301
|
Costa R, Bellesso S, Lualdi S, Manzoli R, Pistorio V, Filocamo M, Moro E. A transcriptional and post-transcriptional dysregulation of Dishevelled 1 and 2 underlies the Wnt signaling impairment in type I Gaucher disease experimental models. Hum Mol Genet 2019; 29:274-285. [DOI: 10.1093/hmg/ddz293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/02/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Bone differentiation defects have been recently tied to Wnt signaling alterations occurring in vitro and in vivo Gaucher disease (GD) models. In this work, we provide evidence that the Wnt signaling multi-domain intracellular transducers Dishevelled 1 and 2 (DVL1 and DVL2) may be potential upstream targets of impaired beta glucosidase (GBA1) activity by showing their misexpression in different type 1 GD in vitro models. We also show that in Gba mutant fish a miR-221 upregulation is associated with reduced dvl2 expression levels and that in type I Gaucher patients single-nucleotide variants in the DVL2 3′ untranslated region are related to variable canonical Wnt pathway activity. Thus, we strengthen the recently outlined relation between bone differentiation defects and Wnt/β-catenin dysregulation in type I GD and further propose novel mechanistic insights of the Wnt pathway impairment caused by glucocerebrosidase loss of function.
Collapse
Affiliation(s)
- Roberto Costa
- Department of Biology, University of Padova, Padova I-35121, Italy
| | - Stefania Bellesso
- Department of Molecular Medicine, University of Padova, Padova I-35121, Italy
| | - Susanna Lualdi
- Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche Giannina Gaslini Institute, Genova 16147, Italy
| | - Rosa Manzoli
- Department of Biology, University of Padova, Padova I-35121, Italy
- Department of Molecular Medicine, University of Padova, Padova I-35121, Italy
| | - Valeria Pistorio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples 80131, Italy
| | - Mirella Filocamo
- Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche Giannina Gaslini Institute, Genova 16147, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padova, Padova I-35121, Italy
| |
Collapse
|
302
|
Growth Plate Chondrocytes: Skeletal Development, Growth and Beyond. Int J Mol Sci 2019; 20:ijms20236009. [PMID: 31795305 PMCID: PMC6929081 DOI: 10.3390/ijms20236009] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 02/01/2023] Open
Abstract
Growth plate chondrocytes play central roles in the proper development and growth of endochondral bones. Particularly, a population of chondrocytes in the resting zone expressing parathyroid hormone-related protein (PTHrP) is now recognized as skeletal stem cells, defined by their ability to undergo self-renewal and clonally give rise to columnar chondrocytes in the postnatal growth plate. These chondrocytes also possess the ability to differentiate into a multitude of cell types including osteoblasts and bone marrow stromal cells during skeletal development. Using single-cell transcriptomic approaches and in vivo lineage tracing technology, it is now possible to further elucidate their molecular properties and cellular fate changes. By discovering the fundamental molecular characteristics of these cells, it may be possible to harness their functional characteristics for skeletal growth and regeneration. Here, we discuss our current understanding of the molecular signatures defining growth plate chondrocytes.
Collapse
|
303
|
Wnt/β-catenin signaling contributes to articular cartilage homeostasis through lubricin induction in the superficial zone. Arthritis Res Ther 2019; 21:247. [PMID: 31771658 PMCID: PMC6880374 DOI: 10.1186/s13075-019-2041-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022] Open
Abstract
Background Both loss- and gain-of-function of Wnt/β-catenin signaling in chondrocytes result in exacerbation of osteoarthritis (OA). Here, we examined the activity and roles of Wnt/β-catenin signaling in the superficial zone (SFZ) of articular cartilage. Methods Wnt/β-catenin signaling activity was analyzed using TOPGAL mice. We generated Prg4-CreERT2;Ctnnb1fl/fl and Prg4-CreERT2;Ctnnb1-ex3fl/wt mice for loss- and gain-of-function, respectively, of Wnt/β-catenin signaling in the SFZ. Regulation of Prg4 expression by Wnt/β-catenin signaling was examined in vitro, as were upstream and downstream factors of Wnt/β-catenin signaling in SFZ cells. Results Wnt/β-catenin signaling activity, as determined by the TOPGAL reporter, was high specifically in the SFZ of mouse adult articular cartilage, where Prg4 is abundantly expressed. In SFZ-specific β-catenin-knockout mice, OA development was significantly accelerated, which was accompanied by decreased Prg4 expression and SFZ destruction. In contrast, Prg4 expression was enhanced and cartilage degeneration was suppressed in SFZ-specific β-catenin-stabilized mice. In primary SFZ cells, Prg4 expression was downregulated by β-catenin knockout, while it was upregulated by β-catenin stabilization by exon 3 deletion or treatment with CHIR99021. Among Wnt ligands, Wnt5a, Wnt5b, and Wnt9a were highly expressed in SFZ cells, and recombinant human WNT5A and WNT5B stimulated Prg4 expression. Mechanical loading upregulated expression of these ligands and further promoted Prg4 transcription. Moreover, mechanical loading and Wnt/β-catenin signaling activation increased mRNA levels of Creb1, a potent transcription factor for Prg4. Conclusions We demonstrated that Wnt/β-catenin signaling regulates Prg4 expression in the SFZ of mouse adult articular cartilage, which plays essential roles in the homeostasis of articular cartilage.
Collapse
|
304
|
Dasgupta K, Chung JU, Asam K, Jeong J. Molecular patterning of the embryonic cranial mesenchyme revealed by genome-wide transcriptional profiling. Dev Biol 2019; 455:434-448. [PMID: 31351040 PMCID: PMC6842427 DOI: 10.1016/j.ydbio.2019.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
In the head of an embryo, a layer of mesenchyme surrounds the brain underneath the surface ectoderm. This cranial mesenchyme gives rise to the meninges, the calvaria (top part of the skull), and the dermis of the scalp. Abnormal development of these structures, especially the meninges and the calvaria, is linked to significant congenital defects in humans. It has been known that different areas of the cranial mesenchyme have different fates. For example, the calvarial bone develops from the cranial mesenchyme on the baso-lateral side of the head just above the eye (supraorbital mesenchyme, SOM), but not from the mesenchyme apical to SOM (early migrating mesenchyme, EMM). However, the molecular basis of this difference is not fully understood. To answer this question, we compared the transcriptomes of EMM and SOM using high-throughput sequencing (RNA-seq). This experiment identified a large number of genes that were differentially expressed in EMM and SOM, and gene ontology analyses found very different terms enriched in each region. We verified the expression of about 40 genes in the head by RNA in situ hybridization, and the expression patterns were annotated to make a map of molecular markers for 6 subdivisions of the cranial mesenchyme. Our data also provided insights into potential novel regulators of cranial mesenchyme development, including several axon guidance pathways, lectin complement pathway, cyclic-adenosine monophosphate (cAMP) signaling pathway, and ZIC family transcription factors. Together, information in this paper will serve as a unique resource to guide future research on cranial mesenchyme development.
Collapse
Affiliation(s)
- Krishnakali Dasgupta
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Jong Uk Chung
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Kesava Asam
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
305
|
Oo WM, Liu X, Hunter DJ. Pharmacodynamics, efficacy, safety and administration of intra-articular therapies for knee osteoarthritis. Expert Opin Drug Metab Toxicol 2019; 15:1021-1032. [PMID: 31709838 DOI: 10.1080/17425255.2019.1691997] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Osteoarthritis (OA) is the leading cause of disability in the elderly, usually presenting with mono-or oligo-arthritis where local drug delivery by intra-articular (IA) injection may be more effective in terms of increased bioavailability, less systemic exposure and reduced adverse events. Several intra-articular medications for symptomatic are available on the market while the new disease-modifying drugs (DMOADs) are progressing into phase 3 pipeline of drug development.Areas covered: This narrative review covered the pharmacokinetic and pharmacodynamics of clinically available IA drugs which include corticosteroids, hyaluronic acid as well as injection techniques, efficacy, adverse effects and contraindications. In addition, the authors briefly describe the newer disease-modifying OA drugs (DMOAD) which are undergoing phase 3 pipeline of development such as Fibroblast growth factor (FGF-18) and Wnt inhibitor.Expert opinion: This is a rapidly evolving area with both new products and new trials regularly emerging. It is also a critically important area in a disease field that lacks for safe and effective treatments, where intra-articular delivery may enhance both local efficacy and reduce systemic toxicity.
