151
|
WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 2013; 19:179-92. [PMID: 23389618 DOI: 10.1038/nm.3074] [Citation(s) in RCA: 1520] [Impact Index Per Article: 126.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 12/18/2012] [Indexed: 12/11/2022]
Abstract
Low bone mass and strength lead to fragility fractures, for example, in elderly individuals affected by osteoporosis or children with osteogenesis imperfecta. A decade ago, rare human mutations affecting bone negatively (osteoporosis-pseudoglioma syndrome) or positively (high-bone mass phenotype, sclerosteosis and Van Buchem disease) have been identified and found to all reside in components of the canonical WNT signaling machinery. Mouse genetics confirmed the importance of canonical Wnt signaling in the regulation of bone homeostasis, with activation of the pathway leading to increased, and inhibition leading to decreased, bone mass and strength. The importance of WNT signaling for bone has also been highlighted since then in the general population in numerous genome-wide association studies. The pathway is now the target for therapeutic intervention to restore bone strength in millions of patients at risk for fracture. This paper reviews our current understanding of the mechanisms by which WNT signalng regulates bone homeostasis.
Collapse
|
152
|
Galea GL, Meakin LB, Sugiyama T, Zebda N, Sunters A, Taipaleenmaki H, Stein GS, van Wijnen AJ, Lanyon LE, Price JS. Estrogen receptor α mediates proliferation of osteoblastic cells stimulated by estrogen and mechanical strain, but their acute down-regulation of the Wnt antagonist Sost is mediated by estrogen receptor β. J Biol Chem 2013; 288:9035-48. [PMID: 23362266 PMCID: PMC3610976 DOI: 10.1074/jbc.m112.405456] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Mechanical strain and estrogens both stimulate osteoblast proliferation through estrogen receptor (ER)-mediated effects, and both down-regulate the Wnt antagonist Sost/sclerostin. Here, we investigate the differential effects of ERα and -β in these processes in mouse long bone-derived osteoblastic cells and human Saos-2 cells. Recruitment to the cell cycle following strain or 17β-estradiol occurs within 30 min, as determined by Ki-67 staining, and is prevented by the ERα antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride. ERβ inhibition with 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-β]pyrimidin-3-yl] phenol (PTHPP) increases basal proliferation similarly to strain or estradiol. Both strain and estradiol down-regulate Sost expression, as does in vitro inhibition or in vivo deletion of ERα. The ERβ agonists 2,3-bis(4-hydroxyphenyl)-propionitrile and ERB041 also down-regulated Sost expression in vitro, whereas the ERα agonist 4,4′,4″-[4-propyl-(1H)-pyrazol-1,3,5-triyl]tris-phenol or the ERβ antagonist PTHPP has no effect. Tamoxifen, a nongenomic ERβ agonist, down-regulates Sost expression in vitro and in bones in vivo. Inhibition of both ERs with fulvestrant or selective antagonism of ERβ, but not ERα, prevents Sost down-regulation by strain or estradiol. Sost down-regulation by strain or ERβ activation is prevented by MEK/ERK blockade. Exogenous sclerostin has no effect on estradiol-induced proliferation but prevents that following strain. Thus, in osteoblastic cells the acute proliferative effects of both estradiol and strain are ERα-mediated. Basal Sost down-regulation follows decreased activity of ERα and increased activity of ERβ. Sost down-regulation by strain or increased estrogens is mediated by ERβ, not ERα. ER-targeting therapy may facilitate structurally appropriate bone formation by enhancing the distinct ligand-independent, strain-related contributions to proliferation of both ERα and ERβ.
Collapse
Affiliation(s)
- Gabriel L Galea
- School of Veterinary Sciences, University of Bristol, Bristol BS40 5DU, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Dwivedi PP, Lam N, Powell BC. Boning up on glypicans-opportunities for new insights into bone biology. Cell Biochem Funct 2013; 31:91-114. [DOI: 10.1002/cbf.2939] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/09/2012] [Accepted: 11/16/2012] [Indexed: 01/01/2023]
Affiliation(s)
| | - N. Lam
- Craniofacial Research Group; Women's and Children's Health Research Institute; North Adelaide; South Australia; Australia
| | | |
Collapse
|
154
|
Abstract
Much of the mammalian skeleton is composed of bones that originate from cartilage templates through endochondral ossification. Elucidating the mechanisms that control endochondral bone development is critical for understanding human skeletal diseases, injury response, and aging. Mouse genetic studies in the past 15 years have provided unprecedented insights about molecules regulating chondrocyte formation, chondrocyte maturation, and osteoblast differentiation, all key processes of endochondral bone development. These include the roles of the secreted proteins IHH, PTHrP, BMPs, WNTs, and FGFs, their receptors, and transcription factors such as SOX9, RUNX2, and OSX, in regulating chondrocyte and osteoblast biology. This review aims to integrate the known functions of extracellular signals and transcription factors that regulate development of the endochondral skeleton.
Collapse
Affiliation(s)
- Fanxin Long
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | |
Collapse
|
155
|
Xu XJ, Shen L, Yang YP, Zhu R, Shuai B, Li CG, Wu MX. Serum β -Catenin Levels Associated with the Ratio of RANKL/OPG in Patients with Postmenopausal Osteoporosis. Int J Endocrinol 2013; 2013:534352. [PMID: 23710175 PMCID: PMC3654357 DOI: 10.1155/2013/534352] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/22/2013] [Accepted: 04/01/2013] [Indexed: 12/13/2022] Open
Abstract
Objective. To demonstrate the role of Wnt/ β -catenin canonical pathway in postmenopausal osteoporosis by evaluating serum β -catenin levels in patients with postmenopausal osteoporosis and analyzing their possible relationship with serum OPG, RANKL, the ratio of RANKL/OPG, sclerostin, and bone turnover markers. Methods. 480 patients with postmenopausal osteoporosis and 170 healthy postmenopausal women were enrolled in the study. Serum β -catenin, OPG, RANKL, and sclerostin levels were measured by enzyme-linked immunosorbent assay. Bone status was assessed by measuring bone mineral density and bone turnover markers. Estradiol levels were also detected. Results. Serum β -catenin levels were lower in postmenopausal osteoporotic women compared to nonosteoporotic postmenopausal women (26.26 ± 14.81 versus 39.33 ± 5.47 pg/mL, P < 0.001). Serum β -catenin was positively correlated with osteoprotegerin (r = 0.232, P < 0.001) and negatively correlated with the ratio of RANKL/OPG, body mass index, and sclerostin (r = -0.128, P = 0.005; r = -0.117, P = 0.010; r = -0.400, P < 0.001, resp.) in patients with postmenopausal osteoporosis. Conclusion. The results indicate that lower serum β -catenin and concomitantly higher ratio of RANKL/OPG may be involved in the pathogenesis of postmenopausal osteoporosis. Functional communication between RANKL/RANK/OPG system and Wnt pathways plays an important role in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Xiao-Juan Xu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lin Shen
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- *Lin Shen: and
| | - Yan-Ping Yang
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
- *Yan-Ping Yang:
| | - Rui Zhu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Bo Shuai
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Cheng-Gang Li
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Man-Xiang Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
156
|
Gatti D, Viapiana O, Fracassi E, Idolazzi L, Dartizio C, Povino MR, Adami S, Rossini M. Sclerostin and DKK1 in postmenopausal osteoporosis treated with denosumab. J Bone Miner Res 2012; 27:2259-63. [PMID: 22692843 DOI: 10.1002/jbmr.1681] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The bone mass benefits of antiresorbers in postmenopausal osteoporosis are limited by the rapid coupling of decreasing bone resorption with bone formation. Wnt signaling is involved in this coupling process during treatment with bisphosphonates, whereas its role during treatment with the anti-receptor activator of NF-κB ligand (RANKL) antibody denosumab is unknown. The study population includes patients participating in a placebo-controlled trial lasting 36 months: 19 women were on placebo and 24 on subcutaneous 60 mg denosumab every 6 months. All measured parameters (serum C-terminal telopeptide of type I collagen [sCTX], serum bone alkaline phosphatase [bAP], Dickkopf-1 [DKK1], and sclerostin) remained unchanged during the observation period in the placebo group. sCTX and bAP were significantly suppressed by denosumab treatment over the entire follow-up. Denosumab treatment was associated with significant (p < 0.05) increases (28% to 32%) in serum sclerostin over the entire study follow-up. Serum DKK1 significantly decreased within the first 6 months with a trend for further continuous decreases, which reached statistical significance (p < 0.05) versus placebo group from the 18th month onward. The changes in DKK1 were significantly and positively related with the changes in sCTX and bAP and negatively with hip bone mineral density (BMD) changes. The changes in sclerostin were significantly and negatively related only with those of bAP. The changes in bone turnover markers associated with denosumab treatment of postmenopausal osteoporosis is associated with significant increase in sclerostin similar to those seen after long-term treatment with bisphosphonates and significant decrease in DKK1. This latter observation might explain the continuous increase over 5 years in BMD observed during treatment of postmenopausal osteoporosis with denosumab.
Collapse
Affiliation(s)
- Davide Gatti
- Unit of Rheumatology, University of Verona, Verona, Italy
| | | | | | | | | | | | | | | |
Collapse
|
157
|
Guo B, Peng S, Liang C, He X, Xiao C, Lu C, Jiang M, Zhao H, Lu A, Zhang G. Recent developments in bone anabolic therapy for osteoporosis. Expert Rev Endocrinol Metab 2012; 7:677-685. [PMID: 30754125 DOI: 10.1586/eem.12.63] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteoporosis is a disorder in which there is a net bone loss and microarchitectural deterioration with an increased risk of bone fracture because of uncoupling of bone formation and bone resorption. The treatment of osteoporosis aims to inhibit bone resorption by osteoclasts and/or promote bone formation by osteoblasts. However, most of the current approaches for treating osteoporosis focus on inhibiting bone resorption. As the only US FDA-approved anabolic agent, the recombinant human parathyroid hormone is recommended for consecutive 2-year period treatment in a clinical setting. Therefore, it is highly desirable to identify novel bone anabolic agents or approaches for osteoporosis treatment. In this review, the authors introduce a new bone anabolic therapy by means of RNAi strategy. Specifically, the authors also discuss the current status and perspectives for RNAi as a novel anabolic approach in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Baosheng Guo
- a Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Songlin Peng
- a Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chao Liang
- a Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaojuan He
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Cheng Xiao
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Cheng Lu
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Miao Jiang
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Hongyan Zhao
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Aiping Lu
- b Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Dongzhimen, Beijing 100700, China
| | - Ge Zhang
- c Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
158
|
Ke HZ, Richards WG, Li X, Ominsky MS. Sclerostin and Dickkopf-1 as therapeutic targets in bone diseases. Endocr Rev 2012; 33:747-83. [PMID: 22723594 DOI: 10.1210/er.2011-1060] [Citation(s) in RCA: 304] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The processes of bone growth, modeling, and remodeling determine the structure, mass, and biomechanical properties of the skeleton. Dysregulated bone resorption or bone formation may lead to metabolic bone diseases. The Wnt pathway plays an important role in bone formation and regeneration, and expression of two Wnt pathway inhibitors, sclerostin and Dickkopf-1 (DKK1), appears to be associated with changes in bone mass. Inactivation of sclerostin leads to substantially increased bone mass in humans and in genetically manipulated animals. Studies in various animal models of bone disease have shown that inhibition of sclerostin using a monoclonal antibody (Scl-Ab) increases bone formation, density, and strength. Additional studies show that Scl-Ab improves bone healing in models of bone repair. Inhibition of DKK1 by monoclonal antibody (DKK1-Ab) stimulates bone formation in younger animals and to a lesser extent in adult animals and enhances fracture healing. Thus, sclerostin and DKK1 are emerging as the leading new targets for anabolic therapies to treat bone diseases such as osteoporosis and for bone repair. Clinical trials are ongoing to evaluate the effects of Scl-Ab and DKK1-Ab in humans for the treatment of bone loss and for bone repair.
