201
|
Dishowitz MI, Terkhorn SP, Bostic SA, Hankenson KD. Notch signaling components are upregulated during both endochondral and intramembranous bone regeneration. J Orthop Res 2012; 30:296-303. [PMID: 21818769 PMCID: PMC3210892 DOI: 10.1002/jor.21518] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Accepted: 07/13/2011] [Indexed: 02/04/2023]
Abstract
Previous studies have demonstrated that Notch signaling regulates endochondral and intramembranous bone formation by controlling cell proliferation and differentiation. Notch signaling has also been shown to regulate healing in a variety of tissues. The objective of this study was to characterize and compare activation of the Notch signaling pathway during endochondral and intramembranous bone healing using tibial fracture and calvarial defect injury models, respectively. Bilateral tibial fractures or bilateral 1.5 mm diameter calvarial defects were created in mice, and tissues were harvested at 0, 5, 10, and 20 days post-fracture. Gene expression of Notch signaling components was upregulated during both tibial fracture and calvarial defect healing, with expression generally higher during tibial fracture healing. The most highly expressed ligand and receptor during healing, Jag1 and Notch2 (specifically the activated receptor, known as NICD2), were similarly localized in mesenchymal cells during both modes of healing, with expression decreasing during chondrogenesis, but remaining present in osteoblasts at all stages of maturity. Results suggest that in addition to embryological bone development, Notch signaling regulates both endochondral and intramembranous bone healing.
Collapse
Affiliation(s)
| | - Shawn P. Terkhorn
- Department of Animal Biology, University of Pennsylvania, Philadelphia, PA
| | - Sandra A. Bostic
- Department of Animal Biology, University of Pennsylvania, Philadelphia, PA
| | - Kurt D. Hankenson
- Department of Animal Biology, University of Pennsylvania, Philadelphia, PA,Correspondence to: Kurt D. Hankenson 311 Hill Pavilion 380 S. University Ave. Philadelphia, PA 19104 (T: 215-746-1873; F: 215-573-5187; )
| |
Collapse
|
202
|
Abstract
Over the past two decades there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point, but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, we have focused on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory, together with detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- School of Dentistry/Department of Oral Biology, University of Missouri, Kansas City, MO, USA.
| | | |
Collapse
|
203
|
Yu X, Xia Z, Wang L, Peng F, Jiang X, Huang J, Rowe D, Wei M. Controlling the structural organization of regenerated bone by tailoring tissue engineering scaffold architecture. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm30332a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
204
|
Born AK, Lischer S, Maniura-Weber K. Watching osteogenesis: Life monitoring of osteogenic differentiation using an osteocalcin reporter. J Cell Biochem 2011; 113:313-21. [DOI: 10.1002/jcb.23357] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
205
|
Ohishi M, Ono W, Ono N, Khatri R, Marzia M, Baker EK, Root SH, Wilson TLS, Iwamoto Y, Kronenberg HM, Aguila HL, Purton LE, Schipani E. A novel population of cells expressing both hematopoietic and mesenchymal markers is present in the normal adult bone marrow and is augmented in a murine model of marrow fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:811-8. [PMID: 22155108 DOI: 10.1016/j.ajpath.2011.10.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 09/29/2011] [Accepted: 10/31/2011] [Indexed: 01/13/2023]
Abstract
Bone marrow (BM) fibrosis is a feature of severe hyperparathyroidism. Consistent with this observation, mice expressing constitutively active parathyroid hormone (PTH)/PTH-related peptide receptors (PPR) in osteoblasts (PPR*Tg) display BM fibrosis. To obtain insight into the nature of BM fibrosis in such a model, a double-mutant mouse expressing constitutively active PPR and green fluorescent protein (GFP) under the control of the type I collagen promoter (PPR*Tg/GFP) was generated. Confocal microscopy and flow cytometry revealed the presence of a cell population expressing GFP (GFP(+)) that was also positive for the hematopoietic marker CD45 in the BM of both PPR*Tg/GFP and control animals. This cell population was expanded in PPR*Tg/GFP. The existence of cells expressing both type I collagen and CD45 in the adult BM was confirmed by IHC and fluorescence-activated cell sorting. An analysis of total RNA extracted from sorted GFP(+)CD45(+) cells showed that these cells produced type I collagen and PTH/PTH-related peptide receptor and receptor activator for NF-κB mRNAs, further supporting their features of being both mesenchymal and hematopoietic lineages. Similar cells, known as fibrocytes, are also present in pathological fibroses. Our findings, thus, indicate that the BM is a permissive microenvironment for the differentiation of fibrocyte-like cells and raise the possibility that these cells could contribute to the pathogenesis of BM fibrosis.
Collapse
Affiliation(s)
- Masanobu Ohishi
- Endocrine Unit, the Department of Medicine, Faculty of Medical Sciences, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Elefteriou F, Yang X. Genetic mouse models for bone studies--strengths and limitations. Bone 2011; 49:1242-54. [PMID: 21907838 PMCID: PMC3331798 DOI: 10.1016/j.bone.2011.08.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/15/2011] [Accepted: 08/18/2011] [Indexed: 11/25/2022]
Abstract
Mice have become a preferred model system for bone research because of their genetic and pathophysiological similarities to humans: a relatively short reproductive period, leading to relatively low cost of maintenance and the availability of the entire mouse genome sequence information. The success in producing the first transgenic mouse line that expressed rabbit β-globin protein in mouse erythrocytes three decades ago marked the beginning of the use of genetically engineered mice as model system to study human diseases. Soon afterward the development of cultured pluripotent embryonic stem cells provided the possibility of gene replacement or gene deletion in mice. These technologies have been critical to identify new genes involved in bone development, growth, remodeling, repair, and diseases, but like many other approaches, they have limitations. This review will introduce the approaches that allow the generation of transgenic mice and global or conditional (tissue-specific and inducible) mutant mice. A list of the various promoters used to achieve bone-specific gene deletion or overexpression is included. The limitations of these approaches are discussed, and general guidelines related to the analysis of genetic mouse models are provided.
Collapse
Affiliation(s)
- Florent Elefteriou
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| | - Xiangli Yang
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| |
Collapse
|
207
|
Dvorak-Ewell MM, Chen TH, Liang N, Garvey C, Liu B, Tu C, Chang W, Bikle DD, Shoback DM. Osteoblast extracellular Ca2+ -sensing receptor regulates bone development, mineralization, and turnover. J Bone Miner Res 2011; 26:2935-47. [PMID: 21956637 PMCID: PMC3222747 DOI: 10.1002/jbmr.520] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The extracellular Ca(2+) -sensing receptor (CaR), a G protein-coupled receptor responsible for maintenance of calcium homeostasis, is implicated in regulation of skeletal metabolism. To discern the role of the osteoblast CaR in regulation of bone development and remodeling, we generated mice in which the CaR is excised in a broad population of osteoblasts expressing the 3.6-kb a(1) (I) collagen promoter. Conditional knockouts had abnormal skeletal histology at birth and developed progressively reduced mineralization secondary to retarded osteoblast differentiation, evident by significantly reduced numbers of osteoblasts and decreased expression of collagen I, osteocalcin, and sclerostin mRNAs. Elevated expression of ankylosis protein, ectonucleotide pyrophosphatase/phosphodiesterase 1, and osteopontin mRNAs in the conditional knockout indicate altered regulation of genes important in mineralization. Knockout of the osteoblast CaR also resulted in increased expression of the receptor activator of NF-κB ligand (RANKL), the major stimulator of osteoclast differentiation and function, consistent with elevated osteoclast numbers in vivo. Osteoblasts from the conditional knockouts exhibited delayed differentiation, reduced mineralizing capacity, altered expression of regulators of mineralization, and increased ability to promote osteoclastogenesis in coculture experiments. We conclude that CaR signaling in a broad population of osteoblasts is essential for bone development and remodeling and plays an important role in the regulation of differentiation and expression of regulators of bone resorption and mineralization.
Collapse
Affiliation(s)
- Melita M Dvorak-Ewell
- Endocrine Research Unit, Department of Veterans Affairs Medical Center, Department of Medicine, University of California, San Francisco, CA 94121, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Ferraro F, Lymperi S, Méndez-Ferrer S, Saez B, Spencer JA, Yeap BY, Masselli E, Graiani G, Prezioso L, Rizzini EL, Mangoni M, Rizzoli V, Sykes SM, Lin CP, Frenette PS, Quaini F, Scadden DT. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med 2011; 3:104ra101. [PMID: 21998408 PMCID: PMC3754876 DOI: 10.1126/scitranslmed.3002191] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Success with transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) in patients depends on adequate collection of these cells after mobilization from the bone marrow niche by the cytokine granulocyte colony-stimulating factor (G-CSF). However, some patients fail to achieve sufficient HSPC mobilization. Retrospective analysis of bone marrow transplant patient records revealed that diabetes correlated with poor mobilization of CD34+ HSPCs. In mouse models of type 1 and type 2 diabetes (streptozotocin-induced and db/db mice, respectively), we found impaired egress of murine HSPCs from the bone marrow after G-CSF treatment. Furthermore, HSPCs were aberrantly localized in the marrow niche of the diabetic mice, and abnormalities in the number and function of sympathetic nerve termini were associated with this mislocalization. Aberrant responses to β-adrenergic stimulation of the bone marrow included an inability of marrow mesenchymal stem cells expressing the marker nestin to down-modulate the chemokine CXCL12 in response to G-CSF treatment (mesenchymal stem cells are reported to be critical for HSPC mobilization). The HSPC mobilization defect was rescued by direct pharmacological inhibition of the interaction of CXCL12 with its receptor CXCR4 using the drug AMD3100. These data suggest that there are diabetes-induced changes in bone marrow physiology and microanatomy and point to a potential intervention to overcome poor HSPC mobilization in diabetic patients.