Collapse
Affiliation(s)
- Win Min Oo
- Rheumatology Department, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Sydney, Australia
| | - Xiaoqian Liu
- Rheumatology Department, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Sydney, Australia
| | - David J Hunter
- Rheumatology Department, Royal North Shore Hospital and Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
306
|
Wang R, Zhang Y, Jin F, Li G, Sun Y, Wang X. High-glucose-induced miR-214-3p inhibits BMSCs osteogenic differentiation in type 1 diabetes mellitus. Cell Death Discov 2019; 5:143. [PMID: 31728209 PMCID: PMC6851127 DOI: 10.1038/s41420-019-0223-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/20/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an autoimmune insulin-dependent disease associated with destructive bone homeostasis. Accumulating evidence has proven that miRNAs are widely involved in the regulation of bone homeostasis. However, whether miRNAs also regulate osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in T1DM mice is under exploration. In this study, miRNA microarray was utilized to screen the differentially expressed miRNAs, which uncovered that miR-214-3p potentially inhibited BMSCs osteogenic differentiation in T1DM mice. We found that high glucose suppressed BMSCs osteogenic differentiation with significant elevation of the miR-214-3p expression. Further study found that the osteogenic differentiation of BMSCs was inhibited by AgomiR-214-3p while enhanced by AntagomiR-214-3p in BMSCs supplemented with high glucose. Moreover, we found that miR-214-3p knockout T1DM mice were resistant to high-glucose-induced bone loss. These results provide a novel insight into an inhibitory role of high-glucose-induced miR-214-3p in BMSCs osteogenic differentiation both in vitro and in vivo. Molecular studies revealed that miR-214-3p inhibits BMSCs osteogenic differentiation by targeting the 3′-UTR of β-catenin, which was further corroborated in human bone specimens and BMSCs of T1DM patients. Taken together, our study discovered that miR-214-3p is a pivotal regulator of BMSCs osteogenic differentiation in T1DM mice. Our findings also suggest that miR-214-3p could be a potential target in the treatment of bone disorders in patients with T1DM.
Collapse
Affiliation(s)
- Rongze Wang
- 1Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yuanxu Zhang
- 2Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Fujun Jin
- 2Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.,3Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Gongchen Li
- 4Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yao Sun
- 4Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiaogang Wang
- 1Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.,5Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
307
|
Fayed HA, Barakat BM, Elshaer SS, Abdel-Naim AB, Menze ET. Antiosteoporotic activities of isoquercitrin in ovariectomized rats: Role of inhibiting hypoxia inducible factor-1 alpha. Eur J Pharmacol 2019; 865:172785. [PMID: 31712059 DOI: 10.1016/j.ejphar.2019.172785] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 01/13/2023]
Abstract
Postmenopausal osteoporosis is a common and disabling disorder that increases the risk of bone fractures due to estrogen deprivation; this can be simulated in rats by ovariectomy. Hypoxia inducible factor-1 alpha (HIF-1α) expression in osteoclasts predominantly leads to its activation increasing bone resorption. Premenopausal, estrogen prevents HIF-1α expression maintaining bone density. Unfortunately postmenopausal estrogen replacement therapy is not recommended due to its potential tumor development risk. Isoquercitrin, a common edible plants phytoestrogen, is known to inhibit HIF-1α. This study was conducted to investigate the potential antiosteoporotic activity of isoquercitrin (15, 30 and 60 mg/kg/day) in ovariectomized rats with reference to 17β-estradiol (25 mcg/kg/day). Animals were bilaterally ovariectomized to induce osteoporosis and one month later they were assigned into groups and administered isoquercitrin and 17β-estradiol for 8 weeks. Ovariectomy reduced lumbar compression strength, distorted bone microscopic architecture, inducing cartilage and trabecular dystrophy, and increased the markers of bone turnover (serum alkaline phosphatase and osteocalcin and urinary calcium, phosphorus and creatinine). It also increased the gene expression of HIF-1α and the levels of nuclear factor-kappa B (NF-κB) and decreased the expression of vascular endothelial growth factor (VEGF) and β-catenin in the femurs. Isoquercitrin was found to improve bone histological features, increase lumbar strength and improve most of the biochemical markers of bone turnover in a manner comparable to 17β-estradiol. Isoquercitrin also attenuated the increased HIF-1α expression while increased that of the VEGF and β-catenin. It also decreased the levels of NF-κB. Therefore isoquercitrin may be considered a safer alternative for managing osteoporosis.
Collapse
Affiliation(s)
- Hadeer A Fayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Bassant M Barakat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Shereen S Elshaer
- Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abbasia, Cairo, 11566, Egypt.
| |
Collapse
|
308
|
Luther J, Yorgan TA, Rolvien T, Ulsamer L, Koehne T, Liao N, Keller D, Vollersen N, Teufel S, Neven M, Peters S, Schweizer M, Trumpp A, Rosigkeit S, Bockamp E, Mundlos S, Kornak U, Oheim R, Amling M, Schinke T, David JP. Wnt1 is an Lrp5-independent bone-anabolic Wnt ligand. Sci Transl Med 2019; 10:10/466/eaau7137. [PMID: 30404864 DOI: 10.1126/scitranslmed.aau7137] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
WNT1 mutations in humans are associated with a new form of osteogenesis imperfecta and with early-onset osteoporosis, suggesting a key role of WNT1 in bone mass regulation. However, the general mode of action and the therapeutic potential of Wnt1 in clinically relevant situations such as aging remain to be established. Here, we report the high prevalence of heterozygous WNT1 mutations in patients with early-onset osteoporosis. We show that inactivation of Wnt1 in osteoblasts causes severe osteoporosis and spontaneous bone fractures in mice. In contrast, conditional Wnt1 expression in osteoblasts promoted rapid bone mass increase in developing young, adult, and aged mice by rapidly increasing osteoblast numbers and function. Contrary to current mechanistic models, loss of Lrp5, the co-receptor thought to transmit extracellular WNT signals during bone mass regulation, did not reduce the bone-anabolic effect of Wnt1, providing direct evidence that Wnt1 function does not require the LRP5 co-receptor. The identification of Wnt1 as a regulator of bone formation and remodeling provides the basis for development of Wnt1-targeting drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Julia Luther
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tim Rolvien
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lorenz Ulsamer
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Till Koehne
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.,Department of Orthodontics, University Medical Center Hamburg-Eppendorf, D 20246 Hamburg, Germany
| | - Nannan Liao
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Daniela Keller
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nele Vollersen
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Teufel
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stephanie Peters
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michaela Schweizer
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, D 20251 Hamburg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ), D 69120 Heidelberg, Germany
| | - Sebastian Rosigkeit
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Ernesto Bockamp
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Stefan Mundlos
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Max Planck Institute for Molecular Genetics, D 14195 Berlin, Germany
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, D 13353 Berlin, Germany.,Max Planck Institute for Molecular Genetics, D 14195 Berlin, Germany
| | - Ralf Oheim
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Jean-Pierre David
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
309
|
Wang X, Cornelis FMF, Lories RJ, Monteagudo S. Exostosin-1 enhances canonical Wnt signaling activity during chondrogenic differentiation. Osteoarthritis Cartilage 2019; 27:1702-1710. [PMID: 31330188 DOI: 10.1016/j.joca.2019.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 07/07/2019] [Accepted: 07/09/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Exostosin-1 (Ext1) encodes a glycosyltransferase required for heparan sulfate (HS) chain elongation in HS-proteoglycan biosynthesis. HS chains serve as binding partners for signaling proteins, affecting their distribution and activity. The Wnt/β-catenin pathway emerged as critical regulator of chondrogenesis. Yet, how EXT1 and HS affect Wnt/β-catenin signaling during chondrogenesis remains unexplored. METHOD Ext1 was stably knocked-down or overexpressed in ATDC5 chondrogenic cells cultured as micromasses. HS content was determined using ELISA. Chondrogenic markers Sox9, Col2a1, Aggrecan, and Wnt direct target gene Axin2 were measured by RT-qPCR. Proteoglycan content was evaluated by Alcian blue and DMMB assay, canonical Wnt signaling activation by β-catenin Western blot and TOP/FOP assay. ATDC5 cells and human articular chondrocytes were treated with Wnt activators CHIR99021 and recombinant WNT3A. RESULTS Ext1 knock-down reduced HS, and increased chondrogenic markers and proteoglycan accumulation. Ext1 knock-down reduced active Wnt/β-catenin signaling. Conversely, Ext1 overexpressing cells, with higher HS content, showed decreased chondrogenic differentiation and enhanced Wnt/β-catenin signaling. Wnt/β-catenin signaling activation led to a down-regulation of Ext1 expression in ATDC5 cells and in human articular chondrocytes. CONCLUSIONS EXT1 affects chondrogenic differentiation of precursor cells, in part via changes in the activity of Wnt/β-catenin signaling. Wnt/β-catenin signaling controls Ext1 expression, suggesting a regulatory loop between EXT1 and Wnt/β-catenin signaling during chondrogenesis.