Collapse
Affiliation(s)
- Hua Zhu Ke
- Metabolic Disorders Research, Amgen Inc., One Amgen Center Drive, MS 29-M-B, Thousand Oaks, California 91320, USA.
| | | | | | | |
Collapse
|
159
|
Ehnert S, Zhao J, Pscherer S, Freude T, Dooley S, Kolk A, Stöckle U, Nussler AK, Hube R. Transforming growth factor β1 inhibits bone morphogenic protein (BMP)-2 and BMP-7 signaling via upregulation of Ski-related novel protein N (SnoN): possible mechanism for the failure of BMP therapy? BMC Med 2012; 10:101. [PMID: 22958403 PMCID: PMC3523027 DOI: 10.1186/1741-7015-10-101] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 09/07/2012] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Bone morphogenic proteins (BMPs) play a key role in bone formation. Consequently, it was expected that topical application of recombinant human (rh)BMP-2 and rhBMP-7 would improve the healing of complex fractures. However, up to 36% of fracture patients do not respond to this therapy. There are hints that a systemic increase in transforming growth factor β1 (TGFβ1) interferes with beneficial BMP effects. Therefore, in the present work we investigated the influence of rhTGFβ1 on rhBMP signaling in primary human osteoblasts, with the aim of more specifically delineating the underlying regulatory mechanisms. METHODS BMP signaling was detected by adenoviral Smad-binding-element-reporter assays. Gene expression was determined by reverse transcription polymerase chain reaction (RT-PCR) and confirmed at the protein level by western blot. Histone deacetylase (HDAC) activity was determined using a test kit. Data sets were compared by one-way analysis of variance. RESULTS Our findings showed that Smad1/5/8-mediated rhBMP-2 and rhBMP-7 signaling is completely blocked by rhTGFβ1. We then investigated expression levels of genes involved in BMP signaling and regulation (for example, Smad1/5/8, TGFβ receptors type I and II, noggin, sclerostin, BMP and activin receptor membrane bound inhibitor (BAMBI), v-ski sarcoma viral oncogene homolog (Ski), Ski-related novel protein N (SnoN) and Smad ubiquitination regulatory factors (Smurfs)) and confirmed the expression of regulated genes at the protein level. Smad7 and SnoN were significantly induced by rhTGFβ1 treatment while expression of Smad1, Smad6, TGFβRII and activin receptor-like kinase 1 (Alk1) was reduced. Elevated SnoN expression was accompanied by increased HDAC activity. Addition of an HDAC inhibitor, namely valproic acid, fully abolished the inhibitory effect of rhTGFβ1 on rhBMP-2 and rhBMP-7 signaling. CONCLUSIONS rhTGFβ1 effectively blocks rhBMP signaling in osteoblasts. As possible mechanism, we postulate an induction of SnoN that increases HDAC activity and thereby reduces the expression of factors required for efficient BMP signaling. Thus, inhibition of HDAC activity may support bone healing during rhBMP therapy in patients with elevated TGFβ serum levels.
Collapse
Affiliation(s)
- Sabrina Ehnert
- BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076, Tübingen, Germany
| | - Jian Zhao
- Department of Traumatology, MRI, Technische Universität München, Ismaninger Str. 22, D-81675 München, Germany
| | - Stefan Pscherer
- Department Nephrology, Klinikum Traunstein, Kliniken Südostbayern AG, Cuno-Niggl-Str. 3, D-83278 Traunstein, Germany
| | - Thomas Freude
- BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076, Tübingen, Germany
| | - Steven Dooley
- Department of Medicine II, University Hospital Mannheim, Ruprecht-Karls-Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, D-68175 Mannheim, Germany
| | - Andreas Kolk
- Department of Oro- and Maxillofacial Surgery, MRI, Technische Universität München, Str. 22, D-81675 München, Germany
| | - Ulrich Stöckle
- BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076, Tübingen, Germany
| | - Andreas Klaus Nussler
- BG Trauma Center, Eberhard Karls Universität Tübingen, Schnarrenbergstr. 95, D-72076, Tübingen, Germany
- Department of Traumatology, MRI, Technische Universität München, Ismaninger Str. 22, D-81675 München, Germany
| | - Robert Hube
- Department of Orthopaedic Surgery, OCM-Clinic Munich, Steinerstr. 6, D-81368 München, Germany
| |
Collapse
|
160
|
Niziolek PJ, Warman ML, Robling AG. Mechanotransduction in bone tissue: The A214V and G171V mutations in Lrp5 enhance load-induced osteogenesis in a surface-selective manner. Bone 2012; 51:459-65. [PMID: 22750014 PMCID: PMC3784262 DOI: 10.1016/j.bone.2012.05.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 05/08/2012] [Accepted: 05/25/2012] [Indexed: 10/28/2022]
Abstract
Mechanotransduction in bone requires components of the Wnt signaling pathway to produce structurally adapted bone elements. In particular, the Wnt co-receptor LDL-receptor-related protein 5 (LRP5) appears to be a crucial protein in the mechanotransduction cascades that translate physical tissue deformation into new bone formation. Recently discovered missense mutations in LRP5 are associated with high bone mass (HBM), and the altered function of these proteins provide insight into LRP5 function in many skeletal processes, including mechanotransduction. We further investigated the role of LRP5 in bone cell mechanotransduction by applying mechanical stimulation in vivo to two different mutant mouse lines, which harbor HBM-causing missense mutations in Lrp5. Axial tibia loading was applied to mature male Lrp5 G171V and Lrp5 A214V knock-in mice, and to their wild type controls. Fluorochrome labeling revealed that 3 days of loading resulted in a significantly enhanced periosteal response in the A214V knock in mice, whereas the G171V mice exhibited a lowered osteogenic threshold on the endocortical surface. In summary, our data further highlight the importance of Lrp5 in bone cell mechanotransduction, and indicate that the HBM-causing mutations in Lrp5 can alter the anabolic response to mechanical stimulation in favor of increased bone gain.
Collapse
Affiliation(s)
- Paul J. Niziolek
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Matthew L. Warman
- Department of Orthopaedic Surgery, Children’s Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Alexander G. Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis (IUPUI), Indianapolis, IN, USA
| |
Collapse
|
161
|
Zhong Z, Williams BO. Integration of cellular adhesion and Wnt signaling: Interactions between N-cadherin and LRP5 and their role in regulating bone mass. J Bone Miner Res 2012; 27:1849-51. [PMID: 22903578 PMCID: PMC3904542 DOI: 10.1002/jbmr.1715] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Zhendong Zhong
- Center for Skeletal Disease Research, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | |
Collapse
|
162
|
Watanabe T, Tamamura Y, Hoshino A, Makino Y, Kamioka H, Amagasa T, Yamaguchi A, Iimura T. Increasing participation of sclerostin in postnatal bone development, revealed by three-dimensional immunofluorescence morphometry. Bone 2012; 51:447-58. [PMID: 22766096 DOI: 10.1016/j.bone.2012.06.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 06/13/2012] [Accepted: 06/19/2012] [Indexed: 12/19/2022]
Abstract
Confocal immunofluorescence tiling imaging revealed the spatio-temporal distributions of osterix and sclerostin in femurs from 3-day-old, 2-week-old and 4-week-old rats to be reciprocally exclusive at the tissue level. Further quantitative three-dimensional immuno fluorescence morphometry demonstrated the increasing distribution of sclerostin in the osteocytic lacuno-canalicular system specifically in diaphysis, which paralleled the cooperative participation and depletion of osterix and β-catenin in adjacent periosteum cells. Treating MC3T3-E1 cells with BIO (a GSK3 inhibitor) induced the stabilization of β-catenin and nuclear translocation of osterix, and negatively regulated osteocalcin/BGLAP and Dmp1. These results collectively demonstrate that the increasing distribution of sclerostin in diaphyseal cortical bone appears to be involved in the attenuation of osterix and β-catenin in adjacent periosteum cells, thus possibly contributing to osteoblast maturation and reducing the osteoblast formation at this bone site. Our confocal microscopy-based imaging analyses provide a comprehensive and detailed view of the spatio-temporal distribution of sclerostin, β-catenin and osterix at the tissue to subcellular level in a coherent manner, and uncovered their spatio-temporal cooperation in postnatal bone development, thus providing evidence that they link skeletogenic growth and functional bone development.
Collapse
Affiliation(s)
- Takashi Watanabe
- Section of Oral Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | | | |
Collapse
|
163
|
Abstract
The WNT signal transduction cascade controls myriad biological phenomena throughout development and adult life of all animals. In parallel, aberrant Wnt signaling underlies a wide range of pathologies in humans. In this Review, we provide an update of the core Wnt/β-catenin signaling pathway, discuss how its various components contribute to disease, and pose outstanding questions to be addressed in the future.
Collapse
Affiliation(s)
- Hans Clevers
- Hubrecht Institute, KNAW and University Medical Center Utrecht, The Netherlands.
| | | |
Collapse
|
164
|
Gkotzamanidou M, Dimopoulos MA, Kastritis E, Christoulas D, Moulopoulos LA, Terpos E. Sclerostin: a possible target for the management of cancer-induced bone disease. Expert Opin Ther Targets 2012; 16:761-9. [DOI: 10.1517/14728222.2012.697154] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
165
|
Holdsworth G, Slocombe P, Doyle C, Sweeney B, Veverka V, Le Riche K, Franklin RJ, Compson J, Brookings D, Turner J, Kennedy J, Garlish R, Shi J, Newnham L, McMillan D, Muzylak M, Carr MD, Henry AJ, Ceska T, Robinson MK. Characterization of the interaction of sclerostin with the low density lipoprotein receptor-related protein (LRP) family of Wnt co-receptors. J Biol Chem 2012; 287:26464-77. [PMID: 22696217 DOI: 10.1074/jbc.m112.350108] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
LRP5 and LRP6 are proteins predicted to contain four six-bladed β-propeller domains and both bind the bone-specific Wnt signaling antagonist sclerostin. Here, we report the crystal structure of the amino-terminal region of LRP6 and using NMR show that the ability of sclerostin to bind to this molecule is mediated by the central core of sclerostin and does not involve the amino- and carboxyl-terminal flexible arm regions. We show that this structured core region interacts with LRP5 and LRP6 via an NXI motif (found in the sequence PNAIG) within a flexible loop region (loop 2) within the central core region. This sequence is related closely to a previously identified motif in laminin that mediates its interaction with the β-propeller domain of nidogen. However, the NXI motif is not involved in the interaction of sclerostin with LRP4 (another β-propeller containing protein in the LRP family). A peptide derived from the loop 2 region of sclerostin blocked the interaction of sclerostin with LRP5/6 and also inhibited Wnt1 but not Wnt3A or Wnt9B signaling. This suggests that these Wnts interact with LRP6 in different ways.