Collapse
Affiliation(s)
- Francesca Ferraro
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Stefania Lymperi
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Simón Méndez-Ferrer
- Cardiovascular Developmental Biology Department. Centro Nacional de Investigaciones Cardiovasculares, 28029 Madrid, Spain
| | - Borja Saez
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Joel A Spencer
- Advanced Microscopy Program, Center for System Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | - Beow Y Yeap
- Department of Medicine Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Elena Masselli
- Department of Hematology and Bone Marrow Transplantation University of Parma, Parma, Italy
| | - Gallia Graiani
- Department of Internal Medicine and Biomedical Science University of Parma, Parma 43100, Italy
| | - Lucia Prezioso
- Department of Internal Medicine and Biomedical Science University of Parma, Parma 43100, Italy
| | - Elisa Lodi Rizzini
- Department of Hematology and Bone Marrow Transplantation University of Parma, Parma, Italy
| | - Marcellina Mangoni
- Department of Hematology and Bone Marrow Transplantation University of Parma, Parma, Italy
| | - Vittorio Rizzoli
- Department of Hematology and Bone Marrow Transplantation University of Parma, Parma, Italy
| | - Stephen M Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Charles P. Lin
- Advanced Microscopy Program, Center for System Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Paul S. Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Federico Quaini
- Department of Internal Medicine and Biomedical Science University of Parma, Parma 43100, Italy
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
209
|
Igwe JC, Gao Q, Kizivat T, Kao WW, Kalajzic I. Keratocan is expressed by osteoblasts and can modulate osteogenic differentiation. Connect Tissue Res 2011; 52:401-7. [PMID: 21405980 PMCID: PMC3574643 DOI: 10.3109/03008207.2010.546536] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Keratocan is an extracellular matrix protein that belongs to the small leucine-rich proteoglycan family that also includes lumican, biglycan, decorin, mimecan, and fibromodulin. Members of this family are known to play a role in regulating cellular processes such as proliferation and modulation of osteoprogenitor lineage differentiation. The aims of this study were to evaluate the expression pattern of the keratocan within the osteoprogenitor lineage and to assess its role in regulating osteoblast maturation and function. Results from gene expression analyses of cells at different maturation stages within the osteoblast lineage indicate that keratocan is differentially expressed by osteoblasts and shows little or no expression by osteocytes. During primary osteoblast cultures, high keratocan mRNA expression was observed on day 14, whereas lower expression was detected at days 7 and 21. To assess the effects of keratocan on osteoprogenitor cell differentiation, we evaluated primary calvarial cell cultures from keratocan-deficient mice. The mineralization of calvarial osteoblast cultures derived from keratocan null (Kera-/-) mice was lower than in wild-type osteoblast cultures. Furthermore, analysis of RNA derived from Kera-/- calvarial cell cultures showed a reduction in the mature osteoblast differentiation markers, that is, bone sialoprotein and osteocalcin. In addition, we have evaluated the bone formation in keratocan-deficient mice. Histomorphometric analysis indicated that homozygous knockout mice have significantly decreased rates of bone formation and mineral apposition. Taken together, our results demonstrate the expression of keratocan by osteoblast lineage cells and its ability to modulate osteoblast function.
Collapse
Affiliation(s)
- John C. Igwe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Qi Gao
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Tomislav Kizivat
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Winston W. Kao
- Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
210
|
Wu JY, Aarnisalo P, Bastepe M, Sinha P, Fulzele K, Selig MK, Chen M, Poulton IJ, Purton LE, Sims NA, Weinstein LS, Kronenberg HM. Gsα enhances commitment of mesenchymal progenitors to the osteoblast lineage but restrains osteoblast differentiation in mice. J Clin Invest 2011; 121:3492-504. [PMID: 21804192 DOI: 10.1172/jci46406] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 06/08/2011] [Indexed: 12/17/2022] Open
Abstract
The heterotrimeric G protein subunit Gsα stimulates cAMP-dependent signaling downstream of G protein-coupled receptors. In this study, we set out to determine the role of Gsα signaling in cells of the early osteoblast lineage in vivo by conditionally deleting Gsα from osterix-expressing cells. This led to severe osteoporosis with fractures at birth, a phenotype that was found to be the consequence of impaired bone formation rather than increased resorption. Osteoblast number was markedly decreased and osteogenic differentiation was accelerated, resulting in the formation of woven bone. Rapid differentiation of mature osteoblasts into matrix-embedded osteocytes likely contributed to depletion of the osteoblast pool. In addition, the number of committed osteoblast progenitors was diminished in both bone marrow stromal cells (BMSCs) and calvarial cells of mutant mice. In the absence of Gsα, expression of sclerostin and dickkopf1 (Dkk1), inhibitors of canonical Wnt signaling, was markedly increased; this was accompanied by reduced Wnt signaling in the osteoblast lineage. In summary, we have shown that Gsα regulates bone formation by at least two distinct mechanisms: facilitating the commitment of mesenchymal progenitors to the osteoblast lineage in association with enhanced Wnt signaling; and restraining the differentiation of committed osteoblasts to enable production of bone of optimal mass, quality, and strength.
Collapse
Affiliation(s)
- Joy Y Wu
- Endocrine Unit, Massachusetts General Hospital, 50 Blossom Street, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Abdallah BM, Ditzel N, Mahmood A, Isa A, Traustadottir GA, Schilling AF, Ruiz-Hidalgo MJ, Laborda J, Amling M, Kassem M. DLK1 is a novel regulator of bone mass that mediates estrogen deficiency-induced bone loss in mice. J Bone Miner Res 2011; 26:1457-71. [PMID: 21308776 DOI: 10.1002/jbmr.346] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Delta-like 1/fetal antigen 1 (DLK1/FA-1) is a transmembrane protein belonging to the Notch/Delta family that acts as a membrane-associated or a soluble protein to regulate regeneration of a number of adult tissues. Here we examined the role of DLK1/FA-1 in bone biology using osteoblast-specific Dlk1-overexpressing mice (Col1-Dlk1). Col1-Dlk1 mice displayed growth retardation and significantly reduced total body weight and bone mineral density (BMD). Micro-computed tomographis (µCT) scanning revealed a reduced trabecular and cortical bone volume fraction. Tissue-level histomorphometric analysis demonstrated decreased bone-formation rate and enhanced bone resorption in Col1-Dlk1 mice compared with wild-type mice. At a cellular level, Dlk1 markedly reduced the total number of bone marrow (BM)-derived colony-forming units fibroblasts (CFU-Fs), as well as their osteogenic capacity. In a number of in vitro culture systems, Dlk1 stimulated osteoclastogenesis indirectly through osteoblast-dependent increased production of proinflammatory bone-resorbing cytokines (eg, Il7, Tnfa, and Ccl3). We found that ovariectomy (ovx)-induced bone loss was associated with increased production of Dlk1 in the bone marrow by activated T cells. Interestingly, Dlk1(-/-) mice were significantly protected from ovx-induced bone loss compared with wild-type mice. Thus we identified Dlk1 as a novel regulator of bone mass that functions to inhibit bone formation and to stimulate bone resorption. Increasing DLK1 production by T cells under estrogen deficiency suggests its possible use as a therapeutic target for preventing postmenopausal bone loss.
Collapse
Affiliation(s)
- Basem M Abdallah
- Endocrine Research Laboratory, KMEB, Department of Endocrinology, and Medical Biotechnlogy Center, Odense University Hospital and University of Southern Denmark, Odense, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
|
213
|
Hall SL, Chen ST, Wergedal JE, Gridley DS, Mohan S, Lau KHW. Stem cell antigen-1 positive cell-based systemic human growth hormone gene transfer strategy increases endosteal bone resorption and bone loss in mice. J Gene Med 2011; 13:77-88. [PMID: 21322098 DOI: 10.1002/jgm.1542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The present study assesses the effect of the stem cell antigen-1 positive (Sca-1(+) ) cell-based human growth hormone (hGH) ex vivo gene transfer strategy on endosteal bone mass in the mouse. METHODS Sublethally irradiated recipient mice were transplanted with Sca-1(+) cells transduced with lentiviral vectors expressing hGH or β-galactosidase control genes. Bone parameters were assessed by micro-computed tomography and histomorphometry. RESULTS This hGH strategy drastically increased hGH mRNA levels in bone marrow cells and serum insulin-like growth factor-I (IGF-I) (by nearly 50%, p < 0.002) in hGH recipient mice. Femoral trabecular bone volume of the hGH mice was significantly reduced by 35% (p < 0.002). The hGH mice also had decreased trabecular number (by 26%; p < 0.0001), increased trabecular separation (by 38%; p < 0.0002) and reduced trabecular connectivity density (by 64%; p < 0.001), as well as significantly more osteoclasts (2.5-fold; p < 0.05) and greater osteoclastic surface per bone surface (2.6-fold; p < 0.01). CONCLUSIONS Targeted expression of hGH in cells of marrow cavity through the Sca-1(+) cell-based gene transfer strategy increased circulating IGF-I and decreased endosteal bone mass through an increase in resorption in recipient mice. These results indicate that high local levels of hGH or IGF-I in the bone marrow microenvironment enhanced resorption, which is consistent with previous findings in transgenic mice with targeted bone IGF-I expression showing that high local IGF-I expression increased bone remodeling, favoring a net bone loss. Thus, GH and/or IGF-I would not be an appropriate transgene for use in this Sca-1(+) cell-based gene transfer strategy to promote endosteal bone formation. Published 2011 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Susan L Hall
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, CA, USA.
| | | | | | | | | | | |
Collapse
|
214
|
Wnt-inhibitory factor 1 dysregulation of the bone marrow niche exhausts hematopoietic stem cells. Blood 2011; 118:2420-9. [PMID: 21652676 DOI: 10.1182/blood-2010-09-305664] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The role of Wnt signaling in hematopoietic stem cell fate decisions remains controversial. We elected to dysregulate Wnt signaling from the perspective of the stem cell niche by expressing the pan Wnt inhibitor, Wnt inhibitory factor 1 (Wif1), specifically in osteoblasts. Here we report that osteoblastic Wif1 overexpression disrupts stem cell quiescence, leading to a loss of self-renewal potential. Primitive stem and progenitor populations were more proliferative and elevated in bone marrow and spleen, manifesting an impaired ability to maintain a self-renewing stem cell pool. Exhaustion of the stem cell pool was apparent only in the context of systemic stress by chemotherapy or transplantation of wild-type stem cells into irradiated Wif1 hosts. Paradoxically this is mediated, at least in part, by an autocrine induction of canonical Wnt signaling in stem cells on sequestration of Wnts in the environment. Additional signaling pathways are dysregulated in this model, primarily activated Sonic Hedgehog signaling in stem cells as a result of Wif1-induced osteoblastic expression of Sonic Hedgehog. We find that dysregulation of the stem cell niche by overexpression of an individual component impacts other unanticipated regulatory pathways in a combinatorial manner, ultimately disrupting niche mediated stem cell fate decisions.
Collapse
|
215
|
Wang M, Jin H, Tang D, Huang S, Zuscik M, Chen D. Smad1 plays an essential role in bone development and postnatal bone formation. Osteoarthritis Cartilage 2011; 19:751-62. [PMID: 21420501 PMCID: PMC3113680 DOI: 10.1016/j.joca.2011.03.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 02/27/2011] [Accepted: 03/11/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To determine the role of Smad1 in bone development and postnatal bone formation. METHODS Col2a1-Cre transgenic mice were bred with Smad1(fx/fx) mice to produce chondrocyte-specific Smad1 conditional knockout (cKO) mice. Embryonic skeletal preparation and staining were performed, alkaline phosphatase activity (ALP) and relative gene expression were examined in isolated primary cells. Smad1(fx/fx) mice were also bred with Col1a1-Cre transgenic mice to produce osteoblast-specific Smad1 cKO mice. Postnatal bone formation was assessed by micro-computed tomography (μCT) and histological analyses in 2-month-old mice. Mineralized bone nodule formation assay, 5-bromo-2'-deoxy-uridine (BrdU) labeling and gene expression analysis were performed. RESULTS Mice with chondrocyte- and osteoblast-specific deletion of the Smad1 gene are viable and fertile. Calvarial bone development was delayed in chondrocyte-specific Smad1 cKO mice. In osteoblast-specific Smad1 cKO mice, BMP signaling was partially inhibited and mice developed an osteopenic phenotype. Osteoblast proliferation and differentiation were impaired in osteoblast-specific Smad1 cKO mice. CONCLUSIONS Smad1 plays an essential role in bone development and postnatal bone formation.