Collapse
Affiliation(s)
- X Wang
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| | - F M F Cornelis
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| | - R J Lories
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium.
| | - S Monteagudo
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.
| |
Collapse
|
310
|
Yanai R, Tetsuo F, Ito S, Itsumi M, Yoshizumi J, Maki T, Mori Y, Kubota Y, Kajioka S. Extracellular calcium stimulates osteogenic differentiation of human adipose-derived stem cells by enhancing bone morphogenetic protein-2 expression. Cell Calcium 2019; 83:102058. [DOI: 10.1016/j.ceca.2019.102058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 06/19/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022]
|
311
|
Sun X, Zhang R, Liu M, Chen H, Chen L, Luo F, Zhang D, Huang J, Li F, Ni Z, Qi H, Su N, Jin M, Yang J, Tan Q, Du X, Chen B, Huang H, Chen S, Yin L, Xu X, Deng C, Luo L, Xie Y, Chen L. Rmrp Mutation Disrupts Chondrogenesis and Bone Ossification in Zebrafish Model of Cartilage-Hair Hypoplasia via Enhanced Wnt/β-Catenin Signaling. J Bone Miner Res 2019; 34:2101-2116. [PMID: 31237961 DOI: 10.1002/jbmr.3820] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 01/09/2023]
Abstract
Cartilage-hair hypoplasia (CHH) is an autosomal recessive metaphyseal chondrodysplasia characterized by bone dysplasia and many other highly variable features. The gene responsible for CHH is the RNA component of the mitochondrial RNA-processing endoribonuclease (RMRP) gene. Currently, the pathogenesis of osteochondrodysplasia and extraskeletal manifestations in CHH patients remains incompletely understood; in addition, there are no viable animal models for CHH. We generated an rmrp KO zebrafish model to study the developmental mechanisms of CHH. We found that rmrp is required for the patterning and shaping of pharyngeal arches. Rmrp mutation inhibits the intramembranous ossification of skull bones and promotes vertebrae ossification. The abnormalities of endochondral bone ossification are variable, depending on the degree of dysregulated chondrogenesis. Moreover, rmrp mutation inhibits cell proliferation and promotes apoptosis through dysregulating the expressions of cell-cycle- and apoptosis-related genes. We also demonstrate that rmrp mutation upregulates canonical Wnt/β-catenin signaling; the pharmacological inhibition of Wnt/β-catenin could partially alleviate the chondrodysplasia and increased vertebrae mineralization in rmrp mutants. Our study, by establishing a novel zebrafish model for CHH, partially reveals the underlying mechanism of CHH, hence deepening our understanding of the role of rmrp in skeleton development.
Collapse
Affiliation(s)
- Xianding Sun
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Mi Liu
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hangang Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dali Zhang
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fangfang Li
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhenhong Ni
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Bo Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Haiyang Huang
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shuai Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Xiaoling Xu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, Chongqing, 400715, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| |
Collapse
|
312
|
Yu SM, Han Y, Kim SJ. Simvastatin induces differentiation in rabbit articular chondrocytes via Wnt/β-catenin pathway. Eur J Pharmacol 2019; 863:172672. [DOI: 10.1016/j.ejphar.2019.172672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 09/05/2019] [Accepted: 09/18/2019] [Indexed: 01/31/2023]
|
313
|
Abstract
Osteoblasts are specialized mesenchymal cells that synthesize bone matrix and coordinate the mineralization of the skeleton. These cells work in harmony with osteoclasts, which resorb bone, in a continuous cycle that occurs throughout life. The unique function of osteoblasts requires substantial amounts of energy production, particularly during states of new bone formation and remodelling. Over the last 15 years, studies have shown that osteoblasts secrete endocrine factors that integrate the metabolic requirements of bone formation with global energy balance through the regulation of insulin production, feeding behaviour and adipose tissue metabolism. In this article, we summarize the current understanding of three osteoblast-derived metabolic hormones (osteocalcin, lipocalin and sclerostin) and the clinical evidence that suggests the relevance of these pathways in humans, while also discussing the necessity of specific energy substrates (glucose, fatty acids and amino acids) to fuel bone formation and promote osteoblast differentiation.
Collapse
Affiliation(s)
- Naomi Dirckx
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Megan C Moorer
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Baltimore Veterans Administration Medical Center, Baltimore, MD, USA.
| |
Collapse
|
314
|
Hung CC, Chaya A, Liu K, Verdelis K, Sfeir C. The role of magnesium ions in bone regeneration involves the canonical Wnt signaling pathway. Acta Biomater 2019; 98:246-255. [PMID: 31181262 DOI: 10.1016/j.actbio.2019.06.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 05/28/2019] [Accepted: 06/03/2019] [Indexed: 12/14/2022]
Abstract
Magnesium (Mg)-based implants have become of interest to both academia and the medical industry. The attraction largely is due to Mg's biodegradability and ability to enhance bone healing and formation. However, the underlying mechanism of how Mg regulates osteogenesis is still unclear. Based on our previous in vivo and molecular signaling work demonstrating the osteogenic effect of Mg, the current study aims to extend this work at the molecular level especially that we also observed and quantified mineral deposits in the bone marrow space in a rabbit ulna fracture model with Mg plates and screws. Histological analysis and quantitative results of micro-CT showed mineralized deposition and a significant increase in bone volume at 8 weeks and 16 weeks post-operative. These in vivo results led us to focus on studying the effect of Mg2+ on human bone marrow stromal cells (hBMSCs). The data presented in this manuscript demonstrate the activation of the canonical Wnt signaling pathway in hBMSCs when treated with 10 mM Mg2+. With additional Mg2+ present, the protein expression of active β-catenin was significantly increased to a level similar to that of the positive control. Immunocytochemistry and the increased expression of LEF1 and Dkk1, downstream target genes that are controlled directly by active β-catenin, demonstrated the protein translocation and the activation of transcription. Taken together, these data suggest that Mg2+ induces an osteogenic effect in the bone marrow space by activating the canonical Wnt signaling pathway, which in turn causes BMSCs to differentiate toward the osteoblast lineage. STATEMENT OF SIGNIFICANCE: Magnesium (Mg)-based alloys are being studied to be used in the field of implantable medical devices due to its natural biodegradability and the potential ability to promote bone regeneration. Despite many in vivo studies that demonstrated an increased new bone growth by implanting Mg-based devices, the underlying mechanism of this effect is still unclear. In order to safely use Mg-based implants on human and better control the osteogenic effect, it is necessary to understand the corresponding cellular response in the targeted area. The present study provides the rationale to study Mg ions on bone marrow stromal cells and shows the activation of canonical Wnt signaling pathway that promotes osteogenesis by in vivo and in vitro approaches.