Collapse
Affiliation(s)
- Gill Holdsworth
- Department of Biology, UCB Pharma, 216 Bath Road, Slough SL1 4EN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Ardawi MSM, Al-Sibiany AM, Bakhsh TM, Rouzi AA, Qari MH. Decreased serum sclerostin levels in patients with primary hyperparathyroidism: a cross-sectional and a longitudinal study. Osteoporos Int 2012; 23:1789-97. [PMID: 22041864 DOI: 10.1007/s00198-011-1806-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Accepted: 09/27/2011] [Indexed: 11/24/2022]
Abstract
UNLABELLED Decreased serum sclerostin was evident in patients with primary hyperparathyroidism and was inversely related to parathyroid hormone (PTH). Sclerostin normalized earlier than biochemical bone turnover markers (BTMs) following parathyroidectomy. INTRODUCTION There is limited information on the changes of serum sclerostin in conditions with chronic PTH excess in humans. The main objectives of the present study were to: (1) examine cross-sectionally the changes of serum sclerostin levels in patients with primary hyperparathyroidism (PHPT), (2) study the time course changes in serum sclerostin in PHPT patients following parathyroidectomy (PTX) followed up longitudinally for 12 months, and (3) compare the changes in serum sclerostin to that of BTMs. METHODS We studied 60 PHPT patients and compared them with 74 PTX patients together with 268 age- and sex-matched healthy controls. Also, we followed 27 PTX patients longitudinally at 2, 4, 6, 10, 30, 60, 180, and 360 days postoperatively. Serum sclerostin, BTMs, and minerals were measured. Also, bone mineral density was determined by dual energy X-ray absorptiometry. RESULTS Patients with PHPT exhibited significantly lower mean serum sclerostin [mean, in picomoles per liter; 95% confidence interval (CI)] (28.98; 27.94-30.03) than that obtained for PTX patients (37.01; 35.75-38.27) and healthy controls (46.22; 45.13-47.31) (P < 0.0001, for each case), respectively. Serum PTH inversely correlated with serum sclerostin (r = -0.651, P < 0.0001). Serum sclerostin was normalized in PTX patients by the tenth day postoperatively and remained within the expected reference range thereafter. CONCLUSIONS Significantly decreased serum sclerostin was evidenced in PHPT patients as compared with PTX and euparathyroid controls. The inverse PTH and sclerostin relationship suggests that sclerostin is downregulated by PTH in humans. Serum sclerostin normalized earlier than BTMs following parathyroidectomy.
Collapse
Affiliation(s)
- M-S M Ardawi
- Center of Excellence for Osteoporosis Research, and Faculty of Medicine, King Abdulaziz University, P.O. Box No. 20724, Jeddah, 21465, Saudi Arabia.
| | | | | | | | | |
Collapse
|
167
|
McNulty MS, Bedell VM, Greenwood TM, Craig TA, Ekker SC, Kumar R. Expression of sclerostin in the developing zebrafish (Danio rerio) brain and skeleton. Gene Expr Patterns 2012; 12:228-35. [PMID: 22575304 DOI: 10.1016/j.gep.2012.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/28/2012] [Accepted: 04/27/2012] [Indexed: 11/15/2022]
Abstract
Sclerostin is a highly conserved, secreted, cystine-knot protein which regulates osteoblast function. Humans with mutations in the sclerostin gene (SOST), manifest increased axial and appendicular skeletal bone density with attendant complications. In adult bone, sclerostin is expressed in osteocytes and osteoblasts. Danio rerio sclerostin-like protein is closely related to sea bass sclerostin, and is related to chicken and mammalian sclerostins. Little is known about the expression of sclerostin in early developing skeletal or extra-skeletal tissues. We assessed sclerostin (sost) gene expression in developing zebrafish (D. rerio) embryos with whole mount is situ hybridization methods. The earliest expression of sost mRNA was noted during 12h post-fertilization (hpf). At 15 hpf, sost mRNA was detected in the developing nervous system and in Kupffer's vesicle. At 18, 20 and 22 hpf, expression in rhombic lip precursors was seen. By 24 hpf, expression in the upper and lower rhombic lip and developing spinal cord was noted. Expression in the rhombic lip and spinal cord persisted through 28 hpf and then diminished in intensity through 44 hpf. At 28 hpf, sost expression was noted in developing pharyngeal cartilage; expression in pharyngeal cartilage increased with time. By 48 hpf, sost mRNA was clearly detected in the developing pharyngeal arch cartilage. Sost mRNA was abundantly expressed in the pharyngeal arch cartilage, and in developing pectoral fins, 72, 96 and 120 hpf. Our study is the first detailed analysis of sost gene expression in early metazoan development.
Collapse
Affiliation(s)
- Melissa S McNulty
- Division of Nephrology and Hypertension, Mayo Clinic, 200 1st St., Southwest, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
168
|
Gatti D, Viapiana O, Adami S, Idolazzi L, Fracassi E, Rossini M. Bisphosphonate treatment of postmenopausal osteoporosis is associated with a dose dependent increase in serum sclerostin. Bone 2012; 50:739-42. [PMID: 22178539 DOI: 10.1016/j.bone.2011.11.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 11/24/2011] [Accepted: 11/30/2011] [Indexed: 12/17/2022]
Abstract
The benefits coming from long-term treatment of postmenopausal osteoporosis with bisphophonates are limited by a coupled decrease in bone formation. The objective of this study is to determine whether this decrease in bone formation is associated with changes in serum levels of the WNT signaling antagonist sclerostin or Dickkopf-1 (DKK1). This is an ancillary observation from patients participating in a 12 months, phase 2, randomized clinical trial. We analyzed 107 patients given either monthly intramuscular neridronate (12.5, 25 or 50 mg) or placebo. Serum C-terminal telopeptide of type I collagen (sCTX, a bone-resorption marker) decreased by 61%, 75% and 73% in the 12.5, 25 and 50 mg dose groups, respectively. Mean changes in bone alkaline phosphatase (bAP) at 12 months were -47%, -60.0% and -52.6% in the groups receiving 12.5, 25 or 50 mg neridronate, respectively. Serum DKK1 remained unchanged at all time points in the 3 groups. Serum sclerostin increased versus placebo group gradually and significantly only in patients treated with 25 or 50 mg neridronate monthly, reaching 138-148% of baseline values (P<0.001). Changes in serum sclerostin at 12 months were negatively correlated with changes in bAP (P<0.001) even when data were adjusted for sCTX changes and only treated patients were included. In conclusions, decreased bone formation after several months of bisphosphonate therapy is associated with increased serum levels of sclerostin. This might suggest that Wnt signaling may play a role in the coupling between resorption and formation.
Collapse
Affiliation(s)
- Davide Gatti
- Rheumatology Unit, Department of Medicine, University of Verona, Italy.
| | | | | | | | | | | |
Collapse
|
169
|
Baron R, Hesse E. Update on bone anabolics in osteoporosis treatment: rationale, current status, and perspectives. J Clin Endocrinol Metab 2012; 97:311-25. [PMID: 22238383 PMCID: PMC3275361 DOI: 10.1210/jc.2011-2332] [Citation(s) in RCA: 250] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteoporosis is defined as low bone mineral density associated with skeletal fractures secondary to minimal or no trauma, most often involving the spine, the hip, and the forearm. The decrease in bone mineral density is the consequence of an unbalanced bone remodeling process, with higher bone resorption than bone formation. Osteoporosis affects predominantly postmenopausal women, but also older men. This chronic disease represents a considerable medical and socioeconomic burden for modern societies. The therapeutic options for the treatment of osteoporosis have so far comprised mostly antiresorptive drugs, in particular bisphosphonates and more recently denosumab, but also calcitonin and, for women, estrogens or selective estrogen receptor modulators. These drugs have limitations, however, in particular the fact that they lead to a low turnover state where bone formation decreases with the decrease in bone-remodeling activity. In this review, we discuss the alternative class of osteoporosis drugs, i.e. bone anabolics, their biology, and the perspectives they offer for our therapeutic armamentarium. We focus on the two main osteoanabolic pathways identified as of today: PTH, the only anabolic drug currently on the market; and activation of canonical Wnt signaling through inhibition of the endogenous inhibitors sclerostin and dickkopf1. Each approach is based on a different molecular mechanism, but most recent evidence suggests that these two pathways may actually converge, at least in part. Whereas recombinant human PTH treatment is being revisited with different formulations and attempts to regulate endogenous PTH secretion via the calcium-sensing receptor, antibodies to sclerostin and dickkopf1 are currently in clinical trials and may prove to be even more efficient at increasing bone mass, possibly independent of bone turnover. Each of these anabolic approaches has its own limitations and safety issues, but the prospects of effective anabolic therapy for osteoporosis are indeed bright.
Collapse
Affiliation(s)
- Roland Baron
- Department of Medicine, Harvard Medical School, Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
170
|
Shi C, Li J, Wang W, Cao W, Cao X, Wan M. Antagonists of LRP6 regulate PTH-induced cAMP generation. Ann N Y Acad Sci 2012; 1237:39-46. [PMID: 22082363 DOI: 10.1111/j.1749-6632.2011.06226.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
LRP6 is a common coreceoptor for different G protein-coupled seven-transmembrane receptors in production of cAMP. Extracelluar proteins sclerostin and DKK1, initially identified as antagonists for Wnt signaling by binding to LRP6, are negative regulators for bone formation. Here, we show that both sclerostin and DKK1 inhibit PTH-stimulated cAMP production. In addition, PTH suppresses expression of sclerostin in osteocytes in mice. We also found that sclerostin and DKK1 binds to LRP6 as antagonists to increase the availability of LRP6 to facilitate PTH signaling in a positive-feedback fashion. These studies reveal a previously unrecognized function of sclerostin and DKK1, which provides an alternative explanation for the application of sclerostin and DKK1 neutralization on enhancing bone formation as a potential therapy for skeletal diseases.