Collapse
Affiliation(s)
- M. Wang
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - H. Jin
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA, Institute of Orthopaedics and Traumatology, Zhejiang Chinese Medical University, Hangzhou, China
| | - D. Tang
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - S. Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - M.J. Zuscik
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - D. Chen
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA,Address correspondence and reprint requests to: D. Chen, Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA. Tel: 1-585-273-5631; Fax: 1-585-275-1121. (D. Chen)
| |
Collapse
|
216
|
Chen J, Utreja A, Kalajzic Z, Sobue T, Rowe D, Wadhwa S. Isolation and characterization of murine mandibular condylar cartilage cell populations. Cells Tissues Organs 2011; 195:232-43. [PMID: 21646777 DOI: 10.1159/000325148] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2011] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES The mandibular condylar cartilage is a heterogeneous tissue containing cells at various stages of chondrocyte maturation organized into 4 zones: superficial, polymorphic, flattened, and hypertrophic. The goal of this study was to use transgenic mice containing chondrocyte maturation markers fused to fluorescent protein transgenes to isolate and characterize homogenous cell populations of the mandibular condylar cartilage. METHODS Fluorescent reporter expression in the mandibular condylar cartilage of transgenic mice containing the 3.6-kb fragment of the rat collagen type 1 promoter fused to a topaz-fluorescent protein (Col3.6-tpz), collagen type 2 promoter fused to a cyan-fluorescent protein (Col2-cyan), and/or collagen type 10 promoter fused to cherry-fluorescent protein (Col10-cherry) was examined. Mandibular condylar cartilage cells were analyzed by fluorescence-activated cell sorting (FACS) and either used for gene expression analysis or plated in cell cultures and exposed to adipogenic, osteogenic, or chondrogenic conditions. To determine cell fate, transgenic mice containing the Col3.6-cre recombinase were bred with cre reporter mice. RESULTS Localization and analysis of gene expression revealed that Col3.6-tpz-positive cells corresponded to the polymorphic/flattened zones and Col2-cyan-positive cells corresponded to the flattened/hypertrophic zones of the mandibular condylar cartilage. Mandibular condylar cartilage FACS-sorted Col3.6-tpz-positive cells have the potential to differentiate into bone, cartilage, and fat. Cell fate mapping revealed that Col3.6 cells are precursors of some of the hypertrophic chondrocytes in the mandibular condylar cartilage. CONCLUSION Col3.6-tpz cells represent an earlier stage of the mandibular condylar cartilage maturation pathway.
Collapse
Affiliation(s)
- J Chen
- Division of Orthodontics, Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | |
Collapse
|
217
|
Maye P, Fu Y, Butler DL, Chokalingam K, Liu Y, Floret J, Stover ML, Wenstrup R, Jiang X, Gooch C, Rowe D. Generation and characterization of Col10a1-mcherry reporter mice. Genesis 2011; 49:410-8. [PMID: 21328521 PMCID: PMC5638041 DOI: 10.1002/dvg.20733] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We report here on the generation of a new fluorescent protein reporter transgenic mouse line, Col10a1-mCherry, which can be used as a tool to study chondrocyte biology and pathology. Collagen, Type X, alpha 1 (Col10a1) is highly expressed in hypertrophic chondrocytes and commonly used as a gene marker for this cell population. The Col10a1-mCherry reporter line was generated using a bacterial recombination strategy with the mouse BAC clone RP23-192A7. To aid in the characterization of this animal model, we intercrossed Col10a1-mCherry mice with Collagen, Type II, alpha 1 (Col2a1) enhanced cyan fluorescent protein (ECFP) reporter mice and characterized the expression of both chondrocyte reporters during embryonic skeletal development from days E10.5 to E17.5. Additionally, at postnatal day 0, Col10a1-mCherry reporter expression was compared to endogenous Col10a1 mRNA expression in long bones and revealed that mCherry fluorescence extended past the Col10a1 expression domain. However, in situ hybridization for mCherry was consistent with the zone of Col10a1 mRNA expression, indicating that the persistent detection of mCherry fluorescence was a result of the long protein half life of mCherry in conjunction with a very rapid phase of skeletal growth and not due to aberrant transcriptional regulation. Taking advantage of the continued fluorescence of hypertrophic chondrocytes at the chondro-osseus junction, we intercrossed Col10a1-mCherry mice with two different Collagen, Type 1, alpha 1, (Col1a1) osteoblast reporter mice, pOBCol3.6-Topaz and pOBCol2.3-Emerald to investigate the possibility that hypertrophic chondrocytes transdifferentiate into osteoblasts. Evaluation of long bones at birth suggests that residual hypertrophic chondrocytes and osteoblasts in the trabecular zone exist as two completely distinct cell populations. genesis 49:410-418, 2011.
Collapse
Affiliation(s)
- Peter Maye
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Uribe F, Kalajzic Z, Bibko J, Nanda R, Olson C, Rowe D, Wadhwa S. Early effects of orthodontic forces on osteoblast differentiation in a novel mouse organ culture model. Angle Orthod 2011; 81:284-91. [PMID: 21208081 DOI: 10.2319/052410-279.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE To develop a mouse orthodontic organ culture model and examine early-induced changes in osteoblast differentiation markers within the periodontal ligament (PDL) and alveolar bone. METHODS Mandibles from 4- to 12-week-old transgenic mice were dissected and hemisected. A conventional superelastic orthodontic spring (25 grams) was bonded to the incisor and first molar on one side of the mandible; the other side served as a control. Dissected mandibles were cultured for 6 hours and then were histologically analyzed for proliferation (BrdU immunostaining) and fluorescent protein expression. Additionally, an in vivo model using the same methods was applied to 3.6 Col1-GFP transgenic mice. RESULTS In vitro, after 6 hours of orthodontic loading, a significant increase was noted in 3.6Col1-GFP- and BSP-GFP-positive cells within the tension side of the PDL compared with unloaded controls. On the compression side, a significant decrease in positive cells in 3.6Col1-GFP mice was observed in the PDL compared with unloaded controls. In vivo, the same tendencies were found. CONCLUSION This novel in vitro mandibular tooth movement organ culture model coupled with transgenic mouse technology provides a powerful tool for delineating initial cellular and molecular events of orthodontic tooth movement.
Collapse
Affiliation(s)
- Flavio Uribe
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, 06032, USA.
| | | | | | | | | | | | | |
Collapse
|
219
|
Gorski JP, Huffman NT, Chittur S, Midura RJ, Black C, Oxford J, Seidah NG. Inhibition of proprotein convertase SKI-1 blocks transcription of key extracellular matrix genes regulating osteoblastic mineralization. J Biol Chem 2011; 286:1836-49. [PMID: 21075843 PMCID: PMC3023479 DOI: 10.1074/jbc.m110.151647] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/22/2010] [Indexed: 11/06/2022] Open
Abstract
Mineralization, a characteristic phenotypic property of osteoblastic lineage cells, was blocked by 4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride (AEBSF) and decanoyl-Arg-Arg-Leu-Leu-chloromethyl ketone (dec-RRLL-cmk), inhibitors of SKI-1 (site 1; subtilisin kexin like-1) protease. Because SKI-1 is required for activation of SREBP and CREB (cAMP-response element-binding protein)/ATF family transcription factors, we tested the effect of these inhibitors on gene expression. AEBSF decreased expression of 140 genes by 1.5-3.0-fold including Phex, Dmp1, COL1A1, COL11A1, and fibronectin. Direct comparison of AEBSF and dec-RRLL-cmk, a more specific SKI-1 inhibitor, demonstrated that expression of Phex, Dmp1, COL11A1, and fibronectin was reduced by both, whereas COL1A2 and HMGCS1 were reduced only by AEBSF. AEBSF and dec-RRLL-cmk decreased the nuclear content of SKI-1-activated forms of transcription factors SREBP-1, SREBP-2, and OASIS. In contrast to AEBSF, the actions of dec-RRLL-cmk represent the sum of its direct actions on SKI-1 and indirect actions on caspase-3. Specifically, dec-RRLL-cmk reduced intracellular caspase-3 activity by blocking the formation of activated 19-kDa caspase-3. Conversely, overexpression of SKI-1-activated SREBP-1a and CREB-H in UMR106-01 osteoblastic cells increased the number of mineralized foci and altered their morphology to yield mineralization nodules, respectively. In summary, SKI-1 regulates the activation of transmembrane transcription factor precursors required for expression of key genes required for mineralization of osteoblastic cultures in vitro and bone formation in vivo. Our results indicate that the differentiated phenotype of osteoblastic cells and possibly osteocytes depends upon the non-apoptotic actions of SKI-1.
Collapse
Affiliation(s)
- Jeff P Gorski
- Center of Excellence in the Study of Musculoskeletal and Dental Tissues and Department of Oral Biology, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA.
| | | | | | | | | | | | | |
Collapse
|
220
|
Chen IP, Wang L, Jiang X, Aguila HL, Reichenberger EJ. A Phe377del mutation in ANK leads to impaired osteoblastogenesis and osteoclastogenesis in a mouse model for craniometaphyseal dysplasia (CMD). Hum Mol Genet 2010; 20:948-61. [PMID: 21149338 DOI: 10.1093/hmg/ddq541] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Craniometaphyseal dysplasia (CMD) is a rare genetic disorder with hyperostosis of craniofacial bones and widened metaphyses in long bones. Patients often suffer from neurological symptoms due to obstruction of cranial foramina. No proven treatment is available and the pathophysiology is largely unknown. A Phe377 (TTC(1130-1132)) deletion in exon 9 of the pyrophosphate (PPi) transporter ANK leads to CMD-like features in an Ank(KI/KI) mouse model. Here, we investigated the effects of CMD-mutant ANK on mineralization and bone mass at a cellular level. Ank(KI/KI) osteoblast cultures showed decreased mineral deposition. Expression of bone mineralization regulating genes Mmp13, Ocn, Osx and Phex was reduced in Ank(KI/KI) osteoblasts, while the Fgf23 mRNA level was highly elevated in Ank(KI/KI) calvarial and femoral bones. Since ANK is a known PPi transporter, we examined other regulators of Pi/PPi homeostasis Enpp1 and Tnap. Significantly increased ENPP1 activity may compensate for dysfunctional mutant ANK leading to comparable extracellular PPi levels in Ank(+/+) osteoblasts. Similar to Ank(KI/KI) bone marrow-derived macrophage cultures, peripheral blood cultures from CMD patients exhibited reduced osteoclastogenesis. Cell-autonomous effects in Ank(KI/KI) osteoclasts resulted in disrupted actin ring formation and cell fusion. In addition, Ank(KI/KI) osteoblasts failed to adequately support osteoclastogenesis. Increased bone mass could partially be rescued by bone marrow transplants supporting our hypothesis that reduced osteoclastogenesis contributes at least in part to hyperostosis. We conclude that the Phe377del mutation in ANK causes impaired osteoblastogenesis and osteoclastogenesis resulting in hypomineralization and a high bone mass phenotype.