Collapse
Affiliation(s)
- Chu-Chih Hung
- The Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amy Chaya
- The Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai Liu
- The Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA
| | - Konstantinos Verdelis
- The Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA; Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Endodontics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charles Sfeir
- The Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Oral Biology, University of Pittsburgh, Pittsburgh, PA, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Periodontics and Preventive Dentistry, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
315
|
Marín-Llera JC, Garciadiego-Cázares D, Chimal-Monroy J. Understanding the Cellular and Molecular Mechanisms That Control Early Cell Fate Decisions During Appendicular Skeletogenesis. Front Genet 2019; 10:977. [PMID: 31681419 PMCID: PMC6797607 DOI: 10.3389/fgene.2019.00977] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/13/2019] [Indexed: 12/02/2022] Open
Abstract
The formation of the vertebrate skeleton is orchestrated in time and space by a number of gene regulatory networks that specify and position all skeletal tissues. During embryonic development, bones have two distinct origins: bone tissue differentiates directly from mesenchymal progenitors, whereas most long bones arise from cartilaginous templates through a process known as endochondral ossification. Before endochondral bone development takes place, chondrocytes form a cartilage analgen that will be sequentially segmented to form joints; thus, in the cartilage template, either the cartilage maturation programme or the joint formation programme is activated. Once the cartilage differentiation programme starts, the growth plate begins to form. In contrast, when the joint formation programme is activated, a capsule begins to form that contains special articular cartilage and synovium to generate a functional joint. In this review, we will discuss the mechanisms controlling the earliest molecular events that regulate cell fate during skeletogenesis in long bones. We will explore the initial processes that lead to the recruitment of mesenchymal stem/progenitor cells, the commitment of chondrocyte lineages, and the formation of skeletal elements during morphogenesis. Thereafter, we will review the process of joint specification and joint morphogenesis. We will discuss the links between transcription factor activity, cell–cell interactions, cell–extracellular matrix interactions, growth factor signalling, and other molecular interactions that control mesenchymal stem/progenitor cell fate during embryonic skeletogenesis.
Collapse
Affiliation(s)
- Jessica Cristina Marín-Llera
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | | | - Jesús Chimal-Monroy
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
316
|
Autosomal-Recessive Mutations in MESD Cause Osteogenesis Imperfecta. Am J Hum Genet 2019; 105:836-843. [PMID: 31564437 DOI: 10.1016/j.ajhg.2019.08.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/16/2019] [Indexed: 11/21/2022] Open
Abstract
Osteogenesis imperfecta (OI) comprises a genetically heterogeneous group of skeletal fragility diseases. Here, we report on five independent families with a progressively deforming type of OI, in whom we identified four homozygous truncation or frameshift mutations in MESD. Affected individuals had recurrent fractures and at least one had oligodontia. MESD encodes an endoplasmic reticulum (ER) chaperone protein for the canonical Wingless-related integration site (WNT) signaling receptors LRP5 and LRP6. Because complete absence of MESD causes embryonic lethality in mice, we hypothesized that the OI-associated mutations are hypomorphic alleles since these mutations occur downstream of the chaperone activity domain but upstream of ER-retention domain. This would be consistent with the clinical phenotypes of skeletal fragility and oligodontia in persons deficient for LRP5 and LRP6, respectively. When we expressed wild-type (WT) and mutant MESD in HEK293T cells, we detected WT MESD in cell lysate but not in conditioned medium, whereas the converse was true for mutant MESD. We observed that both WT and mutant MESD retained the ability to chaperone LRP5. Thus, OI-associated MESD mutations produce hypomorphic alleles whose failure to remain within the ER significantly reduces but does not completely eliminate LRP5 and LRP6 trafficking. Since these individuals have no eye abnormalities (which occur in individuals completely lacking LRP5) and have neither limb nor brain patterning defects (both of which occur in mice completely lacking LRP6), we infer that bone mass accrual and dental patterning are more sensitive to reduced canonical WNT signaling than are other developmental processes. Biologic agents that can increase LRP5 and LRP6-mediated WNT signaling could benefit individuals with MESD-associated OI.
Collapse
|
317
|
Catala M, Khonsari RH, Paternoster G, Arnaud É. [Development and growth of the vault of the skull]. Neurochirurgie 2019; 65:210-215. [PMID: 31586575 DOI: 10.1016/j.neuchi.2019.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/20/2019] [Accepted: 09/21/2019] [Indexed: 02/03/2023]
Abstract
The vault of the skull is a region of the neurocranium formed by a process of membranous ossification. It consists of several bones: frontal bone, parietal bone, squamous part of the temporal bone, lamina ascendens of the sphenoid, and interparietal bone. The embryological origin of the bones of the skull vault is still the subject of controversy. This can be explained by the different animal models used for these purposes, but also by the various techniques applied to this problem. At all events, it seems that the cells of the neural crest generate some of the bones of the vault and that the others are derived from the mesoderm. This uncertainty should lead readers to be extremely cautious before using the presumptive maps published in the literature. Several tissues interact with osteo-progenitor cells: neural tube, surface ectoderm and dura mater. Analysis of genes in which mutations lead to abnormalities of the skull vault has partly revealed the molecular interactions. These are very complex and are the field of very numerous experimental investigations. In the relatively near future, we can hope to discover some of the molecular networks leading to the formation of these bony structures.
Collapse
Affiliation(s)
- M Catala
- UMR biologie du développement (Sorbonne université, CNRS, Inserm, IBPS), Sorbonne université (site Pierre-et-Marie-Curie), 9, quai Saint-Bernard, bâtiment C, 75252 Paris cedex 05, France.
| | - R H Khonsari
- Service de chirurgie maxillo-faciale et plastique, centre de référence maladies rares MAFACE, filière maladies rares CRANIOST, université Sorbonne Paris Cité, université Paris Descartes, hôpital Necker-Enfants-Malades, Assistance publique-Hôpitaux de Paris, Paris, France
| | - G Paternoster
- Service de neurochirurgie pédiatrique, hôpital Necker-Enfants-Malades, Assistance publique-Hôpitaux de Paris, Paris, France
| | - É Arnaud
- 34, avenue d'Eylau, Paris, France
| |
Collapse
|
318
|
Zhou Y, Lin J, Shao J, Zuo Q, Wang S, Wolff A, Nguyen DT, Rintoul L, Du Z, Gu Y, Peng YY, Ramshaw JAM, Long X, Xiao Y. Aberrant activation of Wnt signaling pathway altered osteocyte mineralization. Bone 2019; 127:324-333. [PMID: 31260814 DOI: 10.1016/j.bone.2019.06.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 12/20/2022]
Abstract
Mineralization of bone is a dynamic process, involving a complex interplay between cells, secreted macromolecules, signaling pathways, and enzymatic reactions; the dysregulation of bone mineralization may lead to serious skeletal disorders, including hypophosphatemic rickets, osteoporosis, and rheumatoid arthritis. Very few studies have reported the role of osteocytes - the most abundant bone cells in the skeletal system and the major orchestrators of bone remodeling in bone mineralization, which is owed to their nature of being deeply embedded in the mineralized bone matrix. The Wnt/β-catenin signaling pathway is actively involved in various life processes including osteogenesis; however, the role of Wnt/β-catenin signaling in the terminal mineralization of bone, especially in the regulation of osteocytes, is largely unknown. This research demonstrates that during the terminal mineralization process, the Wnt/β-catenin pathway is downregulated, and when Wnt/β-catenin signaling is activated in osteocytes, dendrite development is suppressed and the expression of dentin matrix protein 1 (DMP1) is inhibited. Aberrant activation of Wnt/β-catenin signaling in osteocytes leads to the spontaneous deposition of extra-large mineralized nodules on the surface of collagen fibrils. The altered mineral crystal structure and decreased bonding force between minerals and the organic matrix indicate the inferior integration of minerals and collagen. In conclusion, Wnt/β-catenin signaling plays a critical role in the terminal differentiation of osteocytes and as such, targeting Wnt/β-catenin signaling in osteocytes may serve as a potential therapeutic approach for the management of bone-related diseases.