Collapse
Affiliation(s)
- Chenhui Shi
- Shihezi Medical College, Shihezi University, Xinjiang, China
| | | | | | | | | | | |
Collapse
|
171
|
Devarajan-Ketha H, Craig TA, Madden BJ, Robert Bergen H, Kumar R. The sclerostin-bone protein interactome. Biochem Biophys Res Commun 2011; 417:830-5. [PMID: 22206666 DOI: 10.1016/j.bbrc.2011.12.048] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 12/13/2011] [Indexed: 01/25/2023]
Abstract
The secreted glycoprotein, sclerostin alters bone formation. To gain insights into the mechanism of action of sclerostin, we examined the interactions of sclerostin with bone proteins using a sclerostin affinity capture technique. Proteins from decalcified rat bone were captured on a sclerostin-maltose binding protein (MBP) amylose column, or on a MBP amylose column. The columns were extensively washed with low ionic strength buffer, and bound proteins were eluted with buffer containing 1M sodium chloride. Eluted proteins were separated by denaturing sodium-dodecyl sulfate gel electrophoresis and were identified by mass spectrometry. Several previously unidentified full-length sclerostin-interacting proteins such as alkaline phosphatase, carbonic anhydrase, gremlin-1, fetuin A, midkine, annexin A1 and A2, and collagen α1, which have established roles in bone formation or resorption processes, were bound to the sclerostin-MBP amylose resin but not to the MBP amylose resin. Other full-length sclerostin-interacting proteins such as casein kinase II and secreted frizzled related protein 4 that modulate Wnt signaling were identified. Several peptides derived from proteins such as Phex, asporin and follistatin that regulate bone metabolism also bound sclerostin. Sclerostin interacts with multiple proteins that alter bone formation and resorption and is likely to function by altering several biologically relevant pathways in bone.
Collapse
Affiliation(s)
- Hemamalini Devarajan-Ketha
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
172
|
Building strong bones: molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol 2011; 13:27-38. [DOI: 10.1038/nrm3254] [Citation(s) in RCA: 773] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
173
|
Yu L, van der Valk M, Cao J, Han CYE, Juan T, Bass MB, Deshpande C, Damore MA, Stanton R, Babij P. Sclerostin expression is induced by BMPs in human Saos-2 osteosarcoma cells but not via direct effects on the sclerostin gene promoter or ECR5 element. Bone 2011; 49:1131-40. [PMID: 21890009 DOI: 10.1016/j.bone.2011.08.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 08/09/2011] [Accepted: 08/14/2011] [Indexed: 12/15/2022]
Abstract
Sclerostin is a secreted inhibitor of Wnt signaling and plays an essential role in the regulation of bone mass. The expression of sclerostin is largely restricted to osteocytes although its mode of transcriptional regulation is not well understood. We observed regulated expression of sclerostin mRNA and protein that was directly correlated with the mineralization response in cultured human Saos-2 osteosarcoma cells and rat primary calvarial cells. Sclerostin mRNA and protein levels were increased following treatment of cells with BMP2, BMP4 and BMP7. Analysis of deletion mutants from the -7.4 kb upstream region of the human sclerostin promoter did not reveal any specific regions that were responsive to BMPs, Wnt3a, PTH, TGFβ1 or Activin A in Saos-2 cells. The downstream ECR5 element did not show enhancer activity in Saos-2 cells and also was not affected when Saos-2 cells were treated with BMPs or PTH. Genome-wide microarray analysis of Saos-2 cells treated with BMP2 showed significant changes in expression of several transcription factors with putative consensus DNA binding sites in the region of the sclerostin promoter. However, whereas most factors tested showed either a range of inhibitory activity (DLX family, MSX2, HEY1, SMAD6/7) or lack of activity on the sclerostin promoter including SMAD9, only MEF2B showed a positive effect on both the promoter and ECR5 element. These results suggest that the dramatic induction of sclerostin gene expression by BMPs in Saos-2 cells occurs indirectly and is associated with late stage differentiation of osteoblasts and the mineralization process.
Collapse
MESH Headings
- Activins/pharmacology
- Adaptor Proteins, Signal Transducing
- Animals
- Bone Morphogenetic Proteins/genetics
- Bone Morphogenetic Proteins/metabolism
- Bone Morphogenetic Proteins/pharmacology
- Calcification, Physiologic/drug effects
- Calcification, Physiologic/genetics
- Cell Line, Tumor
- Enhancer Elements, Genetic/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Genetic Markers/genetics
- Humans
- Osteoblasts/drug effects
- Osteoblasts/metabolism
- Osteogenesis/drug effects
- Osteogenesis/genetics
- Osteosarcoma/genetics
- Osteosarcoma/pathology
- Parathyroid Hormone/pharmacology
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transforming Growth Factor beta1/pharmacology
- Wnt3A Protein/pharmacology
Collapse
Affiliation(s)
- Longchuan Yu
- Department of Metabolic Disorders, One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Zhang F, Ren LF, Lin HS, Yin MN, Tong YQ, Shi GS. The optimal dose of recombinant human osteogenic protein-1 enhances differentiation of mouse osteoblast-like cells: an in vitro study. Arch Oral Biol 2011; 57:460-8. [PMID: 22054726 DOI: 10.1016/j.archoralbio.2011.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 09/01/2011] [Accepted: 10/10/2011] [Indexed: 12/25/2022]
Abstract
OBJECTIVE There is no certain conclusion on the effect of recombinant human Osteogenic Protein-1 (OP-1, BMP-7) on the proliferation of the osteoblast-like cell line, MC3T3-E1. Furthermore, the optimal dose of rhOP-1 on cell differentiation still needs to be elucidated. This investigation aims to delineate the biofunctional characteristics of rhOP-1 in inducing osteoblastogenesis of MC3T3-E1 through in vitro time-course and dose-response studies. DESIGN MC3T3-E1 cells were cultured for 1, 4, 7 days with the addition of different rhOP-1 concentrations (0, 10, 20, 50, 100, 200, 400 ng/ml), and cell proliferation and cell differentiation were examined. RESULTS MC3T3-E1 cell proliferation was stimulated by rhOP-1 in a dose-dependent manner (0-400 ng/ml) on day 1, whereas on day 4 and 7, it was still stimulated at low concentrations (10, 20, 50 ng/ml) but inhibited at high ones (200, 400 ng/ml). The alkaline phosphatase (ALP) activity, osteocalcin (OC) production, collagen deposition and extracellular matrix mineralization were dramatically elevated by rhOP-1 treatment, as a function of culture time and rhOP-1 concentration, and all of them reached a plateau at the concentration of 200 ng/ml. Real-time quantitative RT-PCR results showed Runx2, AKP-2, OC and Nog mRNA expressions increased in a dose- and time-dependent manner, and their expressions were significantly higher at high rhOP-1 concentrations than that of low ones. No significant differences were found between the effects of 200 ng/ml rhOP-1 and 400 ng/ml rhOP-1 on the differentiation of MC3T3-E1 cells, except the expression of Nog mRNA, whose expression level was much higher at 400 ng/ml than that at 200 ng/ml. CONCLUSIONS These results suggest that cell proliferation of MC3T3-E1 is depended on culture time and rhOP-1 concentration, rhOP-1 could stimulate the differentiation of MC3T3-E1 cells and the optimal concentration could be 200 ng/ml.
Collapse
Affiliation(s)
- Feng Zhang
- Taizhou Hospital of Zhejiang Province, Linhai, PR China
| | | | | | | | | | | |
Collapse
|
175
|
Update on Wnt signaling in bone cell biology and bone disease. Gene 2011; 492:1-18. [PMID: 22079544 DOI: 10.1016/j.gene.2011.10.044] [Citation(s) in RCA: 298] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 10/13/2011] [Accepted: 10/20/2011] [Indexed: 12/17/2022]
Abstract
For more than a decade, Wnt signaling pathways have been the focus of intense research activity in bone biology laboratories because of their importance in skeletal development, bone mass maintenance, and therapeutic potential for regenerative medicine. It is evident that even subtle alterations in the intensity, amplitude, location, and duration of Wnt signaling pathways affects skeletal development, as well as bone remodeling, regeneration, and repair during a lifespan. Here we review recent advances and discrepancies in how Wnt/Lrp5 signaling regulates osteoblasts and osteocytes, introduce new players in Wnt signaling pathways that have important roles in bone development, discuss emerging areas such as the role of Wnt signaling in osteoclastogenesis, and summarize progress made in translating basic studies to clinical therapeutics and diagnostics centered around inhibiting Wnt pathway antagonists, such as sclerostin, Dkk1 and Sfrp1. Emphasis is placed on the plethora of genetic studies in mouse models and genome wide association studies that reveal the requirement for and crucial roles of Wnt pathway components during skeletal development and disease.
Collapse
|
176
|
Niziolek PJ, Farmer T, Cui Y, Turner CH, Warman ML, Robling AG. High-bone-mass-producing mutations in the Wnt signaling pathway result in distinct skeletal phenotypes. Bone 2011; 49:1010-9. [PMID: 21855668 PMCID: PMC3412139 DOI: 10.1016/j.bone.2011.07.034] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/18/2011] [Accepted: 07/21/2011] [Indexed: 10/17/2022]
Abstract
Mutations among genes that participate in the canonical Wnt signaling pathway can lead to drastically different skeletal phenotypes, ranging from severe osteoporosis to severe osteosclerosis. Many high-bone-mass (HBM) causing mutations that occur in the LRP5 gene appear to impart the HBM phenotype, in part, by increasing resistance to soluble Wnt signaling inhibitors, including sclerostin. Sost loss-of-function mutant mice (Sost knock-out) and Lrp5 gain-of-function mutant mice (Lrp5 HBM knock-in) have high bone mass. These mutants potentially would be predicted to be phenocopies of one another, because in both cases, the sclerostin-Lrp5 interaction is disrupted. We measured bone mass, size, geometry, architecture, and strength in bones from three different genetic mouse models (Sost knock-out, Lrp5 A214V knock-in, and Lrp5 G171V knock-in) of HBM. We found that all three mouse lines had significantly elevated bone mass in the appendicular skeleton and in the cranium. Sost mutants and Lrp5 A214V mutants were statistically indistinguishable from one another in most endpoints, whereas both were largely different from the Lrp5 G171V mutants. Lrp5 G171V mutants preferentially added bone endocortically, whereas Lrp5 A214V and Sost mutants preferentially added bone periosteally. Cranial thickness and cranial nerve openings were similarly altered in all three HBM models. We also assessed serum serotonin levels as a possible mechanism accounting for the observed changes in bone mass, but no differences in serum serotonin were found in any of the three HBM mouse lines. The skeletal dissimilarities of the Lrp5 G171V mutant to the other mutants suggest that other, non-sclerostin-associated mechanisms might account for the changes in bone mass resulting from this mutation.