Collapse
Affiliation(s)
- I-Ping Chen
- Department of Oral Health and Diagnostic Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | |
Collapse
|
221
|
Zanotti S, Smerdel-Ramoya A, Canalis E. HES1 (hairy and enhancer of split 1) is a determinant of bone mass. J Biol Chem 2010; 286:2648-57. [PMID: 21084301 DOI: 10.1074/jbc.m110.183038] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
HES1 (hairy and enhancer of split) is a transcription factor that regulates osteoblastogenesis in vitro. The skeletal effects of HES1 misexpression were studied. Transgenic mice where a 3.6-kilobase fragment of the collagen type 1 α1 promoter directs HES1 overexpression were created. Transgenics were osteopenic due to decreased osteoblast function in female and increased bone resorption in male mice. HES1 impaired osteoblastogenesis in vitro, and transgenic osteoblasts enhanced the resorptive activity of co-cultured osteoclast precursors. Mice homozygous for a Hes1 loxP-targeted allele were bred to transgenics, where the paired-related homeobox gene enhancer or the osteocalcin promoter direct Cre recombinase expression to inactivate Hes1 in the limb bud or in osteoblasts. To avoid genetic compensation, Hes1 was inactivated in the context of the global deletion of Hes3 and Hes5. Hes3 and Hes5 null mice had no skeletal phenotype. Hes1 inactivation in the limb bud increased femoral length and trabecular number. Hes1 inactivation in osteoblasts increased trabecular bone volume, number, and connectivity due to increased mineral apposition rate and suppressed bone resorption. Hes1 inactivation in vitro increased alkaline phosphatase expression and suppressed the resorptive activity of co-cultured osteoclast precursors. In conclusion, by inhibiting osteoblast function and inducing bone resorption, HES1 is an intracellular determinant of bone mass and structure.
Collapse
Affiliation(s)
- Stefano Zanotti
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut 06105, USA.
| | | | | |
Collapse
|
222
|
Balic A, Aguila HL, Mina M. Identification of cells at early and late stages of polarization during odontoblast differentiation using pOBCol3.6GFP and pOBCol2.3GFP transgenic mice. Bone 2010; 47:948-58. [PMID: 20728593 PMCID: PMC2957651 DOI: 10.1016/j.bone.2010.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 08/12/2010] [Accepted: 08/13/2010] [Indexed: 02/09/2023]
Abstract
Transgenic mouse lines in which GFP expression is under the control of tissue- and stage specific promoters have provided powerful experimental tools for identification and isolation of cells at specific stage of differentiation along a lineage. In the present study, we used primary cell cultures derived from the dental pulp from pOBCol3.6GFP and pOBCol2.3GFP transgenic mice as a model to develop markers for early stages of odontoblast differentiation from progenitor cells. We analyzed the temporal and spatial expression of 2.3-GFP and 3.6-GFP during in vitro mineralization. Using FACS to separate cells based on GFP expression, we obtained relatively homogenous subpopulations of cells and analyzed their dentinogenic potentials and their progression into odontoblasts. Our observations showed that these transgenes were activated before the onset of matrix deposition and in cells at different stages of polarization. The 3.6-GFP transgene was activated in cells in early stages of polarization, whereas the 2.3-GFP transgene was activated at a later stage of polarization just before or at the time of formation of secretory odontoblast.
Collapse
Affiliation(s)
- Anamaria Balic
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT
| | - H. Leonardo Aguila
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT
| | - Mina Mina
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
223
|
Janeway KA, Walkley CR. Modeling human osteosarcoma in the mouse: From bedside to bench. Bone 2010; 47:859-65. [PMID: 20696288 DOI: 10.1016/j.bone.2010.07.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 07/28/2010] [Accepted: 07/30/2010] [Indexed: 01/06/2023]
Abstract
Osteosarcoma (OS) is the most common primary tumour of bone, occurring predominantly in the second decade of life. High-dose cytotoxic chemotherapy and surgical resection have improved prognosis, with long-term survival for patients with localized (non-metastatic) disease approaching 70%. At presentation approximately 20% of patients have metastases and almost all patients with recurrent OS have metastatic disease and cure rates for patients with metastatic or recurrent disease remain poor (<20% survival). Over the past 20 years, considerable progress has been made in the understanding of OS pathogenesis, yet these insights have not translated into substantial therapeutic advances and clinical outcomes. Further progress is essential in order to develop molecularly based therapies that target both primary lesions as well as metastatic disease. The increasing sophistication with which gene expression can be modulated in the mouse, both positively and negatively in addition to temporally, has allowed for the recent generation of more faithful OS models than have previously been available. These murine OS models can recapitulate all aspects of the disease process, from initiation and establishment to invasion and dissemination to distant sites. The development and utilisation of murine models that faithfully recapitulate human osteosarcoma, complementing existing approaches using human and canine disease, holds significant promise in furthering our understanding of the genetic basis of the disease and, more critically, in advancing pre-clinical studies aimed at the rational development and trialing of new therapeutic approaches.
Collapse
Affiliation(s)
- Katherine A Janeway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Division of Hematology/Oncology, Children's Hospital Boston, Harvard Medical School, 44 Binney St, Boston, MA 02115, USA.
| | | |
Collapse
|
224
|
Fusby JS, Kassmeier MD, Palmer VL, Perry GA, Anderson DK, Hackfort BT, Alvarez GK, Cullen DM, Akhter MP, Swanson PC. Cigarette smoke-induced effects on bone marrow B-cell subsets and CD4+:CD8+ T-cell ratios are reversed by smoking cessation: influence of bone mass on immune cell response to and recovery from smoke exposure. Inhal Toxicol 2010; 22:785-96. [PMID: 20482464 DOI: 10.3109/08958378.2010.483258] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Cigarette smoking adversely affects the immune system, and is a risk factor for developing osteoporosis. How smoking contributes to osteoporosis is unclear, but since lymphocytes help maintain bone homeostasis and lymphocyte depletion results in bone loss, one potential mechanism for how smoke exposure promotes osteoporosis is by reducing bone marrow lymphocytes. Since the risk for developing osteoporosis is reportedly greater in smokers with polymorphisms in LRP5, a gene involved in canonical Wnt signaling that regulates bone metabolism, smoking-induced effects on lymphocytes may be influenced by Lrp5 functionality. To test these possibilities, we examined how the duration and cessation of cigarette smoke exposure affects lymphocyte distribution and function in normal mice and mice predisposed to low or high bone mass due to disruption or mutation of Lrp5. We find that, independent of genotype, mice exposed to cigarette smoke for 3-12 weeks showed a significant reduction in bone marrow B220(+)CD43(-) B cells and splenic transitional T1 B cells, and exhibited a splenic CD4(+):CD8(+) T-cell ratio that was skewed toward CD8(+) T cells. Smoke exposure had little or no effect on other lymphocyte subsets or on lymphocyte function ex vivo. Interestingly, these differences were no longer apparent after 6 weeks without smoke exposure, except in mice with high bone mass where bone marrow B220(+)CD43(-) B cells failed to fully recover. These data provide the first evidence that smoke exposure reduces bone marrow B cells, providing a plausible mechanism for how smoking contributes to osteoporosis.
Collapse
Affiliation(s)
- Jenny S Fusby
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska 68178, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Yang M, Trettel LB, Adams DJ, Harrison JR, Canalis E, Kream BE. Col3.6-HSD2 transgenic mice: a glucocorticoid loss-of-function model spanning early and late osteoblast differentiation. Bone 2010; 47:573-82. [PMID: 20541046 PMCID: PMC2926146 DOI: 10.1016/j.bone.2010.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 05/26/2010] [Accepted: 06/01/2010] [Indexed: 01/08/2023]
Abstract
The goal of this study was to characterize the bone phenotype and molecular alterations in Col3.6-HSD2 mice in which a 3.6-kb Col1a1 promoter fragment drives 11beta-HSD2 expression broadly in the osteoblast lineage to reduce glucocorticoid signaling. Serum corticosterone was unchanged in transgenic females excluding a systemic effect of the transgene. Adult transgenic mice showed reduced vertebral trabecular bone volume and reduced femoral and tibial sub-periosteal and sub-endosteal areas as assessed by microCT. In adult female transgenic mice, histomorphometry showed that vertebral bone mass and trabecular number were reduced but that osteoblast and osteoclast numbers and the mineral apposition and bone formation rates were not changed, suggesting a possible developmental defect in the formation of trabeculae. In a small sample of male mice, osteoblast number and percent osteoid surface were increased but the mineral apposition bone formation rates were not changed, indicating subtle sex-specific phenotypic differences in Col3.6-HSD2 bone. Serum from transgenic mice had decreased levels of the C-terminal telopeptide of alpha1(I) collagen but increased levels of osteocalcin. Transgenic calvarial osteoblast and bone marrow stromal cultures showed decreased alkaline phosphatase and mineral staining, reduced levels of Col1a1, bone sialoprotein and osteocalcin mRNA expression, and decreased cell growth and proliferation. Transgenic bone marrow cultures treated with RANKL and M-CSF showed greater osteoclast formation; however, osteoclast activity as assessed by resorption of a calcium phosphate substrate was decreased in transgenic cultures. Gene profiling of cultured calvarial osteoblasts enriched in the Col3.6-HSD2 transgene showed modest but significant changes in gene expression, particularly in cell cycle and integrin genes. In summary, Col3.6-HSD2 mice showed a low bone mass phenotype, with decreased ex vivo osteogenesis. These data further strengthen the concept that endogenous glucocorticoid signaling is required for optimal bone mass acquisition and highlight the complexities of glucocorticoid signaling in bone cell lineages.
Collapse
Affiliation(s)
- Maobin Yang
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Lorin B. Trettel
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Douglas J. Adams
- Department of Orthopaedic Surgery, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - John R. Harrison
- Department of Craniofacial Sciences, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, 114 Woodland, Street Hartford, Hartford, CT 06105-1299
| | - Barbara E. Kream
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA, and Department of Research, Saint Francis Hospital and Medical Center, 114 Woodland, Street Hartford, Hartford, CT 06105-1299
| |
Collapse
|
226
|
Boban I, Barisic-Dujmovic T, Clark SH. Parabiosis model does not show presence of circulating osteoprogenitor cells. Genesis 2010; 48:171-82. [PMID: 20127800 DOI: 10.1002/dvg.20602] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The goal of this study was to determine the presence of osteoprogenitor cells in the peripheral blood. Experiments were conducted with a parabiosis model in which osteoblast specific transgenic mice (Col2.3GFP or hOC-GFP) were surgically joined with a transgenic mouse where herpes virus thymidine kinase gene is under the control of the collagen alpha1 promoter (Col2.3DeltaTK). This method permits conditional ablation of osteoblasts by ganciclovir (GCV) treatment. In parabionts treated with GCV for 15 days or 1.5-2 months, GFP (hOC-GFP or Col2.3GFP) expression was not detected in histological preparations or in marrow stromal cell cultures from the Col2.3DeltaTK parabiont. Finally, Col2.3GFP/Col2.3DeltaTK pairs were treated with GCV for 15 days and allowed to recover from GCV for 3 months. Again there was a failure to detect Col2.3GFP expressing cells in the Col2.3DeltaTK parabiont. These observations, at least within the limits of this model system, allow the conclusion that osteoprogenitor cells do not readily circulate.