Collapse
Affiliation(s)
- Yinghong Zhou
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 51050, China; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Jinying Lin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Implantology, Affiliated Stomatological Hospital of Xiamen Medical College, Fujian 361000, China; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Jin Shao
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Qiliang Zuo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Implantology, Affiliated Stomatological Hospital of Xiamen Medical College, Fujian 361000, China; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia
| | - Shengfang Wang
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Annalena Wolff
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Dung Trung Nguyen
- Department of Engineering and Computer Science, Seattle Pacific University, Seattle, WA 98119, USA.
| | - Llew Rintoul
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Zhibin Du
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Yuantong Gu
- School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Yong Y Peng
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia.
| | - John A M Ramshaw
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| | - Yin Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 51050, China; School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; The Australia-China Centre for Tissue Engineering and Regenerative Medicine (ACCTERM), Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia.
| |
Collapse
|
319
|
Diederichs S, Tonnier V, März M, Dreher SI, Geisbüsch A, Richter W. Regulation of WNT5A and WNT11 during MSC in vitro chondrogenesis: WNT inhibition lowers BMP and hedgehog activity, and reduces hypertrophy. Cell Mol Life Sci 2019; 76:3875-3889. [PMID: 30980110 PMCID: PMC11105731 DOI: 10.1007/s00018-019-03099-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022]
Abstract
Re-directing mesenchymal stromal cell (MSC) chondrogenesis towards a non-hypertrophic articular chondrocyte-(AC)-like phenotype is important for improving articular cartilage neogenesis to enhance clinical cartilage repair strategies. This study is the first to demonstrate that high levels of non-canonical WNT5A followed by WNT11 and LEF1 discriminated MSC chondrogenesis from AC re-differentiation. Moreover, β-catenin seemed incompletely silenced in differentiating MSCs, which altogether suggested a role for WNT signaling in hypertrophic MSC differentiation. WNT inhibition with the small molecule IWP-2 supported MSC chondrogenesis according to elevated proteoglycan deposition and reduced the characteristic upregulation of BMP4, BMP7 and their target ID1, as well as IHH and its target GLI1 observed during endochondral differentiation. Along with the pro-hypertrophic transcription factor MEF2C, multiple hypertrophic downstream targets including IBSP and alkaline phosphatase activity were reduced by IWP-2, demonstrating that WNT activity drives BMP and hedgehog upregulation, and MSC hypertrophy. WNT inhibition almost matched the strong anti-hypertrophic capacity of pulsed parathyroid hormone-related protein application, and both outperformed suppression of BMP signaling with dorsomorphin, which also reduced cartilage matrix deposition. Yet, hypertrophic marker expression under IWP-2 remained above AC level, and in vivo mineralization and ectopic bone formation were reduced but not eliminated. Overall, the strong anti-hypertrophic effects of IWP-2 involved inhibition but not silencing of pro-hypertrophic BMP and IHH pathways, and more advanced silencing of WNT activity as well as combined application of IHH or BMP antagonists should next be considered to install articular cartilage neogenesis from human MSCs.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Veronika Tonnier
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie März
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Geisbüsch
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
320
|
Differential Gene Expression in Articular Cartilage and Subchondral Bone of Neonatal and Adult Horses. Genes (Basel) 2019; 10:genes10100745. [PMID: 31557843 PMCID: PMC6826356 DOI: 10.3390/genes10100745] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/11/2019] [Accepted: 09/21/2019] [Indexed: 12/26/2022] Open
Abstract
Skeletogenesis is complex and incompletely understood. Derangement of this process likely underlies developmental skeletal pathologies. Examination of tissue-specific gene expression may help elucidate novel skeletal developmental pathways that could contribute to disease risk. Our aim was to identify and functionally annotate differentially expressed genes in equine neonatal and adult articular cartilage (AC) and subchondral bone (SCB). RNA was sequenced from healthy AC and SCB from the fetlock, hock, and stifle joints of 6 foals (≤4 weeks of age) and six adults (8–12 years of age). There was distinct clustering by age and tissue type. After differential expression analysis, functional annotation and pathway analysis were performed using PANTHER and Reactome. Approximately 1115 and 3574 genes were differentially expressed between age groups in AC and SCB, respectively, falling within dozens of overrepresented gene ontology terms and enriched pathways reflecting a state of growth, high metabolic activity, and tissue turnover in the foals. Enriched pathways were dominated by those related to extracellular matrix organization and turnover, and cell cycle and signal transduction. Additionally, we identified enriched pathways related to neural development and neurotransmission in AC and innate immunity in SCB. These represent novel potential mechanisms for disease that can be explored in future work.
Collapse
|
321
|
Tsang KY, Cheah KS. The extended chondrocyte lineage: implications for skeletal homeostasis and disorders. Curr Opin Cell Biol 2019; 61:132-140. [PMID: 31541943 DOI: 10.1016/j.ceb.2019.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 01/14/2023]
Abstract
Endochondral bone formation relies on a finely controlled sequence of chondrocyte proliferation, maturation and hypertrophy that establishes the growth plate which, combined with the deposition of bone upon the cartilage template, mediates longitudinal skeletal growth. Recent lineage studies support a chondrocyte-osteoblast differentiation continuum and the presence of skeletal stem cells within cartilage. The biological significance of the lineage extension and the mechanisms controlling the process are unclear. In this review, we describe recent work on the extended chondrocyte-osteoblast lineage and its contribution to the development, growth and repair of bone and to bone disorders that provides insight into the process and the molecular controls involved. The implications for skeletal homeostasis are discussed.
Collapse
Affiliation(s)
- Kwok Yeung Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kathryn Se Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
322
|
Schlesinger PH, Blair HC, Beer Stolz D, Riazanski V, Ray EC, Tourkova IL, Nelson DJ. Cellular and extracellular matrix of bone, with principles of synthesis and dependency of mineral deposition on cell membrane transport. Am J Physiol Cell Physiol 2019; 318:C111-C124. [PMID: 31532718 DOI: 10.1152/ajpcell.00120.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bone differs from other connective tissues; it is isolated by a layer of osteoblasts that are connected by tight and gap junctions. This allows bone to create dense lamellar type I collagen, control pH, mineral deposition, and regulate water content forming a compact and strong structure. New woven bone formed after degradation of mineralized cartilage is rapidly degraded and resynthesized to impart structural order for local bone strength. Ossification is regulated by thickness of bone units and by patterning via bone morphogenetic receptors including activin, other bone morphogenetic protein receptors, transforming growth factor-β receptors, all part of a receptor superfamily. This superfamily interacts with receptors for additional signals in bone differentiation. Important features of the osteoblast environment were established using recent tools including osteoblast differentiation in vitro. Osteoblasts deposit matrix protein, over 90% type I collagen, in lamellae with orientation alternating parallel or orthogonal to the main stress axis of the bone. Into this organic matrix, mineral is deposited as hydroxyapatite. Mineral matrix matures from amorphous to crystalline hydroxyapatite. This process includes at least two-phase changes of the calcium-phosphate mineral as well as intermediates involving tropocollagen fibrils to form the bone composite. Beginning with initiation of mineral deposition, there is uncertainty regarding cardinal processes, but the driving force is not merely exceeding the calcium-phosphate solubility product. It occurs behind a epithelial-like layer of osteoblasts, which generate phosphate and remove protons liberated during calcium-phosphate salt deposition. The forming bone matrix is discontinuous from the general extracellular fluid. Required adjustment of ionic concentrations and water removal from bone matrix are important details remaining to be addressed.