Collapse
Affiliation(s)
- Paul J. Niziolek
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Takeisha Farmer
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yajun Cui
- Department of Genetics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Charles H. Turner
- Department of Biomedical Engineering, Indiana University–Purdue University at Indianapolis, Indianapolis, IN, USA
| | - Matthew L. Warman
- Department of Orthopaedic Surgery, Children’s Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Alexander G. Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Engineering, Indiana University–Purdue University at Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
177
|
Ahn VE, Chu MLH, Choi HJ, Tran D, Abo A, Weis WI. Structural basis of Wnt signaling inhibition by Dickkopf binding to LRP5/6. Dev Cell 2011; 21:862-73. [PMID: 22000856 DOI: 10.1016/j.devcel.2011.09.003] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 08/07/2011] [Accepted: 09/12/2011] [Indexed: 12/17/2022]
Abstract
LDL receptor-related proteins 5 and 6 (LRP5/6) are coreceptors for Wnt growth factors, and also bind Dkk proteins, secreted inhibitors of Wnt signaling. The LRP5/6 ectodomain contains four β-propeller/EGF-like domain repeats. The first two repeats, LRP6(1-2), bind to several Wnt variants, whereas LRP6(3-4) binds other Wnts. We present the crystal structure of the Dkk1 C-terminal domain bound to LRP6(3-4), and show that the Dkk1 N-terminal domain binds to LRP6(1-2), demonstrating that a single Dkk1 molecule can bind to both portions of the LRP6 ectodomain and thereby inhibit different Wnts. Small-angle X-ray scattering analysis of LRP6(1-4) bound to a noninhibitory antibody fragment or to full-length Dkk1 shows that in both cases the ectodomain adopts a curved conformation that places the first three repeats at a similar height relative to the membrane. Thus, Wnts bound to either portion of the LRP6 ectodomain likely bear a similar spatial relationship to Frizzled coreceptors.
Collapse
Affiliation(s)
- Victoria E Ahn
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
178
|
Cheng Z, Biechele T, Wei Z, Morrone S, Moon RT, Wang L, Xu W. Crystal structures of the extracellular domain of LRP6 and its complex with DKK1. Nat Struct Mol Biol 2011; 18:1204-10. [PMID: 21984209 DOI: 10.1038/nsmb.2139] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 08/12/2011] [Indexed: 12/14/2022]
Abstract
Low-density-lipoprotein (LDL) receptor-related proteins 5 and 6 (LRP5/6) are Wnt co-receptors essential for Wnt/β-catenin signaling. Dickkopf 1 (DKK1) inhibits Wnt signaling by interacting with the extracellular domains of LRP5/6 and is a drug target for multiple diseases. Here we present the crystal structures of a human LRP6-E3E4-DKK1 complex and the first and second halves of human LRP6's four propeller-epidermal growth factor (EGF) pairs (LRP6-E1E2 and LRP6-E3E4). Combined with EM analysis, these data demonstrate that LRP6-E1E2 and LRP6-E3E4 form two rigid structural blocks, with a short intervening hinge that restrains their relative orientation. The C-terminal domain of DKK1 (DKK1c) interacts with the top surface of the LRP6-E3 YWTD propeller and given their structural similarity, probably also that of the LRP6-E1 propeller, through conserved hydrophobic patches buttressed by a network of salt bridges and hydrogen bonds. Our work provides key insights for understanding LRP5/6 structure and the interaction of LRP5/6 with DKK, as well as for drug discovery.
Collapse
Affiliation(s)
- Zhihong Cheng
- Department of Biological Structure, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
179
|
Yang N, Schindeler A, McDonald MM, Seto JT, Houweling PJ, Lek M, Hogarth M, Morse AR, Raftery JM, Balasuriya D, MacArthur DG, Berman Y, Quinlan KGR, Eisman JA, Nguyen TV, Center JR, Prince RL, Wilson SG, Zhu K, Little DG, North KN. α-Actinin-3 deficiency is associated with reduced bone mass in human and mouse. Bone 2011; 49:790-8. [PMID: 21784188 DOI: 10.1016/j.bone.2011.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 06/28/2011] [Accepted: 07/07/2011] [Indexed: 10/18/2022]
Abstract
Bone mineral density (BMD) is a complex trait that is the single best predictor of the risk of osteoporotic fractures. Candidate gene and genome-wide association studies have identified genetic variations in approximately 30 genetic loci associated with BMD variation in humans. α-Actinin-3 (ACTN3) is highly expressed in fast skeletal muscle fibres. There is a common null-polymorphism R577X in human ACTN3 that results in complete deficiency of the α-actinin-3 protein in approximately 20% of Eurasians. Absence of α-actinin-3 does not cause any disease phenotypes in muscle because of compensation by α-actinin-2. However, α-actinin-3 deficiency has been shown to be detrimental to athletic sprint/power performance. In this report we reveal additional functions for α-actinin-3 in bone. α-Actinin-3 but not α-actinin-2 is expressed in osteoblasts. The Actn3(-/-) mouse displays significantly reduced bone mass, with reduced cortical bone volume (-14%) and trabecular number (-61%) seen by microCT. Dynamic histomorphometry indicated this was due to a reduction in bone formation. In a cohort of postmenopausal Australian women, ACTN3 577XX genotype was associated with lower BMD in an additive genetic model, with the R577X genotype contributing 1.1% of the variance in BMD. Microarray analysis of cultured osteoprogenitors from Actn3(-/-) mice showed alterations in expression of several genes regulating bone mass and osteoblast/osteoclast activity, including Enpp1, Opg and Wnt7b. Our studies suggest that ACTN3 likely contributes to the regulation of bone mass through alterations in bone turnover. Given the high frequency of R577X in the general population, the potential role of ACTN3 R577X as a factor influencing variations in BMD in elderly humans warrants further study.
Collapse
Affiliation(s)
- Nan Yang
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney 2145, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
180
|
Bourhis E, Wang W, Tam C, Hwang J, Zhang Y, Spittler D, Huang OW, Gong Y, Estevez A, Zilberleyb I, Rouge L, Chiu C, Wu Y, Costa M, Hannoush RN, Franke Y, Cochran AG. Wnt antagonists bind through a short peptide to the first β-propeller domain of LRP5/6. Structure 2011; 19:1433-42. [PMID: 21944579 DOI: 10.1016/j.str.2011.07.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/30/2011] [Accepted: 07/02/2011] [Indexed: 01/27/2023]
Abstract
The Wnt pathway inhibitors DKK1 and sclerostin (SOST) are important therapeutic targets in diseases involving bone loss or damage. It has been appreciated that Wnt coreceptors LRP5/6 are also important, as human missense mutations that result in bone overgrowth (bone mineral density, or BMD, mutations) cluster to the E1 propeller domain of LRP5. Here, we report a crystal structure of LRP6 E1 bound to an antibody, revealing that the E1 domain is a peptide recognition module. Remarkably, the consensus E1 binding sequence is a close match to a conserved tripeptide motif present in all Wnt inhibitors that bind LRP5/6. We show that this motif is important for DKK1 and SOST binding to LRP6 and for inhibitory function, providing a detailed structural explanation for the effect of the BMD mutations.
Collapse
Affiliation(s)
- Eric Bourhis
- Department of Early Discovery Biochemistry, Genentech Research and Early Development, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
181
|
Involvement of Wnt activation in the micromechanical vibration-enhanced osteogenic response of osteoblasts. J Orthop Sci 2011; 16:598-605. [PMID: 21833614 DOI: 10.1007/s00776-011-0124-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 06/13/2011] [Indexed: 02/05/2023]
Abstract
BACKGROUND Low-magnitude vibration has been widely used as a tool for rehabilitation, enhancing physical performance, and stimulating bone development. Although mechanical stimulation generated by vibrations is regarded as important factor in bone remodeling, the underlying cellular and molecular regulatory mechanisms of this response, which may be important in the development of new mechanobiological strategies, currently remain unclear. METHODS In this study, to investigate the mechanobiological mechanisms of vibration-enhanced osteogenic responses in osteoblasts, MC3T3-E1 cells were subjected to vibrations of different amplitude (0.06, 0.14, 0.32, 0.49, 0.66, and 0.8 × g) at 40 Hz for 30 min/day over 3 days. The osteogenesis-related transcription factors Wnt10B, Sclerostin, OPG, and RANKL were analyzed for mRNA and protein expression. RESULTS The results revealed that protein expression of Wnt10B and OPG was increased in a magnitude-dependent manner by mechanical vibrations at amplitudes of 0.06, 0.14, 0.32, and 0.49 × g; the maximum increases were 2.4-fold (p < 0.001) and 7.9-fold (p < 0.001), respectively, at 0.49 × g. Sclerostin and RANKL levels were reduced at all amplitudes. On the basis of mRNA levels, the reduced expression of RANKL was further downregulated (p < 0.05) whereas OPG expression was further increased (p < 0.01) when the MC3T3-E1 cells were treated with LiCl compared with the effects of vibration alone. CONCLUSIONS The findings may indicate that Wnt signaling is involved in mechanotransduction at low-magnitude vibration; this may provide a cellular basis, and impetus for further development of, biomechanically based intervention for enhancing bone strength and accelerating implant osseointegration.
Collapse
|
182
|
Pangrazio A, Boudin E, Piters E, Damante G, Iacono NL, D'Elia AV, Vezzoni P, Van Hul W, Villa A, Sobacchi C. Identification of the first deletion in the LRP5 gene in a patient with autosomal dominant osteopetrosis type I. Bone 2011; 49:568-71. [PMID: 21600326 PMCID: PMC3149657 DOI: 10.1016/j.bone.2011.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/04/2011] [Accepted: 05/04/2011] [Indexed: 10/28/2022]
Abstract
In the last decade, the low-density lipoprotein receptor-related protein 5 (LRP5) gene, coding for a coreceptor in the canonical Wnt signalling pathway, has been shown to play an important role in regulating bone mass and to be involved in the pathogenesis of several bone disorders. Here we describe a patient who presented with a clinical picture of Autosomal Dominant Osteopetrosis type I (ADO I), in whom we could identify the first deletion in the LRP5 gene causing increased bone mass. This mutation caused the in-frame deletion of two amino acids in the fourth blade of the first propeller of the protein, namely the highly conserved glycine at position 171 and the following glutamate residue. In vitro studies suggested that the pathogenic effect of this novel mutation could be due to a decreased inhibition of Wnt signalling by the antagonistic proteins sclerostin and Dickkopf-1, encoded respectively by the SOST and DKK1 genes, in the presence of mutated LRP5. Our results highlight an increasing molecular heterogeneity in LRP5-related bone diseases.