Collapse
Affiliation(s)
- Ivana Boban
- Department of Genetics and Developmental Biology, University of Connecticut Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
227
|
Kozloff KM, Volakis LI, Marini JC, Caird MS. Near-infrared fluorescent probe traces bisphosphonate delivery and retention in vivo. J Bone Miner Res 2010; 25:1748-58. [PMID: 20200982 DOI: 10.1002/jbmr.66] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Bisphosphonate use has expanded beyond traditional applications to include treatment of a variety of low-bone-mass conditions. Complications associated with long-term bisphosphonate treatment have been noted, generating a critical need for information describing the local bisphosphonate-cell interactions responsible for these observations. This study demonstrates that a fluorescent bisphosphonate analogue, far-red fluorescent pamidronate (FRFP), is an accurate biomarker of bisphosphonate deposition and retention in vivo and can be used to monitor site-specific local drug concentration. In vitro, FRFP is competitively inhibited from the surface of homogenized rat cortical bone by traditional bisphosphonates. In vivo, FRFP delivery to the skeleton is rapid, with fluorescence linearly correlated with bone surface area. Limb fluorescence increases linearly with injected dose of FRFP; injected FRFP does not interfere with binding of standard bisphosphonates at the doses used in this study. Long-term FRFP retention studies demonstrated that FRFP fluorescence decreases in conditions of normal bone turnover, whereas fluorescence was retained in conditions of reduced bone turnover, demonstrating preservation of local FRFP concentration. In the mandible, FRFP localized to the alveolar bone and bone surrounding the periodontal ligament and molar roots, consistent with findings of osteonecrosis of the jaw. These findings support a role for FRFP as an effective in vivo marker for bisphosphonate site-specific deposition, turnover, and long-term retention in the skeleton.
Collapse
Affiliation(s)
- Kenneth M Kozloff
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| | | | | | | |
Collapse
|
228
|
Porphyromonas gingivalis lipids inhibit osteoblastic differentiation and function. Infect Immun 2010; 78:3726-35. [PMID: 20584977 DOI: 10.1128/iai.00225-10] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Porphyromonas gingivalis produces unusual sphingolipids that are known to promote inflammatory reactions in gingival fibroblasts and Toll-like receptor 2 (TLR2)-dependent secretion of interleukin-6 from dendritic cells. The aim of the present study was to examine whether P. gingivalis lipids inhibit osteoblastic function. Total lipids from P. gingivalis and two fractions, phosphoglycerol dihydroceramides and phosphoethanolamine dihydroceramides, were prepared free of lipid A. Primary calvarial osteoblast cultures derived from 5- to 7-day-old CD-1 mice were used to examine the effects of P. gingivalis lipids on mineralized nodule formation, cell viability, apoptosis, cell proliferation, and gene expression. P. gingivalis lipids inhibited osteoblast differentiation and fluorescence expression of pOBCol2.3GFP in a concentration-dependent manner. However, P. gingivalis lipids did not significantly alter osteoblast proliferation, viability, or apoptosis. When administered during specific intervals of osteoblast growth, P. gingivalis total lipids demonstrated inhibitory effects on osteoblast differentiation only after the proliferation stage of culture. Reverse transcription-PCR confirmed the downregulation of osteoblast marker genes, including Runx2, ALP, OC, BSP, OPG, and DMP-1, with concurrent upregulation of RANKL, tumor necrosis factor alpha, and MMP-3 genes. P. gingivalis total lipids and lipid fractions inhibited calvarial osteoblast gene expression and function in vivo, as determined by the loss of expression of another osteoblast differentiation reporter, pOBCol3.6GFPcyan, and reduced uptake of Alizarin complexone stain. Finally, lipid inhibition of mineral nodule formation in vitro was dependent on TLR2 expression. Our results indicate that inhibition of osteoblast function and gene expression by P. gingivalis lipids represents a novel mechanism for altering alveolar bone homeostasis at periodontal disease sites.
Collapse
|
229
|
Goldberg AJ, Liu Y, Advincula MC, Gronowicz G, Habibovic P, Kuhn LT. Fabrication and characterization of hydroxyapatite-coated polystyrene disks for use in osteoprogenitor cell culture. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2010; 21:1371-87. [PMID: 20534191 DOI: 10.1163/092050609x12517190417830] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A simple method is reported for fabricating polystyrene disk inserts coated with biomimetic carbonated hydroxyapatite (cHA) to be used for culturing osteoprogenitor cells or other stem cells. Roughened disks cut from tissue-culture polystyrene (TCPS) were coated in simulated body fluid with 5 x normal physiologic ionic concentrations (SBFx5) by a 2-step, 2-day method. The coatings were rigorously characterized by various methods and assessed in cell culture. An adherent, nearly 10 mm thick, relatively uniform layer of single-phase cHA was formed in two days. MC3T3-E1 and mouse calvaria-derived osteoprogenitor cells (pCOBs) were cultured on the cHA for various time points. Despite less initial attachment of both cell types to the cHA, proliferation rates on cHA were similar to that on TCPS. Two-fold greater cell attachment (P < 0.05) of the MC3T3-E1 cells was observed relative to the pCOBs, on both the TCPS and the cHA. Importantly, the coatings were relatively smooth, without the extensive agglomerates observed in other studies and remained adherent and morphologically unchanged after 21 days of culture. This technique can be used to rapidly produce high-quality cHA-coated TCPS disks for cell-culture studies.
Collapse
Affiliation(s)
- A Jon Goldberg
- Center for Biomaterials, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | | | | | | | | |
Collapse
|
230
|
Balic A, Aguila HL, Caimano MJ, Francone VP, Mina M. Characterization of stem and progenitor cells in the dental pulp of erupted and unerupted murine molars. Bone 2010; 46:1639-51. [PMID: 20193787 PMCID: PMC2881695 DOI: 10.1016/j.bone.2010.02.019] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 02/12/2010] [Accepted: 02/18/2010] [Indexed: 12/15/2022]
Abstract
In the past few years there have been significant advances in the identification of putative stem cells also referred to as "mesenchymal stem cells" (MSC) in dental tissues including the dental pulp. It is thought that MSC in dental pulp share certain similarities with MSC isolated from other tissues. However, cells in dental pulp are still poorly characterized. This study focused on the characterization of progenitor and stem cells in dental pulps of erupted and unerupted mice molars. Our study showed that dental pulps from unerupted molars contain a significant number of cells expressing CD90+/CD45-, CD117+/CD45-, Sca-1+/CD45- and little if any CD45+ cells. Our in vitro functional studies showed that dental pulp cells from unerupted molars displayed extensive osteo-dentinogenic potential but were unable to differentiate into chondrocytes and adipocytes. Dental pulps from erupted molars displayed a reduced number of cells, contained a higher percentage of CD45+ and a lower percentage of cells expressing CD90+/CD45-, CD117+/CD45- as compared to unerupted molars. In vitro functional assays demonstrated the ability of a small fraction of cells to differentiate into odontoblasts, osteoblasts, adipocytes and chondrocytes. There was a significant reduction in the osteo-dentinogenic potential of the pulp cells derived from erupted molars compared to unerupted molars. Furthermore, the adipogenic and chondrogenic differentiation of pulp cells from erupted molars was dependent on a long induction period and were infrequent. Based on these findings we propose that the dental pulp of the erupted molars contain a small population of multipotent cells, whereas the dental pulp of the unerupted molars does not contain multipotent cells but is enriched in osteo-dentinogenic progenitors engaged in the formation of coronal and radicular odontoblasts.
Collapse
Affiliation(s)
- Anamaria Balic
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT
| | - H. Leonardo Aguila
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT
| | - Melissa J. Caimano
- Department of Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT
| | - Victor P. Francone
- Department of Neuroscience, School of Medicine, University of Connecticut Health Center, Farmington, CT
| | - Mina Mina
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
231
|
Hong D, Chen HX, Yu HQ, Liang Y, Wang C, Lian QQ, Deng HT, Ge RS. Morphological and proteomic analysis of early stage of osteoblast differentiation in osteoblastic progenitor cells. Exp Cell Res 2010; 316:2291-300. [PMID: 20483354 DOI: 10.1016/j.yexcr.2010.05.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 04/30/2010] [Accepted: 05/12/2010] [Indexed: 01/28/2023]
Abstract
Bone remodeling relies on a dynamic balance between bone formation and resorption, mediated by osteoblasts and osteoclasts, respectively. Under certain stimuli, osteoprogenitor cells may differentiate into premature osteoblasts and further into mature osteoblasts. This process is marked by increased alkaline phosphatase (ALP) activity and mineralized nodule formation. In this study, we induced osteoblast differentiation in mouse osteoprogenitor MC3T3-E1 cells and divided the process into three stages. In the first stage (day 3), the MC3T3-E1 cell under osteoblast differentiation did not express ALP or deposit a mineralized nodule. In the second stage, the MC3T3-E1 cell expressed ALP but did not form a mineralized nodule. In the third stage, the MC3T3-E1 cell had ALP activity and formed mineralized nodules. In the present study, we focused on morphological and proteomic changes of MC3T3-E1 cells in the early stage of osteoblast differentiation - a period when premature osteoblasts transform into mature osteoblasts. We found that mean cell area and mean stress fiber density were increased in this stage due to enhanced cell spreading and decreased cell proliferation. We further analyzed the proteins in the signaling pathway of regulation of the cytoskeleton using a proteomic approach and found upregulation of IQGAP1, gelsolin, moesin, radixin, and Cfl1. After analyzing the focal adhesion signaling pathway, we found the upregulation of FLNA, LAMA1, LAMA5, COL1A1, COL3A1, COL4A6, and COL5A2 as well as the downregulation of COL4A1, COL4A2, and COL4A4. In conclusion, the signaling pathway of regulation of the cytoskeleton and focal adhesion play critical roles in regulating cell spreading and actin skeleton formation in the early stage of osteoblast differentiation.
Collapse
Affiliation(s)
- Dun Hong
- Population Council, 1230 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
232
|
Abstract
Osteocytes are derived from osteoblasts and make up over 90% of the cells in bone. However, the mechanisms that control the differentiation of osteoblasts into osteocytes embedded in bone matrix are not well understood. With the recent developments of transgenic models for manipulating gene expression in osteocytes and of transgenic mice carrying lineage reporters for osteoblasts and osteocytes, unprecedented new insights are becoming possible. In this article we review recent advances, such as comparative gene and protein expression studies, that are delineating the changes in gene and protein expression that accompany osteocyte differentiation. We also review recent studies in which time-lapse dynamic imaging approaches have been used to visualize osteoblast and osteocyte populations within bone. These approaches reveal the key role of cell motility in bone cell function and highlight the dynamic nature of mineralized tissues. Changes in motile properties of the cell may be key in the transition from osteoblast to osteocyte, as reflected in the altered expression of many molecules involved in cytoskeletal function.