Collapse
Affiliation(s)
| | - Harry C Blair
- Veterans Affairs Medical Center, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Donna Beer Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vladimir Riazanski
- Department of Neurobiology, Pharmacology, and Physiology, University of Chicago, Chicago, Illinois
| | - Evan C Ray
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Irina L Tourkova
- Veterans Affairs Medical Center, Pittsburgh, Pennsylvania.,Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Deborah J Nelson
- Department of Neurobiology, Pharmacology, and Physiology, University of Chicago, Chicago, Illinois
| |
Collapse
|
323
|
Hosseinpour S, He Y, Nanda A, Ye Q. MicroRNAs Involved in the Regulation of Angiogenesis in Bone Regeneration. Calcif Tissue Int 2019; 105:223-238. [PMID: 31175386 DOI: 10.1007/s00223-019-00571-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/01/2019] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) as a newly founded and thriving non-coding endogenous class of molecules which regulate many cellular pathways after transcription have been extensively investigated in regenerative medicine. In this systematic review, we sought to analyze miRNAs-mediated therapeutic approaches for influencing angiogenesis in bone tissue/bone regeneration. An electronic search in MEDLINE, Scopus, EMBASE, Cochrane library, web of science, and google scholar with no time limit were done on English publications. All types of original articles which a miRNA for angiogenesis in bone regeneration were included in our review. In the process of reviewing, we used PRISMA guideline and, SYRCLE's and science in risk assessment and policy tools for analyzing risk of bias. Among 751 initial retrieved records, 16 studies met the inclusion criteria and were fully assessed in this review. 275 miRNAs, one miRNA 195~497 cluster, and one Cysteine-rich 61 short hairpin RNA were differentially expressed during bone regeneration with 24 predicted targets reported in these studies. Among these miRNAs, miRNA-7b, -9, -21, -26a, -27a, -210, -378, -195~497 cluster, -378 and -675 positively promoted both angiogenesis and osteogenesis, whereas miRNA-10a, -222 and -494 inhibited both processes. The most common target was vasculoendothelial growth factor-signaling pathway. Recent evidence has demonstrated that miRNAs actively participated in angio-osteogenic coupling that can improve their therapeutic potentials for the treatment of bone-related diseases and bone regeneration. However, there is still need for further research to unravel the exact mechanisms.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Yan He
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Ashwin Nanda
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Qingsong Ye
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
324
|
Liao F, Liu Y, Liu HH, Hu J, Zhao S, Yang SM. [Effect of Angelica sinensis polysaccharide on the osteogenic differentiation of bone marrow mesenchymal stem cells of rats with high glucose levels]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:193-199. [PMID: 31168987 DOI: 10.7518/hxkq.2019.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study aims to evaluate the effect of Angelica sinensis polysaccharide (ASP) on the osteogenic differentiation of the bone marrow mesenchymal stem cells (BMSCs) of rats with high glucose levels. METHODS Rat BMSCs were isolated and identified by osteogenic and adipogenic differentiation. Then, the BMSCs were divided into three groups as follows: normal control group (5.5 mmol·L⁻¹ glucose), high glucose group (25.5 mmol·L⁻¹ glucose), and ASP+high glucose group (25.5 mmol·L⁻¹ glucose +40 mg·L⁻¹ ASP). The proliferation activities of the BMSCs were detected by CCK8. Alizarin red staining, and alkaline phosphatase activity were used in the examination of osteogenic activity. Quantitative real time-polymerase chain reaction was used to detect the expression levels of the osteogenic genes (Runx2, Osx, OCN, Col-Ⅰ) and the key factors of Wnt/β-catenin signal pathway (CyclinD1, β-catenin). In vivo, a type 2 diabetes rat model was established. The rats were divided into three groups, namely, the normal control group (normal rats), diabetes group (diabetic rats), diabetes+ASP group (diabetic rats, ASP feeding). Then, the tibia bone defect was established. The repair of bone defects in each group was observed through histological examination. RESULTS The proliferation of BMSCs was higher in the high glucose group and ASP+high glucose group than in the normal control group (P<0.05). No significant difference was observed between the high glucose group and ASP+high glucose group (P>0.05). The number of calcium nodules of BMSCs; alkaline phosphatase activity; and the mRNA expression of Runx2, OCN, Osx, Col-Ⅰ, CyclinD1, β-catenin in the high glucose group were lower than those in the normal control and ASP+high glucose groups (P<0.05). No significant difference was observed between the normal control and ASP+high glucose groups (P>0.05). The bone mass was significantly lower in the bone defect of the diabetes group than in the bone defect of the normal control or diabetes+ASP group (P<0.05). No statistical difference was found between the normal control and diabetes+ASP groups (P>0.05). CONCLUSIONS ASP can promote the osteogenic differentiation of rat BMSCs under high glucose culture and induce bone regeneration in rats with type 2 diabetes. These features may be related to the activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Feng Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province & Key Laboratory of Oral Biomedicine Wuhan University, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hang-Hang Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jian Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province & Key Laboratory of Oral Biomedicine Wuhan University, Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China
| | - Shuang Zhao
- Dept. of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan 250001, China
| | - Shi-Mao Yang
- Dept. of Oral and Maxillofacial Surgery, Jinan Stomatology Hospital, Jinan 250001, China
| |
Collapse
|
325
|
Regulation of Hedgehog signaling Offers A Novel Perspective for Bone Homeostasis Disorder Treatment. Int J Mol Sci 2019; 20:ijms20163981. [PMID: 31426273 PMCID: PMC6719140 DOI: 10.3390/ijms20163981] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023] Open
Abstract
The hedgehog (HH) signaling pathway is central to the regulation of bone development and homeostasis. HH signaling is not only involved in osteoblast differentiation from bone marrow mesenchymal stem cells (BM-MSCs), but also acts upstream within osteoblasts via the OPG/RANK/RANKL axis to control the expression of RANKL. HH signaling has been found to up-regulate parathyroid hormone related protein (PTHrP) expression in osteoblasts, which in turn activates its downstream targets nuclear factor of activated T cells (NFAT) and cAMP responsive element binding protein (CREB), and as a result CREB and NFAT cooperatively increase RANKL expression and osteoclastogenesis. Osteoblasts must remain in balance with osteoclasts in order to avoid excessive bone formation or resorption, thereby maintaining bone homeostasis. This review systemically summarizes the mechanisms whereby HH signaling induces osteoblast development and controls RANKL expression through PTHrP in osteoblasts. Proper targeting of HH signaling may offer a therapeutic option for treating bone homeostasis disorders.
Collapse
|
326
|
Li L, Rao S, Cheng Y, Zhuo X, Deng C, Xu N, Zhang H, Yang L. Microbial osteoporosis: The interplay between the gut microbiota and bones via host metabolism and immunity. Microbiologyopen 2019; 8:e00810. [PMID: 31001921 PMCID: PMC6692530 DOI: 10.1002/mbo3.810] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/15/2023] Open
Abstract
The complex relationship between intestinal microbiota and host is a novel field in recent years. A large number of studies are being conducted on the relationship between intestinal microbiota and bone metabolism. Bone metabolism consisted of bone absorption and formation exists in the whole process of human growth and development. The nutrient components, inflammatory factors, and hormone environment play important roles in bone metabolism. Recently, intestinal microbiota has been found to influence bone metabolism via influencing the host metabolism, immune function, and hormone secretion. Here, we searched relevant literature on Pubmed and reviewed the effect of intestinal microbiota on bone metabolism through the three aspects, which may provide new ideas and targets for the clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Lishan Li
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shitao Rao
- School of Biomedical SciencesCUHKShatin, N.THong Kong SARChina
| | - Yanzhen Cheng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Xiaoyun Zhuo
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Caihong Deng
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ningning Xu
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Hua Zhang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Li Yang
- Department of endocrinology and metabolismZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
327
|
Ma ZP, Zhang ZF, Yang YF, Yang Y. Sesamin Promotes Osteoblastic Differentiation and Protects Rats from Osteoporosis. Med Sci Monit 2019; 25:5312-5320. [PMID: 31314750 PMCID: PMC6659468 DOI: 10.12659/msm.915529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Osteoporosis is a common osteopathy, resulting in fractures, especially in elder people. Sesamin has many pharmacological effects, including supplying calcium. However, how sesamin might prevent osteoporosis is still under study. Material/Methods Bone marrow stromal cells (BMSCs) extracted from rat femur were induced for osteoblastic differentiation. Cell proliferation, alkaline phosphatase (ALP), osterix (OSX), SRY-box 9 (SOX9), runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), β-catenin, low density lipoprotein receptor-related protein 5 (LRP5), and glycogen synthase kinase-3β (GSK-3β) levels in BMSCs were detected in the presence or absence of sesamin (1 μM or 10 μM). In addition, FH535 (1 μM) was used to silence Wnt/β-catenin in vitro. Ovariectomized (OVX) rats were established and intragastrically administrated sesamin (80 mg/kg), and then the rat bones were analyzed by micro-computed tomography. Osteocalcin and collagen type I were measured in the rat femurs. Results Sesamin had no influence on BMSC proliferation. Higher sesamin concentration promoted Wnt/β-catenin activity and enhanced more expressions of ALP, OSX, SOX9, RUNX2, and OCN, gradually and significantly (P<0.05). Silencing Wnt/β-catenin weakened the enhancement on RUNX2 and OCN expression. Sesamin (80 mg/kg) promoted bone structure in ovariectomized rats, and significantly enhanced osteocalcin and collage type I expression (P<0.05). Conclusions Sesamin promoted osteoblastic differentiation of rat BMSCs by regulating the Wnt/β-catenin pathway, and improved rat bone structure. Sesamin could have therapeutic and preventive effects on osteoporosis.