Collapse
Affiliation(s)
- Alessandra Pangrazio
- Institute of Genetic and Biomedical Research (IRGB) - Milan Section, National Research Council, 20138 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
| | - Eveline Boudin
- Department of Medical Genetics, University of Antwerp, 2610 Antwerp, Belgium
| | - Elke Piters
- Department of Medical Genetics, University of Antwerp, 2610 Antwerp, Belgium
| | - Giuseppe Damante
- Azienda Ospedaliero-Universitaria di Udine, Istituto di Genetica, 33100 Udine, Italy
| | - Nadia Lo Iacono
- Institute of Genetic and Biomedical Research (IRGB) - Milan Section, National Research Council, 20138 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
| | | | - Paolo Vezzoni
- Institute of Genetic and Biomedical Research (IRGB) - Milan Section, National Research Council, 20138 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
| | - Wim Van Hul
- Department of Medical Genetics, University of Antwerp, 2610 Antwerp, Belgium
| | - Anna Villa
- Institute of Genetic and Biomedical Research (IRGB) - Milan Section, National Research Council, 20138 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
| | - Cristina Sobacchi
- Institute of Genetic and Biomedical Research (IRGB) - Milan Section, National Research Council, 20138 Milan, Italy
- Istituto Clinico Humanitas IRCCS, 20089 Rozzano, Italy
- Corresponding author at: Istituto Clinico Humanitas IRCCS, via Manzoni 113, 20089 Rozzano (MI), Italy. Fax: + 39 0282245191.
| |
Collapse
|
183
|
Dissecting molecular differences between Wnt coreceptors LRP5 and LRP6. PLoS One 2011; 6:e23537. [PMID: 21887268 PMCID: PMC3160902 DOI: 10.1371/journal.pone.0023537] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 07/19/2011] [Indexed: 01/14/2023] Open
Abstract
Low-density lipoprotein receptor-related proteins 5 and 6 (LRP5 and LRP6) serve as Wnt co-receptors for the canonical β-catenin pathway. While LRP6 is essential for embryogenesis, both LRP5 and LRP6 play critical roles for skeletal remodeling, osteoporosis pathogenesis and cancer formation, making LRP5 and LRP6 key therapeutic targets for cancer and disease treatment. LRP5 and LRP6 each contain in the cytoplasmic domain five conserved PPPSPxS motifs that are pivotal for signaling and serve collectively as phosphorylation-dependent docking sites for the scaffolding protein Axin. However existing data suggest that LRP6 is more effective than LRP5 in transducing the Wnt signal. To understand the molecular basis that accounts for the different signaling activity of LRP5 and LRP6, we generated a series of chimeric receptors via swapping LRP5 and LRP6 cytoplasmic domains, LRP5C and LRP6C, and studied their Wnt signaling activity using biochemical and functional assays. We demonstrate that LRP6C exhibits strong signaling activity while LRP5C is much less active in cells. Recombinant LRP5C and LRP6C upon in vitro phosphorylation exhibit similar Axin-binding capability, suggesting that LRP5 and LRP6 differ in vivo at a step prior to Axin-binding, likely at receiving phosphorylation. We identified between the two most carboxyl PPPSPxS motifs an intervening "gap4" region that appears to account for much of the difference between LRP5C and LRP6C, and showed that alterations in this region are sufficient to enhance LRP5 PPPSPxS phosphorylation and signaling to levels comparable to LRP6 in cells. In addition we provide evidence that binding of phosphorylated LRP5 or LRP6 to Axin is likely direct and does not require the GSK3 kinase as a bridging intermediate as has been proposed. Our studies therefore uncover a new and important molecular tuning mechanism for differential regulation of LRP5 and LRP6 phosphorylation and signaling activity.
Collapse
|
184
|
Glantschnig H, Scott K, Hampton R, Wei N, McCracken P, Nantermet P, Zhao JZ, Vitelli S, Huang L, Haytko P, Lu P, Fisher JE, Sandhu P, Cook J, Williams D, Strohl W, Flores O, Kimmel D, Wang F, An Z. A rate-limiting role for Dickkopf-1 in bone formation and the remediation of bone loss in mouse and primate models of postmenopausal osteoporosis by an experimental therapeutic antibody. J Pharmacol Exp Ther 2011; 338:568-78. [PMID: 21531794 DOI: 10.1124/jpet.111.181404] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Genetic studies have linked both osteoporotic and high bone mass phenotypes to low-density lipoprotein receptor-related proteins (LRP4, LRP5, and LRP6). LRPs are receptors for inhibitory Dickkopf-1 (DKK1) protein, and treatment modalities that modulate LRP/DKK1 binding therefore may act as stimulators of bone mass accrual. Here, we report that RH2-18, a fully human monoclonal anti-DKK1 antibody elicits systemic pharmacologic bone efficacy and new bone formation at endosteal bone surfaces in vivo in a mouse model of estrogen-deficiency-induced osteopenia. This was paralleled by partial-to-complete resolution of osteopenia (bone mineral density) at all of the skeletal sites investigated in femur and lumbar-vertebral bodies and the restoration of trabecular bone microarchitecture. More importantly, testing of RH2-18 in adult, osteopenic rhesus macaques demonstrated a rate-limiting role of DKK1 at multiple skeletal sites and responsiveness to treatment. In conclusion, this study provides pharmacologic evidence for the modulation of DKK1 bioactivity in the adult osteopenic skeleton as a viable approach to resolve osteopenia in animal models. Thus, data described here suggest that targeting DKK1 through means such as a fully human anti-DKK1-antibody provides a potential bone-anabolic treatment for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Helmut Glantschnig
- Bone Biology, Merck Research Laboratories, 700 Sumneytown Pike, WP26A-1000, West Point, PA 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Robling AG, Kedlaya R, Ellis SN, Childress PJ, Bidwell JP, Bellido T, Turner CH. Anabolic and catabolic regimens of human parathyroid hormone 1-34 elicit bone- and envelope-specific attenuation of skeletal effects in Sost-deficient mice. Endocrinology 2011; 152:2963-75. [PMID: 21652726 PMCID: PMC3138236 DOI: 10.1210/en.2011-0049] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PTH is a potent calcium-regulating factor that has skeletal anabolic effects when administered intermittently or catabolic effects when maintained at consistently high levels. Bone cells express PTH receptors, but the cellular responses to PTH in bone are incompletely understood. Wnt signaling has recently been implicated in the osteo-anabolic response to the hormone. Specifically, the Sost gene, a major antagonist of Wnt signaling, is down-regulated by PTH exposure. We investigated this mechanism by treating Sost-deficient mice and their wild-type littermates with anabolic and catabolic regimens of PTH and measuring the skeletal responses. Male Sost(+/+) and Sost(-/-) mice were injected daily with human PTH 1-34 (0, 30, or 90 μg/kg) for 6 wk. Female Sost(+/+) and Sost(-/-) mice were continuously infused with vehicle or high-dose PTH (40 μg/kg · d) for 3 wk. Dual energy x-ray absorptiometry-derived measures of intermittent PTH (iPTH)-induced bone gain were impaired in Sost(-/-) mice. Further probing revealed normal or enhanced iPTH-induced cortical bone formation rates but concomitant increases in cortical porosity among Sost(-/-) mice. Distal femur trabecular bone was highly responsive to iPTH in Sost(-/-) mice. Continuous PTH (cPTH) infusion resulted in equal bone loss in Sost(+/+) and Sost(-/-) mice as measured by dual energy x-ray absorptiometry. However, distal femur trabecular bone, but not lumbar spine trabecular bone, was spared the bone-wasting effects of cPTH in Sost(-/-) mice. These results suggest that changes in Sost expression are not required for iPTH-induced anabolism. iPTH-induced resorption of cortical bone might be overstimulated in Sost-deficient environments. Furthermore, Sost deletion protects some trabecular compartments, but not cortical compartments, from bone loss induced by high-dose PTH infusion.
Collapse
Affiliation(s)
- Alexander G Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS 5035, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | | | |
Collapse
|
186
|
Cui Y, Niziolek PJ, MacDonald BT, Zylstra CR, Alenina N, Robinson DR, Zhong Z, Matthes S, Jacobsen CM, Conlon RA, Brommage R, Liu Q, Mseeh F, Powell DR, Yang Q, Zambrowicz B, Gerrits H, Gossen JA, He X, Bader M, Williams BO, Warman ML, Robling AG. Lrp5 functions in bone to regulate bone mass. Nat Med 2011; 17:684-91. [PMID: 21602802 PMCID: PMC3113461 DOI: 10.1038/nm.2388] [Citation(s) in RCA: 350] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 04/27/2011] [Indexed: 12/17/2022]
Abstract
The human skeleton is affected by mutations in low-density lipoprotein receptor-related protein 5 (LRP5). To understand how LRP5 influences bone properties, we generated mice with osteocyte-specific expression of inducible Lrp5 mutations that cause high and low bone mass phenotypes in humans. We found that bone properties in these mice were comparable to bone properties in mice with inherited mutations. We also induced an Lrp5 mutation in cells that form the appendicular skeleton but not in cells that form the axial skeleton; we observed that bone properties were altered in the limb but not in the spine. These data indicate that Lrp5 signaling functions locally, and they suggest that increasing LRP5 signaling in mature bone cells may be a strategy for treating human disorders associated with low bone mass, such as osteoporosis.
Collapse
Affiliation(s)
- Yajun Cui
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Children’s Hospital, Boston, MA
- Department of Genetics, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Paul J. Niziolek
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
- Departments of Anatomy & Cell Biology and Biomedical Engineering, Indiana University School of Medicine, Indianapolis, IN
| | - Bryan T. MacDonald
- F. M. Kirby Neurobiology Center, Children’s Hospital, Boston, Department of Neurology, Harvard Medical School, Boston, MA
| | - Cassandra R. Zylstra
- Cell Signaling and Carcinogenesis Laboratory, Van Andel Research Institute, Grand Rapids, MI
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany
| | - Daniel R. Robinson
- Cell Signaling and Carcinogenesis Laboratory, Van Andel Research Institute, Grand Rapids, MI
| | - Zhendong Zhong
- Cell Signaling and Carcinogenesis Laboratory, Van Andel Research Institute, Grand Rapids, MI
| | - Susann Matthes
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany
| | - Christina M. Jacobsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Children’s Hospital, Boston, MA
| | - Ronald A. Conlon
- Department of Genetics, Case Western Reserve University School of Medicine, Cleveland, OH
| | | | - Qingyun Liu
- Lexicon Pharmaceuticals Incorporated, The Woodlands, TX
| | - Faika Mseeh
- Lexicon Pharmaceuticals Incorporated, The Woodlands, TX
| | | | - Qi Yang
- Lexicon Pharmaceuticals Incorporated, The Woodlands, TX
| | | | - Han Gerrits
- Merck Sharp & Dohme Research Laboratories, Oss, The Netherlands
| | - Jan A. Gossen
- Merck Sharp & Dohme Research Laboratories, Oss, The Netherlands
| | - Xi He
- F. M. Kirby Neurobiology Center, Children’s Hospital, Boston, Department of Neurology, Harvard Medical School, Boston, MA
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin-Buch, Germany
| | - Bart O. Williams
- Cell Signaling and Carcinogenesis Laboratory, Van Andel Research Institute, Grand Rapids, MI
| | - Matthew L. Warman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Children’s Hospital, Boston, MA
- Howard Hughes Medical Institute, Children’s Hospital Boston, and Department of Genetics, Harvard Medical School, Boston, MA
| | - Alexander G. Robling
- Departments of Anatomy & Cell Biology and Biomedical Engineering, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
187
|
Valkenburg KC, Graveel CR, Zylstra-Diegel CR, Zhong Z, Williams BO. Wnt/β-catenin Signaling in Normal and Cancer Stem Cells. Cancers (Basel) 2011; 3:2050-79. [PMID: 24212796 PMCID: PMC3757404 DOI: 10.3390/cancers3022050] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 04/12/2011] [Accepted: 04/13/2011] [Indexed: 12/23/2022] Open
Abstract
The ability of Wnt ligands to initiate a signaling cascade that results in cytoplasmic stabilization of, and nuclear localization of, β-catenin underlies their ability to regulate progenitor cell differentiation. In this review, we will summarize the current knowledge of the mechanisms underlying Wnt/β-catenin signaling and how the pathway regulates normal differentiation of stem cells in the intestine, mammary gland, and prostate. We will also discuss how dysregulation of the pathway is associated with putative cancer stem cells and the potential therapeutic implications of regulating Wnt signaling.