Collapse
Affiliation(s)
- Sarah L Dallas
- Department of Oral Biology, School of Dentistry, University of Missouri, Kansas City, Missouri, USA.
| | | |
Collapse
|
233
|
Kuhn LT, Liu Y, Advincula M, Wang YH, Maye P, Goldberg AJ. A nondestructive method for evaluating in vitro osteoblast differentiation on biomaterials using osteoblast-specific fluorescence. Tissue Eng Part C Methods 2010; 16:1357-66. [PMID: 20337515 DOI: 10.1089/ten.tec.2009.0701] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Transgenic mice with a Col1a1-promoter-driven transgene pOBCol2.3GFP were previously developed to visually identify mature osteoblasts through fluorescent expression. Our goal was to determine if this technology could be used to nondestructively evaluate the in vitro differentiation of osteoprogenitor cells on biomaterials such as biomimetic carbonated hydroxyapatite (cHA). Primary osteoprogenitor cells were harvested from calvaria of neonatal Col2.3GFP transgenic mice and cultured on cHA and a tissue culture polystyrene (TCPS) control. The distribution of intensities and area percentage of green fluorescent protein (GFP)-positive cells were quantified using fluorimetry and image analysis of fluorescent microscopy. At 14 days, an increased area and higher mean intensity of GFP-positive cells was observed on cHA as compared to TCPS, indicating more rapid differentiation on cHA. Notably, there were large continuous regions of GFP-positive osteoblasts on cHA, in contrast to the sparse, nodules of osteoblasts on TCPS, implying that cHA provides an osteogenic cue to cells. Xylenol orange staining was capable of distinguishing osteoblast-initiated mineral from the cHA substrate. With this method the unique pattern of osteoblast differentiation on cHA was clearly observed for the first time. Importantly, the generalized method can be used for rapid, high-throughput, nondestructive screening of biomaterials intended to enhance osteogenic differentiation.
Collapse
Affiliation(s)
- Liisa T Kuhn
- Center for Biomaterials, University of Connecticut Health Center, Farmington, Connecticut 06030-1615, USA.
| | | | | | | | | | | |
Collapse
|
234
|
Wang CJ, Chen IP, Koczon-Jaremko B, Boskey AL, Ueki Y, Kuhn L, Reichenberger EJ. Pro416Arg cherubism mutation in Sh3bp2 knock-in mice affects osteoblasts and alters bone mineral and matrix properties. Bone 2010; 46:1306-15. [PMID: 20117257 PMCID: PMC2854251 DOI: 10.1016/j.bone.2010.01.380] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 01/15/2010] [Accepted: 01/21/2010] [Indexed: 11/22/2022]
Abstract
Cherubism is an autosomal dominant disorder in children characterized by unwarranted symmetrical bone resorption of the jaws with fibrous tissue deposition. Mutations causing cherubism have been identified in the adaptor protein SH3BP2. Knock-in mice with a Pro416Arg mutation in Sh3bp2 exhibit a generalized osteoporotic bone phenotype. In this study, we examined the effects of this "cherubism" mutation on spectroscopic indices of "bone quality" and on osteoblast differentiation. Fourier-transform infrared imaging (FTIRI) analysis of femurs from wild-type and Sh3bp2 knock-in mice showed decreased mineral content, decreased mineral crystallinity/crystal size, and increased collagen maturity in homozygous mutants. To assess osteoblast maturation in vivo, knock-in mice were crossed with transgenic mice over-expressing GFP driven by 3.6-kb or 2.3-kb Col1a1 promoter fragments. Reduced numbers of mature osteoblasts were observed in homozygous mice. Neonatal calvarial cultures, which were enriched for osteoblasts by depletion of hematopoietic cells (negative selection for Ter119- and CD45-positive cells) were investigated for osteoblast-specific gene expression and differentiation, which demonstrated that differentiation and mineralization in homozygous osteoblast cultures was impaired. Co-cultures with calvarial osteoblasts and bone marrow macrophages showed that mutant osteoblasts appear to increase osteoclastogenesis resulting in increased bone resorption on bone chips. In summary, the Sh3bp2 mutation in cherubism mice alters bone quality, reduces osteoblast function, and may contribute to excessive bone resorption by osteoclasts. Our data, together with previous osteoclast studies, demonstrate a critical role of Sh3bp2 in bone remodeling and osteoblast differentiation.
Collapse
Affiliation(s)
- Chiachien J Wang
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | | | | | | | | | | | | |
Collapse
|
235
|
Li H, Jiang X, Delaney J, Franceschetti T, Bilic-Curcic I, Kalinovsky J, Lorenzo JA, Grcevic D, Rowe DW, Kalajzic I. Immature osteoblast lineage cells increase osteoclastogenesis in osteogenesis imperfecta murine. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2405-13. [PMID: 20348238 DOI: 10.2353/ajpath.2010.090704] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
This study addressed the role of impairment of osteoblastic differentiation as a mechanism underlying pathophysiology of the osteogenesis imperfecta (OI). We hypothesized that combination of impaired osteogenic differentiation with increased bone resorption leads to diminished bone mass. By introducing visual markers of distinct stages of osteoblast differentiation, pOBCol3.6GFP (3.6GFP; preosteoblast) and pOBCol2.3GFP (2.3GFP; osteoblast/osteocytes), into the OIM model, we assessed osteoblast maturation and the mechanism of increased osteoclastogenesis. Cultures from oim/oim;2.3GFP mice showed a marked reduction of cells expressing GFP relative to +/+;2.3GFP littermates. No significant difference in expression of 3.6GFP between the +/+ and oim/oim mice was observed. Histological analysis of the oim/oim;3.6GFP mice showed an increased area of GFP-positive cells lining the endocortical surface compared with +/+;3.6GFP mice. In contrast GFP expression was similar between oim/oim;2.3GFP and +/+;2.3GFP mice. These data indicate that the osteoblastic lineage is under continuous stimulation; however, only a proportion of cells attain the mature osteoblast stage. Indeed, immature osteoblasts exhibit a stronger potential to support osteoclast formation and differentiation. We detected a higher Rankl/Opg ratio and higher expression of TNF-alpha in sorted immature osteoblasts. In addition, increased osteoclast formation was observed when osteoclast progenitors were cocultured with oim/oim-derived osteoblasts compared with osteoblasts derived from +/+ mice. Taken together, our data indicate that osteoblast lineage maturation is a critical aspect underlying the pathophysiology of OI.
Collapse
Affiliation(s)
- Haitao Li
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Liu J, Arends R, Martens A, den Brok-Bardoel MHC, Scheepers MGH, van Blitterswijk CA, de Boer J. Noninvasive imaging of bone-specific collagen I expression in a luciferase transgenic mouse model. Tissue Eng Part C Methods 2010; 16:1297-304. [PMID: 20218816 DOI: 10.1089/ten.tec.2009.0594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Luciferase transgenic mice are a very promising tool for noninvasive, quantitative, and longitudinal evaluation of gene expression. The aim of this study was to validate the Col(I)-Luc transgenic mouse model in which the luciferase gene is driven by bone-specific regulatory elements from the mouse collagen α1(I) gene for bioluminescent imaging of bone development and remodeling. We observed strong luciferase activity in skeletal tissues of Col(I)-Luc mice, and observed that the light intensity declined with postnatal bone development. Luciferase activity was enhanced in a tail bone repair model and we were able to monitor the process of ectopic bone formation induced by recombinant human bone morphogenetic protein 2 using bioluminescent imaging. We conclude that Col(I)-Luc transgenic mice can be applied in the field of bone tissue engineering for monitoring bone repair processes and for investigating osteoinductive molecules or scaffolds.
Collapse
Affiliation(s)
- Jun Liu
- Department of Tissue Regeneration, MIRA Research Institute, University of Twente, Enschede, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
237
|
Weber AJ, Li G, Kalak R, Street J, Buttgereit F, Dunstan CR, Seibel MJ, Zhou H. Osteoblast-targeted disruption of glucocorticoid signalling does not delay intramembranous bone healing. Steroids 2010; 75:282-6. [PMID: 20096296 DOI: 10.1016/j.steroids.2010.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 01/06/2010] [Accepted: 01/07/2010] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Glucocorticoids at pharmacological doses have been shown to interfere with fracture repair. The role of endogenous glucocorticoids in fracture healing is not well understood. We examined whether endogenous glucocorticoids affect bone healing in an in vivo model of cortical defect repair. METHODS Experiments were performed using a well characterised mouse model in which intracellular glucocorticoid signalling was disrupted in osteoblasts through transgenic overexpression of 11beta-hydroxysteroid-dehydrogenase type 2 (11beta-HSD2) under the control of a collagen type I promoter (Col2.3-11beta-HSD2). Unicortical bone defects (ø 0.8mm) were created in the tibiae of 7-week-old male transgenic mice and their wild-type littermates. Repair was assessed via histomorphometry, immunohistochemistry and microcomputed tomography (micro-CT) analysis at 1-3 weeks after defect creation. RESULTS At week 1, micro-CT images of the defect demonstrated formation of mineralized intramembranous bone which increased in volume and density by week 2. At week 3, healing of the defect was nearly complete in all animals. Analysis by histomorphometry and micro-CT revealed that repair of the bony defect was similar in Col2.3-11beta-HSD2 transgenic animals and their wild-type littermates at all time-points. CONCLUSION Disrupting endogenous glucocorticoid signalling in mature osteoblasts did not affect intramembranous fracture healing in a tibia defect repair model. It remains to be shown whether glucocorticoid signalling has a role in endochondral fracture healing.
Collapse
Affiliation(s)
- Agnes J Weber
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Hospital Road, Concord NSW 2139, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
238
|
Wiren KM, Semirale AA, Hashimoto JG, Zhang XW. Signaling pathways implicated in androgen regulation of endocortical bone. Bone 2010; 46:710-23. [PMID: 19895913 PMCID: PMC2823843 DOI: 10.1016/j.bone.2009.10.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 10/26/2009] [Accepted: 10/28/2009] [Indexed: 01/18/2023]
Abstract
Periosteal expansion is a recognized response to androgen exposure during bone development and in profoundly hypogonadal adults. However, androgen also suppresses endocortical bone formation, indicating that its effects on bone are dichotomous and envelope-specific. In fact, enhanced androgen signaling has been shown to have dramatic detrimental effects on whole bone biomechanical properties in two different transgenic models with skeletally targeted androgen receptor (AR) overexpression. As the mechanisms underlying this response are uncharacterized, we compared patterns of gene expression in periosteum-free cortical bone samples derived from AR-overexpressing transgenic male mice and their wild-type counterparts. We then assessed direct androgen effects in both wild-type and AR-overexpressing osteoblasts in primary culture. Among major signaling pathways associated with bone formation, focused quantitative RT-PCR (qPCR) array-based analysis of endocortical bone gene expression from wild-type vs. transgenic males identified the transforming growth factor-beta (TGF-beta) superfamily and bone morphogenetic protein (BMP) signaling as significantly altered by androgen in vivo. Bioinformatic analyses indicated proliferation, osteoblast differentiation and mineralization as major biological processes affected. Consistent with the in vivo array data and bioinformatic analyses, inhibition of differentiation observed with androgen exposure was reduced by exogenous BMP2 treatment of AR-overexpressing cultures to stimulate BMP signaling, confirming array pathway analysis. In addition, nonaromatizable dihydrotestosterone (DHT) inhibited osteoblast proliferation, differentiation and several indices of mineralization, including mineral accumulation and mineralized nodule formation in primary cultures from both wild-type and AR-transgenic mice. These findings identify a molecular mechanism based on altered BMP signaling that contributes to androgen inhibition of osteoblast differentiation and mineralization. Such detrimental effects of androgen on osteoblast function may underlie the generally disappointing results of androgen therapy.
