSIONS An in vivo tooth movement model using transgenic mice with promoter-GFP constructs provides an efficient and effective way of investigating the cellular events underlying orthodontic tooth movement. PDL cells may undergo decreased differentiation in response to the compressive force.
Collapse
|
193
|
Reichenberger EJ, Levine MA, Olsen BR, Papadaki ME, Lietman SA. The role of SH3BP2 in the pathophysiology of cherubism. Orphanet J Rare Dis 2012; 7 Suppl 1:S5. [PMID: 22640988 PMCID: PMC3359958 DOI: 10.1186/1750-1172-7-s1-s5] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cherubism is a rare bone dysplasia that is characterized by symmetrical bone resorption limited to the jaws. Bone lesions are filled with soft fibrous giant cell-rich tissue that can expand and cause severe facial deformity. The disorder typically begins in children at ages of 2-5 years and the bone resorption and facial swelling continues until puberty; in most cases the lesions regress spontaneously thereafter. Most patients with cherubism have germline mutations in the gene encoding SH3BP2, an adapter protein involved in adaptive and innate immune response signaling. A mouse model carrying a Pro416Arg mutation in SH3BP2 develops osteopenia and expansile lytic lesions in bone and some soft tissue organs. In this review we discuss the genetics of cherubism, the biological functions of SH3BP2 and the analysis of the mouse model. The data suggest that the underlying cause for cherubism is a systemic autoinflammatory response to physiologic challenges despite the localized appearance of bone resorption and fibrous expansion to the jaws in humans.
Collapse
Affiliation(s)
- Ernst J Reichenberger
- Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut Health Center, Farmington, CT, USA.
| | | | | | | | | |
Collapse
|
194
|
Javaheri B, Sunters A, Zaman G, Suswillo RFL, Saxon LK, Lanyon LE, Price JS. Lrp5 is not required for the proliferative response of osteoblasts to strain but regulates proliferation and apoptosis in a cell autonomous manner. PLoS One 2012; 7:e35726. [PMID: 22567110 PMCID: PMC3342322 DOI: 10.1371/journal.pone.0035726] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/22/2012] [Indexed: 11/19/2022] Open
Abstract
Although Lrp5 is known to be an important contributor to the mechanisms regulating bone mass, its precise role remains unclear. The aim of this study was to establish whether mutations in Lrp5 are associated with differences in the growth and/or apoptosis of osteoblast-like cells and their proliferative response to mechanical strain in vitro. Primary osteoblast-like cells were derived from cortical bone of adult mice lacking functional Lrp5 (Lrp5(-/-)), those heterozygous for the human G171V High Bone Mass (HBM) mutation (LRP5(G171V)) and their WT littermates (WT(Lrp5), WT(HBM)). Osteoblast proliferation over time was significantly higher in cultures of cells from LRP5(G171V) mice compared to their WT(HBM) littermates, and lower in Lrp5(-/-) cells. Cells from female LRP5(G171V) mice grew more rapidly than those from males, whereas cells from female Lrp5(-/-) mice grew more slowly than those from males. Apoptosis induced by serum withdrawal was significantly higher in cultures from Lrp5(-/-) mice than in those from WT(HBM) or LRP5(G171V) mice. Exposure to a single short period of dynamic mechanical strain was associated with a significant increase in cell number but this response was unaffected by genotype which also did not change the 'threshold' at which cells responded to strain. In conclusion, the data presented here suggest that Lrp5 loss and gain of function mutations result in cell-autonomous alterations in osteoblast proliferation and apoptosis but do not alter the proliferative response of osteoblasts to mechanical strain in vitro.