Collapse
Affiliation(s)
- Kenneth C Valkenburg
- Van Andel Research Institute, 333 Bostwick Ave. N.E., Grand Rapids, MI 49503, USA.
| | | | | | | | | |
Collapse
|
188
|
Abstract
BACKGROUND An understanding of the molecular mechanisms controlling bone formation is central to skeletal tissue engineering efforts. The observation that immature animals are able to heal calvarial defects while adult animals are not has proven to be a useful tool for examining these mechanisms. Thus, the authors compared expression of sclerostin, a bone inhibitor, between the calvariae of juvenile and adult mice. METHODS Parietal bone was harvested from juvenile (6-day-old; n = 20) and adult (60-day-old; n = 20) mice. Sclerostin transcript and protein levels were compared between the parietal bone of juvenile and adult mice using polymerase chain reaction, Western blotting, and immunohistochemistry. Finally, osteoblasts from the parietal bone of juvenile and adult mice were harvested and cultured under osteogenic differentiation conditions with and without recombinant sclerostin (200 ng/ml). Terminal osteogenic differentiation was assessed at 21 days with alizarin red staining. RESULTS Polymerase chain reaction, Western blot analysis, and immunohistochemistry all confirmed greater expression of sclerostin in the parietal bone of adult mice when compared with that of juvenile mice. Osteoblasts, whether from juvenile or adult parietal bones, demonstrated reduced capacity for osteogenic differentiation when exposed to recombinant sclerostin. CONCLUSIONS Given the role of sclerostin in inhibiting bone formation, the authors' findings suggest that differences in expression levels of sclerostin may play a role in the differential regenerative capacity of calvariae from juvenile and adult animals. These findings suggest it as a potential target to abrogate in future tissue engineering studies.
Collapse
|
189
|
Leupin O, Piters E, Halleux C, Hu S, Kramer I, Morvan F, Bouwmeester T, Schirle M, Bueno-Lozano M, Fuentes FJR, Itin PH, Boudin E, de Freitas F, Jennes K, Brannetti B, Charara N, Ebersbach H, Geisse S, Lu CX, Bauer A, Van Hul W, Kneissel M. Bone overgrowth-associated mutations in the LRP4 gene impair sclerostin facilitator function. J Biol Chem 2011; 286:19489-500. [PMID: 21471202 DOI: 10.1074/jbc.m110.190330] [Citation(s) in RCA: 209] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Humans lacking sclerostin display progressive bone overgrowth due to increased bone formation. Although it is well established that sclerostin is an osteocyte-secreted bone formation inhibitor, the underlying molecular mechanisms are not fully elucidated. We identified in tandem affinity purification proteomics screens LRP4 (low density lipoprotein-related protein 4) as a sclerostin interaction partner. Biochemical assays with recombinant proteins confirmed that sclerostin LRP4 interaction is direct. Interestingly, in vitro overexpression and RNAi-mediated knockdown experiments revealed that LRP4 specifically facilitates the previously described inhibitory action of sclerostin on Wnt1/β-catenin signaling. We found the extracellular β-propeller structured domain of LRP4 to be required for this sclerostin facilitator activity. Immunohistochemistry demonstrated that LRP4 protein is present in human and rodent osteoblasts and osteocytes, both presumed target cells of sclerostin action. Silencing of LRP4 by lentivirus-mediated shRNA delivery blocked sclerostin inhibitory action on in vitro bone mineralization. Notably, we identified two mutations in LRP4 (R1170W and W1186S) in patients suffering from bone overgrowth. We found that these mutations impair LRP4 interaction with sclerostin and its concomitant sclerostin facilitator effect. Together these data indicate that the interaction of sclerostin with LRP4 is required to mediate the inhibitory function of sclerostin on bone formation, thus identifying a novel role for LRP4 in bone.
Collapse
Affiliation(s)
- Olivier Leupin
- Musculoskeletal Disease Area, Novartis Institutes for Biomedical Research, Novartis Pharma AG, Basel, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Genetos DC, Yellowley CE, Loots GG. Prostaglandin E2 signals through PTGER2 to regulate sclerostin expression. PLoS One 2011; 6:e17772. [PMID: 21436889 PMCID: PMC3059227 DOI: 10.1371/journal.pone.0017772] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 02/14/2011] [Indexed: 11/19/2022] Open
Abstract
The Wnt signaling pathway is a robust regulator of skeletal homeostasis. Gain-of-function mutations promote high bone mass, whereas loss of Lrp5 or Lrp6 co-receptors decrease bone mass. Similarly, mutations in antagonists of Wnt signaling influence skeletal integrity, in an inverse relation to Lrp receptor mutations. Loss of the Wnt antagonist Sclerostin (Sost) produces the generalized skeletal hyperostotic condition of sclerosteosis, which is characterized by increased bone mass and density due to hyperactive osteoblast function. Here we demonstrate that prostaglandin E2 (PGE2), a paracrine factor with pleiotropic effects on osteoblasts and osteoclasts, decreases Sclerostin expression in osteoblastic UMR106.01 cells. Decreased Sost expression correlates with increased expression of Wnt/TCF target genes Axin2 and Tcf3. We also show that the suppressive effect of PGE2 is mediated through a cyclic AMP/PKA pathway. Furthermore, selective agonists for the PGE2 receptor EP2 mimic the effect of PGE2 upon Sost, and siRNA reduction in Ptger2 prevents PGE2-induced Sost repression. These results indicate a functional relationship between prostaglandins and the Wnt/β-catenin signaling pathway in bone.
Collapse
Affiliation(s)
- Damian C Genetos
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America.
| | | | | |
Collapse
|
191
|
Wan M, Li J, Herbst K, Zhang J, Yu B, Wu X, Qiu T, Lei W, Lindvall C, Williams BO, Ma H, Zhang F, Cao X. LRP6 mediates cAMP generation by G protein-coupled receptors through regulating the membrane targeting of Gα(s). Sci Signal 2011; 4:ra15. [PMID: 21406690 DOI: 10.1126/scisignal.2001464] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ligand binding to certain heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) stimulates the rapid synthesis of cyclic adenosine monophosphate (cAMP) through the G protein α(s) subunit, which activates adenylyl cyclase (AC). We found that the transmembrane receptor low-density lipoprotein receptor-related protein 6 (LRP6), a co-receptor for Wnt proteins, bound to the Gα(s)βγ heterotrimer and that knockdown of LRP6 attenuated cAMP production by various GPCRs, including parathyroid hormone receptor 1 (PTH1R). Knockdown of LRP6 disrupted the localization of Gα(s) to the plasma membrane, which led to a decrease in the extent of coupling of Gα(s) to PTH1R and inhibited the production of cAMP and the activation of cAMP-dependent protein kinase (PKA) in response to PTH. PKA phosphorylated LRP6, which enhanced the binding of Gα(s) to LRP6, its localization to the plasma membrane, and the production of cAMP in response to PTH. Decreased PTH-dependent cAMP production was observed in single cells in which LRP6 was knocked down or mutated at the PKA site by monitoring the cAMP kinetics. Thus, we suggest that the binding of Gα(s) to LRP6 is required to establish a functional GPCR-Gα(s)-AC signaling pathway for the production of cAMP, providing an additional regulatory component to the current GPCR-cAMP paradigm.
Collapse
Affiliation(s)
- Mei Wan
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Ohazama A, Porntaveetus T, Ota MS, Herz J, Sharpe PT. Lrp4: A novel modulator of extracellular signaling in craniofacial organogenesis. Am J Med Genet A 2011; 152A:2974-83. [PMID: 21108386 DOI: 10.1002/ajmg.a.33372] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The low-density lipoprotein (LDL) receptor family is a large evolutionarily conserved group of transmembrane proteins. It has been shown that LDL receptor family members can also function as direct signal transducers or modulators for a broad range of cellular signaling pathways. We have identified a novel mode of signaling pathway integration/coordination that occurs outside cells during development that involves an LDL receptor family member. Physical interaction between an extracellular protein (Wise) that binds BMP ligands and an Lrp receptor (Lrp4) that modulates Wnt signaling, acts to link these two pathways. Mutations in either Wise or Lrp4 in mice produce multiple, but identical abnormalities in tooth development that are linked to alterations in BMP and Wnt signaling. Teeth, in common with many other organs, develop by a series of epithelial-mesenchymal interactions, orchestrated by multiple cell signaling pathways. In tooth development, Lrp4 is expressed exclusively in epithelial cells and Wise mainly in mesenchymal cells. Our hypothesis, based on the mutant phenotypes, cell signaling activity changes and biochemical interactions between Wise and Lrp4 proteins, is that Wise and Lrp4 together act as an extracellular mechanism of coordinating BMP and Wnt signaling activities in epithelial-mesenchymal cell communication during development.
Collapse
Affiliation(s)
- Atsushi Ohazama
- Department of Craniofacial Development, Dental Institute, King's College London, Guy's Hospital, London Bridge, London, UK
| | | | | | | | | |
Collapse
|
193
|
Kawai M, Mödder UI, Khosla S, Rosen CJ. Emerging therapeutic opportunities for skeletal restoration. Nat Rev Drug Discov 2011; 10:141-56. [PMID: 21283108 PMCID: PMC3135105 DOI: 10.1038/nrd3299] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteoporosis, a syndrome characterized by thin bones and fractures, has become more prevalent in both women and men. Established therapies for treating this disorder consist primarily of drugs that prevent bone loss, such as the bisphosphonates and selective oestrogen receptor modulators. Although these drugs have been shown to reduce fractures in randomized trials, there is an urgent need for treatments that could lower fracture risk further without additional adverse effects. The introduction of parathyroid hormone (teriparatide), which significantly increases bone mineral density, albeit for a relatively short duration, raised expectations that drugs that stimulate bone formation might cure osteoporosis. After outlining current approaches for treating osteoporosis, this Review focuses on emerging therapeutic opportunities for osteoporosis that are based on recent insights into skeletal physiology. Such novel strategies offer promise not only for reducing age-related bone loss and the associated risk of fractures but also for restoring bone mineral density to healthy levels.