Collapse
Affiliation(s)
- Kristine M Wiren
- Bone and Mineral Research Unit, Portland Veterans Affairs Medical Center, USA.
| | | | | | | |
Collapse
|
239
|
Barisic-Dujmovic T, Boban I, Clark SH. Fibroblasts/myofibroblasts that participate in cutaneous wound healing are not derived from circulating progenitor cells. J Cell Physiol 2010; 222:703-12. [PMID: 20020505 DOI: 10.1002/jcp.21997] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Dermal fibroblasts/myofibroblasts involved in the wound healing are thought to originate from the resident fibroblast progenitors. To test the hypothesis of an extra dermal origin of the dermal fibroblasts/myofibroblasts, bone marrow (BM) transplantation and parabiosis experiments were initiated utilizing a collagen promoter green fluorescent protein (GFP) reporter transgene as a visible marker for dermal fibroblasts/myofibroblasts. BM transplantation experiments using BM from Col3.6GFPsapph transgenic mice showed no evidence that BM derived progenitors differentiated into dermal fibroblasts/myofibroblasts at the wound site. Rather the GFP positive cells (GFP+) observed at the wound site were not dermal fibroblasts/myofibroblasts but immune cells. These GFP+ cells were also detected in the lung and spleen. Furthermore, GFP+ fibroblasts were not detected in primary dermal fibroblast cultures initiated from BM chimeras. Using the same transgenic mice, parabiotic pairs were generated. One partner in the parabiosis carried a GFP expressing transgene while the other partner was a non-transgenic C57BL/6 mouse. Similar to the BM transplantation experiments, GFP+ immune cells were detected in the wound of the non-transgenic parabiont, however, GFP expressing dermal fibroblasts/myofibroblasts were not observed. Collectively, these data suggest that dermal fibroblast/myofibroblast progenitors do not readily circulate. The expression of the Col3.6GFPsapph in the hematopoietic cells confirmed that our methods were sensitive enough to detect Col3.6GFP expressing dermal fibroblasts derived from the peripheral circulation if they had originated in the BM.
Collapse
Affiliation(s)
- Tatjana Barisic-Dujmovic
- Department of Genetics and Developmental Biology, University of Connecticut Stem Cell Institute, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | |
Collapse
|
240
|
Chung HJ, Steplewski A, Uitto J, Fertala A. Fluorescent protein markers to tag collagenous proteins: the paradigm of procollagen VII. Biochem Biophys Res Commun 2009; 390:662-6. [PMID: 19822129 PMCID: PMC2796180 DOI: 10.1016/j.bbrc.2009.10.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Accepted: 10/06/2009] [Indexed: 10/20/2022]
Abstract
Fluorescent proteins are powerful markers allowing tracking expression, intracellular localization, and translocation of tagged proteins but their effects on the structure and assembly of complex extracellular matrix proteins has not been investigated. Here, we analyzed the utility of fluorescent proteins as markers for procollagen VII, a triple-helical protein critical for the integrity of dermal-epidermal junction. DNA constructs encoding a red fluorescent protein-tagged wild type mini-procollagen VII alpha chain and green fluorescent protein-tagged alpha chains harboring selected mutations were genetically engineered. These DNA constructs were co-expressed in HEK-293 cells and the assembly of heterogeneous triple-helical mini-procollagen VII molecules was analyzed. Immunoprecipitation and fluorescence resonance energy transfer assays demonstrated that the presence of different fluorescent protein markers at the C-termini of individual alpha chains neither altered formation of triple-helical molecules nor affected their secretion to the extracellular space. Our study provides a basis for employing fluorescent proteins as tags for complex structural proteins of extracellular matrix.
Collapse
Affiliation(s)
- Hye Jin Chung
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107, U.S.A
| | - Andrzej Steplewski
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107, U.S.A
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107, U.S.A
| | - Andrzej Fertala
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107, U.S.A
| |
Collapse
|
241
|
Abstract
Notch receptors are transmembrane receptors that regulate cell fate decisions. There are four Notch receptors in mammals. Upon binding to members of the Delta and Jagged family of transmembrane proteins, Notch is cleaved and the Notch intracellular domain (NICD) is released. NICD then translocates to the nucleus, where it associates with the CBF-1, Suppressor of Hairless, and Lag-2 (CSL) and Mastermind-Like (MAML) proteins. This complex activates the transcription of Notch target genes, such as Hairy Enhancer of Split (Hes) and Hes-related with YRPF motif (Hey). Notch signaling is critical for the regulation of mesenchymal stem cell differentiation. Misexpression of Notch in skeletal tissue indicates a role as an inhibitor of skeletal development and postnatal bone formation. Overexpression of Notch inhibits endochondral bone formation and osteoblastic differentiation, causing severe osteopenia. Conditional inactivation of Notch in the skeleton causes an increase in cancellous bone volume and enhanced osteoblastic differentiation. Notch ligands are expressed in the hematopoietic stem cell niche and are critical for the regulation of hematopoietic stem cell self-renewal. Dysregulation of Notch signaling is the underlying cause of diseases affecting the skeletal tissue, including Alagille syndrome, spondylocostal dysostosis, and possibly, osteosarcoma.
Collapse
|
242
|
Mak W, Shao X, Dunstan CR, Seibel MJ, Zhou H. Biphasic glucocorticoid-dependent regulation of Wnt expression and its inhibitors in mature osteoblastic cells. Calcif Tissue Int 2009; 85:538-45. [PMID: 19876584 DOI: 10.1007/s00223-009-9303-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 09/30/2009] [Indexed: 11/26/2022]
Abstract
Glucocorticoids exert both anabolic and catabolic effects on bone. Previously, we reported that endogenous glucocorticoids control mesenchymal lineage commitment and osteoblastogenesis through regulation of Wnt signaling in osteoblasts. Here, we investigated the effects of glucocorticoids on Wnt expression in mature osteoblasts. Mature osteoblasts and their immature progenitors were separately isolated from Col2.3-GFP transgenic mice in which mature osteoblasts are identifiable through GFP expression. mRNA levels of Wnt2, Wnt2b, Wnt4, Wnt5a, Wnt10b, and Wnt11 were 4- to 12-fold higher in osteoblasts compared to their progenitors (P < 0.05). Expression of Wnt7b and Wnt10b in osteoblasts was modulated by corticosterone (CS), in a biphasic fashion with 3- to 3.5-fold upregulation at 10 nM CS (P < 0.01) and 50% downregulation at 100 nM CS (P < 0.05). CS 100 nM also increased expression of the Wnt inhibitors sFRP-1 and DKK-1 two- to threefold (P < 0.05). We conclude that the contrasting anabolic and catabolic effects of glucocorticoids on bone are, at least in part, mediated through the regulation of Wnt expression and its inhibitors in mature osteoblasts.
Collapse
Affiliation(s)
- Wendy Mak
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Hospital Road, Concord, 2139, Sydney, NSW, Australia
| | | | | | | | | |
Collapse
|
243
|
Igwe JC, Jiang X, Paic F, Ma L, Adams DJ, Baldock PA, Pilbeam CC, Kalajzic I. Neuropeptide Y is expressed by osteocytes and can inhibit osteoblastic activity. J Cell Biochem 2009; 108:621-30. [PMID: 19670271 DOI: 10.1002/jcb.22294] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Osteocytes are the most abundant osteoblast lineage cells within the bone matrix. They respond to mechanical stimulation and can participate in the release of regulatory proteins that can modulate the activity of other bone cells. We hypothesize that neuropeptide Y (NPY), a neurotransmitter with regulatory functions in bone formation, is produced by osteocytes and can affect osteoblast activity. To study the expression of NPY by the osteoblast lineage cells, we utilized transgenic mouse models in which we can identify and isolate populations of osteoblasts and osteocytes. The Col2.3GFP transgene is active in osteoblasts and osteocytes, while the DMP1 promoter drives green fluorescent protein (GFP) expression in osteocytes. Real-time PCR analysis of RNA from the isolated populations of cells derived from neonatal calvaria showed higher NPY mRNA in the preosteocytes/osteocytes fraction compared to osteoblasts. NPY immunostaining confirmed the strong expression of NPY in osteocytes (DMP1GFP(+)), and lower levels in osteoblasts. In addition, the presence of NPY receptor Y1 mRNA was detected in cavaria and long bone, as well as in primary calvarial osteoblast cultures, whereas Y2 mRNA was restricted to the brain. Furthermore, NPY expression was reduced by 30-40% in primary calvarial cultures when subjected to fluid shear stress. In addition, treatment of mouse calvarial osteoblasts with exogenous NPY showed a reduction in the levels of intracellular cAMP and markers of osteoblast differentiation (osteocalcin, BSP, and DMP1). These results highlight the potential regulation of osteoblast lineage differentiation by local NPY signaling.
Collapse
Affiliation(s)
- John C Igwe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut 06032, USA
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Paic F, Igwe JC, Ravi N, Kronenberg MS, Franceschetti T, Harrington P, Kuo L, Shin DG, Rowe DW, Harris SE, Kalajzic I. Identification of differentially expressed genes between osteoblasts and osteocytes. Bone 2009; 45:682-92. [PMID: 19539797 PMCID: PMC2731004 DOI: 10.1016/j.bone.2009.06.010] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 06/03/2009] [Accepted: 06/04/2009] [Indexed: 11/17/2022]
Abstract
Osteocytes represent the most abundant cellular component of mammalian bones with important functions in bone mass maintenance and remodeling. To elucidate the differential gene expression between osteoblasts and osteocytes we completed a comprehensive analysis of their gene profiles. Selective identification of these two mature populations was achieved by utilization of visual markers of bone lineage cells. We have utilized dual GFP reporter mice in which osteocytes are expressing GFP (topaz) directed by the DMP1 promoter, while osteoblasts are identified by expression of GFP (cyan) driven by 2.3 kb of the Col1a1 promoter. Histological analysis of 7-day-old neonatal calvaria confirmed the expression pattern of DMP1GFP in osteocytes and Col2.3 in osteoblasts and osteocytes. To isolate distinct populations of cells we utilized fluorescent activated cell sorting (FACS). Cell suspensions were subjected to RNA extraction, in vitro transcription and labeling of cDNA and gene expression was analyzed using the Illumina WG-6v1 BeadChip. Following normalization of raw data from four biological replicates, 3444 genes were called present in all three sorted cell populations: GFP negative, Col2.3cyan(+) (osteoblasts), and DMP1topaz(+) (preosteocytes and osteocytes). We present the genes that showed in excess of a 2-fold change for gene expression between DMP1topaz(+) and Col2.3cyan(+) cells. The selected genes were classified and grouped according to their associated gene ontology terms. Genes clustered to osteogenesis and skeletal development such as Bmp4, Bmp8a, Dmp1, Enpp1, Phex and Ank were highly expressed in DMP1topaz(+)cells. Most of the genes encoding extracellular matrix components and secreted proteins had lower expression in DMP1topaz(+) cells, while most of the genes encoding plasma membrane proteins were increased. Interestingly a large number of genes associated with muscle development and function and with neuronal phenotype were increased in DMP1topaz(+) cells, indicating some new aspects of osteocyte biology. Although a large number of genes differentially expressed in DMP1topaz(+) and Col2.3cyan(+) cells in our study have already been assigned to bone development and physiology, for most of them we still lack any substantial data. Therefore, isolation of osteocyte and osteoblast cell populations and their subsequent microarray analysis allowed us to identify a number or genes and pathways with potential roles in regulation of bone mass.