Collapse
Affiliation(s)
- Behzad Javaheri
- Department of Oral Biology, UMKC School of Dentistry, Kansas City, Missouri, United States of America.
| | | | | | | | | | | | | |
Collapse
|
195
|
Aguila HL, Mun SH, Kalinowski J, Adams DJ, Lorenzo JA, Lee SK. Osteoblast-specific overexpression of human interleukin-7 rescues the bone mass phenotype of interleukin-7-deficient female mice. J Bone Miner Res 2012; 27:1030-42. [PMID: 22258693 PMCID: PMC3361560 DOI: 10.1002/jbmr.1553] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-7 is a critical cytokine for lymphoid development and a direct inhibitor of in vitro osteoclastogenesis in murine bone marrow cultures. To explore the role of IL-7 in bone, we generated transgenic mouse lines bearing the 2.3-kb rat collagen 1α1 promoter driving the expression of human IL-7 specifically in osteoblasts. In addition, we crossed these mice with IL-7-deficient mice to determine if the alterations in lymphopoiesis, bone mass, and osteoclast formation observed in the IL-7 knockout (KO) mice could be rescued by osteoblast-specific overexpression of IL-7. Here, we show that mice overexpressing human IL-7 in the osteoblast lineage showed increased trabecular bone volume in vivo by µCT and decreased osteoclast formation in vitro. Furthermore, targeted overexpression of IL-7 in osteoblasts rescued the osteopenic bone phenotype and B-cell development of IL-7 KO mice but did not have an effect on T lymphopoiesis, which occurs in the periphery. The bone phenotypes in IL-7 KO mice and targeted IL-7-overexpressing mouse models were observed only in females. These results likely reflect both direct inhibitory effects of IL-7 on osteoclastogenesis in vivo and sex-specific differences in responses to IL-7.
Collapse
Affiliation(s)
- Hector L. Aguila
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Se Hwan Mun
- UCONN Center on Aging, University of Connecticut Health Center, Farmington, CT 06030
| | - Judith Kalinowski
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Douglas J. Adams
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030
| | - Joseph A. Lorenzo
- Division of Endocrinology, Department of Medicine, University of Connecticut Health Center, Farmington, CT 06030
| | - Sun-Kyeong Lee
- UCONN Center on Aging, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
196
|
Yu X, Wang L, Peng F, Jiang X, Xia Z, Huang J, Rowe D, Wei M. The effect of fresh bone marrow cells on reconstruction of mouse calvarial defect combined with calvarial osteoprogenitor cells and collagen-apatite scaffold. J Tissue Eng Regen Med 2012; 7:974-83. [PMID: 22473786 DOI: 10.1002/term.1490] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 01/10/2012] [Accepted: 01/19/2012] [Indexed: 11/08/2022]
Abstract
Fresh bone marrow cells have already exhibited its advantages as osteogenic donor cells, but the combination between fresh bone marrow cells and other donor cells utilized for bone healing has not been fully explored. To highlight the impact of fresh bone marrow cells on scaffold-based bone regeneration, single or a combination of calvarial osteoprogenitor cells (OPCs) and bone marrow cells (BMCs) were used as donor cells combined with collagen-apatite scaffold for calvarial defect healing. The host and donor contributions to bone formation were assessed using histological and GFP imaging analysis. Although the amount of new bone formed by different cell sources did not show significant differences, the origin of the bone formation in the defects mainly depended on the types of donor cells employed: when only calvarial OPCs were used as donor cells, a donor-derived bone healing instead of host-derived bone ingrowth was observed; when only fresh BMCs were loaded, the host bone could grow into the defect along the lamellar structure of the scaffolds, but the amount of new bone formed was significantly lower than the defect loaded with calvarial OPCs only. The combination of calvarial OPCs and fresh BMCs had similar amount of new bone formation as the group loaded with calvarial osteoprogenitors alone, but did not induce any host-derived bone formation. These results provide compelling evidence of the importance of fresh BMCs to induce host-implant integration in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaohua Yu
- Department of Chemical, Materials and Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | | | | | | | | | | | | | | |
Collapse
|
197
|