Collapse
Affiliation(s)
- Masanobu Kawai
- Center for clinical and translational research, Maine Medical Center Research Institute, Scarborough, Maine, USA
- Department of Bone and Mineral research, Osaka medical center and research institute for maternal and child health, Izumi, Osaka, Japan
| | | | | | - Clifford J Rosen
- Center for clinical and translational research, Maine Medical Center Research Institute, Scarborough, Maine, USA
| |
Collapse
|
194
|
Naka T, Yokose S. Spatiotemporal expression of sclerostin in odontoblasts during embryonic mouse tooth morphogenesis. J Endod 2011; 37:340-5. [PMID: 21329818 DOI: 10.1016/j.joen.2010.11.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 11/22/2010] [Accepted: 11/23/2010] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Sclerostin is the product of the SOST gene. Loss-of-function mutations in the SOST gene result in a high bone mass phenotype, thus confirming that sclerostin is a negative regulator of bone mass. SOST knockdown in humans also causes oral and dental malformations. However, the relationship between sclerostin and tooth development is unclear. METHODS Using immunohistochemical techniques, we investigated sclerostin expression during fetal mouse tooth development and adult mouse tooth morphogenesis. RESULTS Sclerostin was expressed in the secretory odontoblasts located along the ameloblasts of fetal mouse tooth germ and adult incisor. Sclerostin expression was also observed in the fetal and adult osteocytes in the jaw bone. CONCLUSION These results suggest that sclerostin, one of the important regulatory factors of differentiated odontoblast function, may usable in vital pulp therapy.
Collapse
Affiliation(s)
- Takahiro Naka
- Division of Restorative Dentistry, Department of Conservative Dentistry, Ohu University, School of Dentistry, Fukushima, Japan
| | | |
Collapse
|
195
|
|
196
|
Li X, Warmington KS, Niu QT, Asuncion FJ, Barrero M, Grisanti M, Dwyer D, Stouch B, Thway TM, Stolina M, Ominsky MS, Kostenuik PJ, Simonet WS, Paszty C, Ke HZ. Inhibition of sclerostin by monoclonal antibody increases bone formation, bone mass, and bone strength in aged male rats. J Bone Miner Res 2010; 25:2647-56. [PMID: 20641040 DOI: 10.1002/jbmr.182] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 05/20/2010] [Accepted: 07/07/2010] [Indexed: 11/12/2022]
Abstract
The purpose of this study was to evaluate the effects of sclerostin inhibition by treatment with a sclerostin antibody (Scl-AbII) on bone formation, bone mass, and bone strength in an aged, gonad-intact male rat model. Sixteen-month-old male Sprague-Dawley rats were injected subcutaneously with vehicle or Scl-AbII at 5 or 25 mg/kg twice per week for 5 weeks (9-10/group). In vivo dual-energy X-ray absorptiometry (DXA) analysis showed that there was a marked increase in areal bone mineral density of the lumbar vertebrae (L(1) to L(5) ) and long bones (femur and tibia) in both the 5 and 25 mg/kg Scl-AbII-treated groups compared with baseline or vehicle controls at 3 and 5 weeks after treatment. Ex vivo micro-computed tomographic (µCT) analysis demonstrated improved trabecular and cortical architecture at the fifth lumbar vertebral body (L(5) ), femoral diaphysis (FD), and femoral neck (FN) in both Scl-AbII dose groups compared with vehicle controls. The increased cortical and trabecular bone mass was associated with a significantly higher maximal load of L(5) , FD, and FN in the high-dose group. Bone-formation parameters (ie, mineralizing surface, mineral apposition rate, and bone-formation rate) at the proximal tibial metaphysis and tibial shaft were markedly greater on trabecular, periosteal, and endocortical surfaces in both Scl-AbII dose groups compared with controls. These results indicate that sclerostin inhibition by treatment with a sclerostin antibody increased bone formation, bone mass, and bone strength in aged male rats and, furthermore, suggest that pharmacologic inhibition of sclerostin may represent a promising anabolic therapy for low bone mass in aged men.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Metabolic Disorders, Amgen, Inc., Thousand Oaks, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
van Lierop AH, Witteveen JE, Hamdy NAT, Papapoulos SE. Patients with primary hyperparathyroidism have lower circulating sclerostin levels than euparathyroid controls. Eur J Endocrinol 2010; 163:833-7. [PMID: 20817762 DOI: 10.1530/eje-10-0699] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE In vitro and in vivo studies in animal models have shown that parathyroid hormone (PTH) inhibits the expression of the SOST gene, which encodes sclerostin, an osteocyte-derived negative regulator of bone formation. We tested the hypothesis that chronic PTH excess decreases circulating sclerostin in humans. DESIGN We studied 25 patients with elevated serum PTH concentrations due to primary hyperparathyroidism (PHPT) and 49 patients cured from PHPT after successful parathyroidectomy (PTx; euparathyroid controls (EuPTH)). METHODS We measured plasma PTH and serum sclerostin levels and the serum markers of bone turnover alkaline phosphatase, P1NP, and β-CTX. RESULTS As expected by the design of the study, mean plasma PTH was significantly higher (P<0.001) in PHPT patients (15.3 pmol/l; 95% confidence interval (CI): 11.1-19.5) compared with that of EuPTH controls (4.1 pmol/l; 95% CI: 3.6-4.5). PHPT patients had significantly lower serum sclerostin values compared with those in EuPTH subjects (30.5 pg/ml; 95% CI: 26.0-35.1 vs 45.4 pg/ml; 95% CI: 40.5-50.2; P<0.001) and healthy controls (40.0 pg/ml; 95% CI: 37.1-42.9; P=0.01). Plasma PTH concentrations were negatively correlated with serum sclerostin values (r=-0.44; P<0.001). Bone turnover markers were significantly correlated with PTH, but not with sclerostin. CONCLUSION Patients with PHPT have significantly lower serum sclerostin values compared with PTx controls with normal PTH concentrations. The negative correlation between PTH and sclerostin suggests that SOST is downregulated by PTH in humans.
Collapse
Affiliation(s)
- A H van Lierop
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | | | | | | |
Collapse
|
198
|
Li WF, Hou SX, Yu B, Jin D, Férec C, Chen JM. Genetics of osteoporosis: perspectives for personalized medicine. Per Med 2010; 7:655-668. [PMID: 29788568 DOI: 10.2217/pme.10.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteoporosis is the most common metabolic bone disorder worldwide. At least 15 genes (e.g., ESR1, LRP5, SOST, OPG, RANK and RANKL) have been confirmed as osteoporosis susceptibility genes, and another 30 have been highlighted as promising susceptibility genes. Notably, these genes are clustered in three biological pathways: the estrogen endocrine pathway, the Wnt/β-catenin signaling pathway and the RANK/RANKL/osteoprotegerin (OPG) pathway. In this article, using data pertaining to these three biological pathways as examples, we illustrate possible principles of personalized therapy for osteoporosis. In particular, we propose to use inhibitors (e.g., denosumab) of the RANK/RANKL/OPG signaling pathway to circumvent resistance to estrogen-replacement therapy: a novel idea resulting from the consideration of a mechanistic link between the estrogen endocrine pathway and the RANK/RANKL/OPG signaling pathway. In addition, we call for more attention to be focused on rare variants of major effects in future studies.
Collapse
Affiliation(s)
- Wen-Feng Li
- Department of Orthopaedics, The First Affiliated Hospital, General Hospital of the People’s Liberation Army, Beijing, China
| | - Shu-Xun Hou
- Department of Orthopaedics, The First Affiliated Hospital, General Hospital of the People’s Liberation Army, Beijing, China
| | - Bin Yu
- Department of Orthopaedic Trauma, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Jin
- Department of Orthopaedic Trauma, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U613, Brest, France; INSERM, U613 and EFS – Bretagne, 46 rue Félix Le Dantec, 29218 Brest, France
- Etablissement Français du Sang (EFS) – Bretagne, Brest, France
- Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, France
| | | |
Collapse
|
199
|
Krause C, Korchynskyi O, de Rooij K, Weidauer SE, de Gorter DJJ, van Bezooijen RL, Hatsell S, Economides AN, Mueller TD, Löwik CWGM, ten Dijke P. Distinct modes of inhibition by sclerostin on bone morphogenetic protein and Wnt signaling pathways. J Biol Chem 2010; 285:41614-26. [PMID: 20952383 DOI: 10.1074/jbc.m110.153890] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Sclerostin is expressed by osteocytes and has catabolic effects on bone. It has been shown to antagonize bone morphogenetic protein (BMP) and/or Wnt activity, although at present the underlying mechanisms are unclear. Consistent with previous findings, Sclerostin opposed direct Wnt3a-induced but not direct BMP7-induced responses when both ligand and antagonist were provided exogenously to cells. However, we found that when both proteins are expressed in the same cell, sclerostin can antagonize BMP signaling directly by inhibiting BMP7 secretion. Sclerostin interacts with both the BMP7 mature domain and pro-domain, leading to intracellular retention and proteasomal degradation of BMP7. Analysis of sclerostin knock-out mice revealed an inhibitory action of sclerostin on Wnt signaling in both osteoblasts and osteocytes in cortical and cancellous bones. BMP7 signaling was predominantly inhibited by sclerostin in osteocytes of the calcaneus and the cortical bone of the tibia. Our results suggest that sclerostin exerts its potent bone catabolic effects by antagonizing Wnt signaling in a paracrine and autocrine manner and antagonizing BMP signaling selectively in the osteocytes that synthesize simultaneously both sclerostin and BMP7 proteins.
Collapse
Affiliation(s)
- Carola Krause
- Department of Molecular Cell Biology and Centre for Biomedical Genetics, Julius-von-Sachs Institut für Biowissenschaften der UniversitätWürzburg, D-97074 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Craig TA, Kumar R. Sclerostin-erbB-3 interactions: modulation of erbB-3 activity by sclerostin. Biochem Biophys Res Commun 2010; 402:421-4. [PMID: 20951118 DOI: 10.1016/j.bbrc.2010.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 10/11/2010] [Indexed: 11/17/2022]
Abstract
To gain insights into the mechanism of action of sclerostin, a protein that regulates bone mass, we performed yeast two-hybrid analyses using human SOST (sclerostin) cDNA cloned into pGBKT7 DNA-binding domain vector as a bait, and a normalized, high-complexity, universal cDNA library in a GAL4 activating domain vector. We identified an interaction between sclerostin and the carboxyl-terminal portion of the receptor tyrosine-protein kinase erbB-3. To determine the biological relevance of this interaction, we treated MC3T3-E1 mouse osteoblast cells transfected with either a SOST expression plasmid or a control vector, with recombinant heregulin/neuregulin. Phospho-p44/42 (Thr202/Tyr204) MAPK was assessed in heregulin/neuregulin treated cells. We observed an increase in phospho-p44/42 (Thr202/Tyr204) MAPK concentrations in SOST transfected cells but not in cells transfected with a control vector, thus demonstrating a modulatory effect of sclerostin on heregulin/neuregulin signaling in osteoblasts. The data demonstrate that sclerostin functions in part, by modulating the activity of erbB-3.
Collapse
Affiliation(s)
- Theodore A Craig
- Nephrology and Hypertension Research, Department of Internal Medicine, MS 1-120, Mayo Clinic, 200 1st St., Southwest, Rochester, MN 55905, USA
| | | |
Collapse
|