Collapse
Affiliation(s)
- Zhong-Ping Ma
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Zhi-Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Yi-Feng Yang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| | - Yun Yang
- Department of Orthopedics, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China (mainland)
| |
Collapse
|
328
|
Kleber M, Ntanasis-Stathopoulos I, Dimopoulos MA, Terpos E. Monoclonal antibodies against RANKL and sclerostin for myeloma-related bone disease: can they change the standard of care? Expert Rev Hematol 2019; 12:651-663. [PMID: 31268745 DOI: 10.1080/17474086.2019.1640115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Over 80% of the patients with multiple myeloma (MM) develop myeloma bone disease (MBD) during the disease course. The clinical consequences include serious skeletal-related events (SRE) that impact survival and quality of life. Bisphosphonates are the mainstay in the treatment of MBD. Currently, new therapeutic strategies are being introduced and broaden the therapeutic options in MBD. Areas covered: The purpose of this review is to summarize the current clinical management of MBD and present novel data regarding monoclonal antibodies against the receptor activator of NF-kappa B ligand (RANKL) and sclerostin that may change the clinical practice. Expert opinion: Our better understanding of the pathophysiology of MBD has identified several factors as potential therapeutic targets. Recent data have shown that the RANKL inhibitor denosumab constitutes a new promising option. The non-inferiority compared with bisphosphonates in terms of SRE prevention, the potential survival benefit, the convenience of subcutaneous administration, and the favorable toxicity profile makes denosumab a valuable alternative for physicians in the current treatment of MBD. Anti-sclerostin antibodies are currently under clinical development. Further investigations are needed to address open questions in the field including the value of anabolic agents combined with anti-resorptive and anti-MM drugs in MBD.
Collapse
Affiliation(s)
- Martina Kleber
- a Division of Hematology, Department of Medicine, University Hospital Basel , Basel , Switzerland.,b Division of Internal Medicine, Department of Medicine, University Hospital Basel , Basel , Switzerland
| | - Ioannis Ntanasis-Stathopoulos
- c Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens , Athens , Greece
| | - Meletios A Dimopoulos
- c Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens , Athens , Greece
| | - Evangelos Terpos
- c Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
329
|
Wang Y, Chen H, Zhang H. Tanshinone IIA exerts beneficial effects on fracture healing in vitro and in vivo. Chem Biol Interact 2019; 310:108748. [PMID: 31306638 DOI: 10.1016/j.cbi.2019.108748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/04/2019] [Accepted: 07/12/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Fracture healing is a very important process after fracture. Tanshinone IIA (Tan IIA) has been reported to possess beneficial impact on osteoblasts growth. Our study investigated the effects of Tan IIA on fracture healing. METHODS In vitro, mouse pre-osteoblast MC3T3-E1 cells were treated with Tan IIA. Then, the protein levels of Runx2, Osx, Collagen I, JNK and c-Jun, alkaline phosphatase (ALP) activity and calcium deposition were detected, respectively. Furthermore, the roles of microRNA-424 (miR-424) and Bone morphogenetic protein 2 (BMP-2) in Tan IIA-caused MC3T3-E1 cell differentiation were probed. In vivo, mice open osteotomy at femur diaphysis model was established. The callus area, callus intensity, low-density bone volume/callus total volume (BV1/TV), tissue mineral density (TMD) and bone mineral density (BMD) were tested. RESULTS In vitro, Tan IIA promoted MC3T3-E1 cell differentiation via increasing the Runx2, Osx and collagen I expression, along with enhancing ALP activity and calcium deposition. In addition, Tan IIA activated JNK pathway in MC3T3-E1 cells, while inhibition of JNK pathway mitigated the Tan IIA-caused MC3T3-E1 cell differentiation. Moreover, Tan IIA declined the miR-424 expression in MC3T3-E1 cells. Overexpression of miR-424 also weakened the Tan IIA-caused MC3T3-E1 cell differentiation. BMP-2 was a target gene of miR-424. BMP-2 silence reversed the Tan IIA-caused activation of JNK pathway. In vivo, Tan IIA increased the callus area, callus intensity, BV1/TV, TMD and BMD. CONCLUSION Tan IIA could promote fracture healing. In vitro, Tan IIA promoted MC3T3-E1 cell differentiation might be via down-regulating miR-424, up-regulating BMP-2 and then activating JNK pathway.
Collapse
Affiliation(s)
- Yang Wang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin, China
| | - Hongyu Chen
- Department of Orthopaedics, Qingdao West Coast New Area Central Hospital, Qingdao, 266555, Shandong, China
| | - Hanyang Zhang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, 130000, Jilin, China.
| |
Collapse
|
330
|
Han Q, Wang X, Liao X, Han C, Yu T, Yang C, Li G, Han B, Huang K, Zhu G, Liu Z, Zhou X, Su H, Shang L, Gong Y, Song X, Peng T, Ye X. Diagnostic and prognostic value of WNT family gene expression in hepatitis B virus‑related hepatocellular carcinoma. Oncol Rep 2019; 42:895-910. [PMID: 31322232 PMCID: PMC6667889 DOI: 10.3892/or.2019.7224] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to investigate the diagnostic and prognostic value of Wingless-type MMTV integration site (WNT) gene family expression in patients with hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). The clinical data of the patients and gene expression levels were downloaded from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. Receiver operating characteristic curve analysis was used to investigate the diagnostic value of WNT genes. Cox proportional hazard regression analysis and Kaplan-Meier survival analysis were performed to evaluate the association of WNT gene expression level with overall survival (OS) and recurrence-free survival (RFS). A nomogram was constructed for the prediction of prognosis. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. Diagnostic receiver operating characteristic curve analysis suggested that WNT2 had a high diagnostic value, with an area under the curve (AUC) of >0.800 (P<0.0001, AUC=0.810, 95% CI: 0.767–0.852). Survival analysis indicated that the expression level of WNT1 was significantly associated with OS and RFS (adjusted P=0.033, adjusted HR=0.607, 95% CI: 0.384–0.960; and adjusted P=0.007, adjusted HR=0.592, 95% CI: 0.404–0.868, respectively). In the TCGA validation cohort, we also observed that WNT2 was significantly differentially expressed between HCC tissues and adjacent non-tumor tissues, and WNT1 was associated with both the OS and RFS of HCC. Therefore, through the GSE14520 HBV-related HCC cohort we concluded that WNT2 may serve as a diagnostic biomarker and WNT1 may serve as a prognostic biomarker. These results may also be extended to TCGA HCC verification cohort.