Zanotti S, Canalis E. Notch regulation of bone development and remodeling and related skeletal disorders. Calcif Tissue Int 2012; 90:69-75. [PMID: 22002679 PMCID: PMC3272107 DOI: 10.1007/s00223-011-9541-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 09/14/2011] [Indexed: 10/16/2022]
Abstract
Notch signaling mediates cell-to-cell interactions that are critical for embryonic development and tissue renewal. In the canonical signaling pathway, the Notch receptor is cleaved following ligand binding, resulting in the release and nuclear translocation of the Notch intracellular domain (NICD). NICD induces gene expression by forming a ternary complex with the DNA binding protein CBF1/Rbp-Jk, Suppressor of Hairless, Lag1, and Mastermind-Like (Maml). Hairy Enhancer of Split (Hes) and Hes related with YRPW motif (Hey) are classic Notch targets. Notch canonical signaling plays a central role in skeletal development and bone remodeling by suppressing the differentiation of skeletal cells. The skeletal phenotype of mice misexpressing Hes1 phenocopies partially the effects of Notch misexpression, suggesting that Hey proteins mediate most of the skeletal effects of Notch. Dysregulation of Notch signaling is associated with diseases affecting human skeletal development, such as Alagille syndrome, brachydactyly and spondylocostal dysostosis. Somatic mutations in Notch receptors and ligands are found in tumors of the skeletal system. Overexpression of NOTCH1 is associated with osteosarcoma, and overexpression of NOTCH3 or JAGGED1 in breast cancer cells favors the formation of osteolytic bone metastasis. Activating mutations in NOTCH2 cause Hajdu-Cheney syndrome, which is characterized by skeletal defects and fractures, and JAG1 polymorphisms, are associated with variations in bone mineral density. In conclusion, Notch is a regulator of skeletal development and bone remodeling, and abnormal Notch signaling is associated with developmental and postnatal skeletal disorders.
Collapse
Affiliation(s)
- Stefano Zanotti
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, CT 06105 USA
- The University of Connecticut School of Medicine, Farmington, CT 06030, USA
| | - Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, CT 06105 USA
- The University of Connecticut School of Medicine, Farmington, CT 06030, USA
- Address correspondence: Department of Research Saint Francis Hospital and Medical Center 114 Woodland Street Hartford, CT 06105-1299 Tel: (860)714-4068 Fax: (860)714-8053
| |
Collapse
|
198
|
Liang B, Cotter MM, Chen D, Hernandez CJ, Zhou G. Ectopic expression of SOX9 in osteoblasts alters bone mechanical properties. Calcif Tissue Int 2012; 90:76-89. [PMID: 22143895 PMCID: PMC3272153 DOI: 10.1007/s00223-011-9550-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 11/11/2011] [Indexed: 12/28/2022]
Abstract
Osteoporosis is a common skeletal disease characterized by low bone mass and microarchitectural deterioration of bone tissue, with a consequent increase in bone fragility and susceptibility to fracture. We previously demonstrated that Col1a1-SOX9 transgenic (TG) mice, in which SOX9 specifically expresses in osteoblasts driven by a 2.3-kb Col1a1 promoter, display osteopenia during the early postnatal stage. In this study, to further analyze the osteopenia phenotype and especially the effect of the osteoblast-specific expression of SOX9 on bone mechanical properties, we performed bone geometry and mechanical property analysis of long bones from Col1a1-SOX9 TG mice and wild-type littermates (WT) at different time points. Interestingly, after body weight adjustment, TG mice had similar whole-bone strength as WT mice but significantly thinner cortical bone, lower elastic modulus, and higher moment of inertia. Thus, osteoblast-specific SOX9 expression results in altered bone structure and material properties. Furthermore, the expression levels of Pcna, Col1a1, osteocalcin, and the Opg/Rankl ratio in TG mice were significantly lower until 4 months of age compared with WT mice, suggesting that TG mice have dysregulated bone homeostasis. Finally, bone marrow stromal cells (MSCs) isolated from TG mice display enhanced adipocyte differentiation and decreased osteoblast differentiation in vitro, suggesting that osteoblast-specific expression of SOX9 can lead to altered mesenchymal stem cell differentiation potentials. In conclusion, our study implies that SOX9 activity has to be tightly regulated in the adult skeleton to ensure optimal bone quality.