Collapse
Affiliation(s)
- Frane Paic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
- Department of Biology, School of Medicine, Zagreb, Croatia
| | - John C. Igwe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Nori Ravi
- Department of Computer Science, University of Connecticut, Storrs, Connecticut, USA
| | - Mark S. Kronenberg
- 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
| | - Patrick Harrington
- Dept. of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Lynn Kuo
- Dept. of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Don-Guk Shin
- Department of Computer Science, University of Connecticut, Storrs, Connecticut, USA
| | - David W. Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | | | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
245
|
Kumar S, Nagy TR, Ponnazhagan S. Therapeutic potential of genetically modified adult stem cells for osteopenia. Gene Ther 2009; 17:105-16. [PMID: 19741731 DOI: 10.1038/gt.2009.116] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Adult stem cells have therapeutic potential because of their intrinsic capacity for self-renewal, especially for bone regeneration. The present study shows the utility of ex vivo modified mesenchymal stem cells (MSC) to enhance bone density in an immunocompetent mouse model of osteopenia. MSC were transduced ex vivo with a recombinant adeno-associated virus 2 (rAAV2) expressing bone morphogenetic protein 2 (BMP2) under the transcriptional control of collagen type-1alpha promoter. To enrich bone homing in vivo, we further modified the cells to transiently express the mouse alpha4 integrin. The modified MSC were systemically administered to ovariectomized, female C57BL/6 mice. Effects of the therapy were determined by dual-energy X-ray absorptiometry, 3D micro-CT, histology and immunohistochemistry for up to 6 months. Results indicated that mice transplanted with MSC expressing BMP2 showed significant increase in bone mineral density and bone mineral content (P < 0.001) with relatively better proliferative capabilities of bone marrow stromal cells and higher osteocompetent pool of cells compared to control animals. Micro-CT analysis of femora and other bone histomorphometric analyses indicated more trabecular bone following MSC-BMP2 therapy. Results obtained by transplanting genetically modified MSC from green fluorescent protein transgenic mouse suggested that production of BMP2 from transplanted MSC also influenced the mobilization of endogenous progenitors for new bone formation.
Collapse
Affiliation(s)
- S Kumar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | | | | |
Collapse
|
246
|
Askmyr M, Sims NA, Martin TJ, Purton LE. What is the true nature of the osteoblastic hematopoietic stem cell niche? Trends Endocrinol Metab 2009; 20:303-9. [PMID: 19595609 DOI: 10.1016/j.tem.2009.03.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 03/17/2009] [Accepted: 03/23/2009] [Indexed: 01/13/2023]
Abstract
The recently revitalized interest in the regulation of hematopoietic stem cells (HSCs) by the bone marrow microenvironment has resulted in the identification of some important cell types that potentially form the HSC niche. The term 'osteoblast' has commonly been used to describe the endosteal elements of the HSC niche, but these cells are part of a larger family that functions in bone at different stages of differentiation. Given that there is much controversy as to what cell types have important roles in the HSC niche, this review offers an overview of the diverse osteoblastic cell types and discusses the current evidence regarding what roles they have in the HSC niche.
Collapse
Affiliation(s)
- Maria Askmyr
- St Vincent's Institute, Fitzroy, Victoria, 3065, Australia
| | | | | | | |
Collapse
|
247
|
Yin D, Wang Z, Gao Q, Sundaresan R, Parrish C, Yang Q, Krebsbach PH, Lichtler AC, Rowe DW, Hock J, Liu P. Determination of the fate and contribution of ex vivo expanded human bone marrow stem and progenitor cells for bone formation by 2.3ColGFP. Mol Ther 2009; 17:1967-78. [PMID: 19603005 DOI: 10.1038/mt.2009.151] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Bone marrow transplantation can provide an effective cell-based strategy to enhance bone repair. However, the fate of implanted cells and the extent of their contribution to bone osteoinduction remain uncertain. To define the fate of bone marrow-derived cells and their contribution in vivo, we used a bone-specific collagen I promoter (2.3Col) driving green fluorescent protein (GFP) (2.3ColGFP) within a lentiviral vector. Prior to in vivo cell fate determination, we verified a high efficiency of lentiviral transduction in human bone marrow stromal cells (hBMSCs), without altering the proliferation or differentiation potential of these cells. We showed that the 2.3ColGFP marker responded to endogenous transcriptional regulation signals. In a mouse ossicle model, we demonstrated that the 2.3ColGFP marker is able to specifically define human bone marrow-derived stem cells that enter the osteoblast lineage in vivo. In addition, cells labeled with 2.3ColGFP with the donor origin, directly make a major contribution to bone formation. Furthermore, we also demonstrated in a calvarial defect model that a mixture of human bone marrow-derived populations, have stronger bone regenerative potential than that of hBMSCs, and an optimal dose is required for bone regeneration by the mixed populations.
Collapse
Affiliation(s)
- Dezhong Yin
- Aastrom Biosciences Inc., Ann Arbor, Michigan 48105, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Kalak R, Zhou H, Street J, Day RE, Modzelewski JRK, Spies CM, Liu PY, Li G, Dunstan CR, Seibel MJ. Endogenous glucocorticoid signalling in osteoblasts is necessary to maintain normal bone structure in mice. Bone 2009; 45:61-7. [PMID: 19358901 DOI: 10.1016/j.bone.2009.03.673] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 03/19/2009] [Accepted: 03/25/2009] [Indexed: 11/26/2022]
Abstract
The role of endogenous glucocorticosteroids (GC) in bone development is ill-defined. Using the Col2.3-11betaHSD2 transgenic (tg) mouse model, we examined the effect of osteoblast-targeted disruption of intracellular GC signalling on bone growth and strength, and its modulation by factors such as age, gender and skeletal site. Tibiae and L3 vertebrae of 3 and 7-week-old, male and female wild type (WT) mice and their tg, age and sex matched littermates were analysed by micro-CT and mechanical testing. Data were analysed separately for 3 and 7-week-old mice by 2-way ANOVA using genotype (WT, tg), gender and their interactions as factors. Transgenic mice were characterised by lower bone volume, lower trabecular number and higher trabecular separation in tibial trabecular bone, as well as lower tibial cortical bone area and periosteal and endosteal perimeters. These changes resulted in a marked decrease in mechanical bone strength and stiffness in sexually mature, 7-week-old mice. In the tibia, the observed transgene effect was present in 3 and 7-week-old animals, indicating that the biological effect of disrupted GC signalling was independent of sexual maturity. This was not the case for the vertebral bones, where significant differences between tg and WT mice were seen in 7 but not in 3-week-old animals, suggesting that the effects of the transgene at this site may be modulated by age and/or changes in circulating sex hormone levels. Taken together, our results demonstrate that endogenous glucocorticoids may be required for normal bone growth but that their effect on bone structure and strength varies according to the skeletal site and sexual maturity of the animals.
Collapse
Affiliation(s)
- Robert Kalak
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Zhou H, Mak W, Kalak R, Street J, Fong-Yee C, Zheng Y, Dunstan CR, Seibel MJ. Glucocorticoid-dependent Wnt signaling by mature osteoblasts is a key regulator of cranial skeletal development in mice. Development 2009; 136:427-36. [PMID: 19141672 DOI: 10.1242/dev.027706] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Glucocorticoids are important regulators of bone cell differentiation and mesenchymal lineage commitment. Using a cell-specific approach of osteoblast-targeted transgenic disruption of intracellular glucocorticoid signaling, we discovered a novel molecular pathway by which glucocorticoids, mainly through the mature osteoblast, regulate the cellular mechanisms that govern cranial skeleton development. Embryonic and neonatal transgenic mice revealed a distinct phenotype characterized by hypoplasia and osteopenia of the cranial skeleton; disorganized frontal, parietal and interparietal bones; increased suture patency; ectopic differentiation of cartilage in the sagittal suture; and disturbed postnatal removal of parietal cartilage. Concurrently, expression of Mmp14, an enzyme essential for calvarial cartilage removal, was markedly reduced in parietal bone and cartilage of transgenic animals. Expression of Wnt9a and Wnt10b was significantly reduced in osteoblasts with disrupted glucocorticoid signaling, and accumulation of beta-catenin, the upstream regulator of Mmp14 expression, was decreased in osteoblasts, chondrocytes and mesenchymal progenitors of transgenic mice. Supracalvarial injection of Wnt3a protein rescued the transgenic cranial phenotype. These results define novel roles for glucocorticoids in skeletal development and delineate how osteoblasts--under steroid hormone control--orchestrate the intricate process of intramembranous bone formation by directing mesenchymal cell commitment towards osteoblastic differentiation while simultaneously initiating and controlling cartilage dissolution in the postnatal mouse.
Collapse
Affiliation(s)
- Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, NSW 2139, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
250
|
Maye P, Stover ML, Liu Y, Rowe DW, Gong S, Lichtler AC. A BAC-bacterial recombination method to generate physically linked multiple gene reporter DNA constructs. BMC Biotechnol 2009; 9:20. [PMID: 19284652 PMCID: PMC2662825 DOI: 10.1186/1472-6750-9-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Accepted: 03/13/2009] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Reporter gene mice are valuable animal models for biological research providing a gene expression readout that can contribute to cellular characterization within the context of a developmental process. With the advancement of bacterial recombination techniques to engineer reporter gene constructs from BAC genomic clones and the generation of optically distinguishable fluorescent protein reporter genes, there is an unprecedented capability to engineer more informative transgenic reporter mouse models relative to what has been traditionally available. RESULTS We demonstrate here our first effort on the development of a three stage bacterial recombination strategy to physically link multiple genes together with their respective fluorescent protein (FP) reporters in one DNA fragment. This strategy uses bacterial recombination techniques to: (1) subclone genes of interest into BAC linking vectors, (2) insert desired reporter genes into respective genes and (3) link different gene-reporters together. As proof of concept, we have generated a single DNA fragment containing the genes Trap, Dmp1, and Ibsp driving the expression of ECFP, mCherry, and Topaz FP reporter genes, respectively. Using this DNA construct, we have successfully generated transgenic reporter mice that retain two to three gene readouts. CONCLUSION The three stage methodology to link multiple genes with their respective fluorescent protein reporter works with reasonable efficiency. Moreover, gene linkage allows for their common chromosomal integration into a single locus. However, the testing of this multi-reporter DNA construct by transgenesis does suggest that the linkage of two different genes together, despite their large size, can still create a positional effect. We believe that gene choice, genomic DNA fragment size and the presence of endogenous insulator elements are critical variables.
Collapse
Affiliation(s)
- Peter Maye
- Department of Reconstructive Sciences, Center for Regenerative Medicine, University of Connecticut Health Center, Farmington, CT, USA.
| | | | | | | | | | | |
Collapse
|