Collapse
Affiliation(s)
- Bojian Liang
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
| | - Meghan M. Cotter
- Department of Anatomy, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
| | - Dongxing Chen
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
| | - Christopher J. Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY 14853, U.S.A
| | - Guang Zhou
- Department of Orthopaedics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
- Department of Genetics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, U. S. A
- Corresponding author: Guang Zhou, Ph.D., Department of Orthopaedics, BRB, Room 328, Case Western Reserve University, 2109 Adelbert Road, Cleveland, OH 44106, U. S. A., Tel: (216) 368-2260, Fax: (216) 368-1332,
| |
Collapse
|
199
|
Grcevic D, Pejda S, Matthews BG, Repic D, Wang L, Li H, Kronenberg MS, Jiang X, Maye P, Adams DJ, Rowe DW, Aguila HL, Kalajzic I. In vivo fate mapping identifies mesenchymal progenitor cells. Stem Cells 2012; 30:187-96. [PMID: 22083974 PMCID: PMC3560295 DOI: 10.1002/stem.780] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Adult mesenchymal progenitor cells have enormous potential for use in regenerative medicine. However, the true identity of the progenitors in vivo and their progeny has not been precisely defined. We hypothesize that cells expressing a smooth muscle α-actin promoter (αSMA)-directed Cre transgene represent mesenchymal progenitors of adult bone tissue. By combining complementary colors in combination with transgenes activating at mature stages of the lineage, we characterized the phenotype and confirmed the ability of isolated αSMA(+) cells to progress from a progenitor to fully mature state. In vivo lineage tracing experiments using a new bone formation model confirmed the osteogenic phenotype of αSMA(+) cells. In vitro analysis of the in vivo-labeled SMA9(+) cells supported their differentiation potential into mesenchymal lineages. Using a fracture-healing model, αSMA9(+) cells served as a pool of fibrocartilage and skeletal progenitors. Confirmation of the transition of αSMA9(+) progenitor cells to mature osteoblasts during fracture healing was assessed by activation of bone-specific Col2.3emd transgene. Our findings provide a novel in vivo identification of defined population of mesenchymal progenitor cells with active role in bone remodeling and regeneration.
Collapse
Affiliation(s)
- Danka Grcevic
- Department of Physiology and Immunology, University School of Medicine, Zagreb, Croatia
| | - Slavica Pejda
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
- University School of Dental Medicine, Split Croatia
| | - Brya G. Matthews
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dario Repic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
- University School of Dental Medicine, Split Croatia
| | - Liping Wang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Haitao Li
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Mark S. Kronenberg
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Xi Jiang
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Peter Maye
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Douglas J. Adams
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - David W. Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hector L. Aguila
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
200
|
Abdallah BM, Kassem M. New factors controlling the balance between osteoblastogenesis and adipogenesis. Bone 2012; 50:540-5. [PMID: 21745614 DOI: 10.1016/j.bone.2011.06.030] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/20/2011] [Accepted: 06/23/2011] [Indexed: 02/01/2023]
Abstract
The majority of conditions associated with bone loss, including aging, are accompanied by increased marrow adiposity possibly due to shifting of the balance between osteoblast and adipocyte differentiation in bone marrow stromal (skeletal) stem cells (MSC). In order to study the relationship between osteoblastogenesis and adipogenesis in bone marrow, we have characterized cellular models of multipotent MSC as well as pre-osteoblastic and pre-adipocytic cell populations. Using these models, we identified two secreted factors in the bone marrow microenviroment: secreted frizzled-related protein 1 (sFRP-1) and delta-like1 (preadipocyte factor 1) (Dlk1/Pref-1). Both exert regulatory effects on osteoblastogenesis and adipogenesis. Our studies suggest a model for lineage fate determination of MSC that is regulated through secreted factors in the bone marrow microenvironment that mediate a cross-talk between lineage committed cell populations in addition to controlling differentiation choices of multipotent MSC.
Collapse
Affiliation(s)
- Basem M Abdallah
- Endocrine Research Laboratory (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|