Collapse
Affiliation(s)
- Quanfa Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guanghui Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Bowen Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Liming Shang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yizhen Gong
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaowei Song
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
331
|
Sivaji K, Kannan RR, Nandhagopal S, Carlton Ranjith WA, Saleem S. Exogenous human beta amyloid peptide interferes osteogenesis through Sox9a in embryonic zebrafish. Mol Biol Rep 2019; 46:4975-4984. [PMID: 31264162 DOI: 10.1007/s11033-019-04948-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/26/2019] [Indexed: 12/30/2022]
Abstract
The two major hallmarks of Alzheimer's disease (AD) are beta-amyloid plaques and neurofibrillary tangles. Amyloid peptide aggregations in the brain cause loss of synaptic connections and subsequent neurotoxicity leading to neurodegeneration and memory deficits. However, the physiological effects of beta-amyloid on early embryonic development still remain unclear. Administration of human beta-amyloid peptide (1-42) through cerebrospinal ventricular injection was carried out at 24 hpf (hours post fertilization) and it was uptaken into the cellular layers of the early ventricular development without any plaque aggregation. Whole-mount Immunostaining of zebrafish embryos injected with the beta-amyloid at 60 hpf revealed the delay in Sox9a expression. Decreased level of cartilage to bone transformation rate in 15 dpf (days post fertilization) zebrafish was observed by differential staining. These results suggest the possible existence of a genetic relationship between extrinsic amyloid peptide and Sox9a expression. Thus, our results demonstrated that the human beta-amyloid influences bone development through Sox9a expression during osteogenesis in zebrafish.
Collapse
Affiliation(s)
- Kalaiarasi Sivaji
- Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | | | | | | | - Suraiya Saleem
- Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
332
|
Pierce JL, Ding KH, Xu J, Sharma AK, Yu K, Del Mazo Arbona N, Rodriguez-Santos Z, Bernard P, Bollag WB, Johnson MH, Hamrick MW, Begun DL, Shi XM, Isales CM, McGee-Lawrence ME. The glucocorticoid receptor in osteoprogenitors regulates bone mass and marrow fat. J Endocrinol 2019; 243:JOE-19-0230.R1. [PMID: 31370004 PMCID: PMC6938567 DOI: 10.1530/joe-19-0230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Excess fat within bone marrow is associated with lower bone density. Metabolic stressors such as chronic caloric restriction (CR) can exacerbate marrow adiposity, and increased glucocorticoid signaling and adrenergic signaling are implicated in this phenotype. The current study tested the role of glucocorticoid signaling in CR-induced stress by conditionally deleting the glucocorticoid receptor (GR) in bone marrow osteoprogenitors (Osx1-Cre) of mice subjected to CR and ad libitum diets. Conditional knockout of the GR (GR-CKO) reduced cortical and trabecular bone mass as compared to wildtype (WT) mice under both ad libitum and CR conditions. No interaction was detected between genotype and diet, suggesting that the GR is not required for CR-induced skeletal changes. The lower bone mass in GR-CKO mice, and the further suppression of bone by CR, resulted from suppressed bone formation. Interestingly, treatment with the -adrenergic receptor antagonist propranolol mildly but selectively improved metrics of cortical bone mass in GR-CKO mice during CR, suggesting interaction between adrenergic and glucocorticoid signaling pathways that affects cortical bone. GR-CKO mice dramatically increased marrow fat under both ad libitum and CR-fed conditions, and surprisingly propranolol treatment was unable to rescue CR-induced marrow fat in either WT or GR-CKO mice. Additionally, serum corticosterone levels were selectively elevated in GR-CKO mice with CR, suggesting the possibility of bone-hypothalamus-pituitary-adrenal crosstalk during metabolic stress. This work highlights the complexities of glucocorticoid and β-adrenergic signaling in stress-induced changes in bone mass, and the importance of GR function in suppressing marrow adipogenesis while maintaining healthy bone mass.
Collapse
Affiliation(s)
- Jessica L Pierce
- J Pierce, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Ke-Hong Ding
- K Ding, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Jianrui Xu
- J Xu, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Anuj K Sharma
- A Sharma, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Kanglun Yu
- K Yu, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | | | | | - Paul Bernard
- P Bernard, Pediatric Endocrine Specialists of Georgia, Pediatric Endocrine Specialists of Georgia, Duluth, United States
| | - Wendy B Bollag
- W Bollag, Department of Physiology, Medical College of Georgia, Augusta, GA 30912, United States
| | - Maribeth H Johnson
- M Johnson, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Mark W Hamrick
- M Hamrick, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Dana L Begun
- D Begun, Department of Orthopedic Surgery, Mayo Clinic, Rochester, United States
| | - Xing M Shi
- X Ming Shi, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Carlos M Isales
- C Isales, Neuroscience and Regenerative Medicine, Augusta University, Augusta, 30912, United States
| | | |
Collapse
|
333
|
Kaucka M, Adameyko I. Evolution and development of the cartilaginous skull: From a lancelet towards a human face. Semin Cell Dev Biol 2019; 91:2-12. [DOI: 10.1016/j.semcdb.2017.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 11/27/2017] [Accepted: 12/09/2017] [Indexed: 11/16/2022]
|
334
|
Li J, Ayoub A, Xiu Y, Yin X, Sanders JO, Mesfin A, Xing L, Yao Z, Boyce BF. TGFβ-induced degradation of TRAF3 in mesenchymal progenitor cells causes age-related osteoporosis. Nat Commun 2019; 10:2795. [PMID: 31243287 PMCID: PMC6595054 DOI: 10.1038/s41467-019-10677-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/22/2019] [Indexed: 01/08/2023] Open
Abstract
Inflammaging induces osteoporosis by promoting bone destruction and inhibiting bone formation. TRAF3 limits bone destruction by inhibiting RANKL-induced NF-κB signaling in osteoclast precursors. However, the role of TRAF3 in mesenchymal progenitor cells (MPCs) is unknown. Mice with TRAF3 deleted in MPCs develop early onset osteoporosis due to reduced bone formation and enhanced bone destruction. In young mice TRAF3 prevents β-catenin degradation in MPCs and maintains osteoblast formation. However, TRAF3 protein levels decrease in murine and human bone samples during aging when TGFβ1 is released from resorbing bone. TGFβ1 induces degradation of TRAF3 in murine MPCs and inhibits osteoblast formation through GSK-3β-mediated degradation of β-catenin. Thus, TRAF3 positively regulates MPC differentiation into osteoblasts. TRAF3 deletion in MPCs activated NF-κB RelA and RelB to promote RANKL expression and enhance bone destruction. We conclude that pharmacologic stabilization of TRAF3 during aging could treat/prevent age-related osteoporosis by inhibiting bone destruction and promoting bone formation.
Collapse
Affiliation(s)
- Jinbo Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Akram Ayoub
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Yan Xiu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - Xiaoxiang Yin
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Medical Imaging, Henan University First Affiliated Hospital, 357 Ximen Street, Kaifeng, 475001, Henan, P.R. China
| | - James O Sanders
- Department of Orthopaedics and Rehabilitation Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of North Carolina, Chapel Hill, NC, 27514, USA
| | - Addisu Mesfin
- Department of Orthopaedics and Rehabilitation Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
335
|
Wang F, Tarkkonen K, Nieminen‐Pihala V, Nagano K, Majidi RA, Puolakkainen T, Rummukainen P, Lehto J, Roivainen A, Zhang F, Mäkitie O, Baron R, Kiviranta R. Mesenchymal Cell-Derived Juxtacrine Wnt1 Signaling Regulates Osteoblast Activity and Osteoclast Differentiation. J Bone Miner Res 2019; 34:1129-1142. [PMID: 30690791 PMCID: PMC6850336 DOI: 10.1002/jbmr.3680] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/08/2019] [Accepted: 01/28/2019] [Indexed: 12/30/2022]
Abstract
Human genetic evidence demonstrates that WNT1 mutations cause osteogenesis imperfecta (OI) and early-onset osteoporosis, implicating WNT1 as a major regulator of bone metabolism. However, its main cellular source and mechanisms of action in bone remain elusive. We generated global and limb bud mesenchymal cell-targeted deletion of Wnt1 in mice. Heterozygous deletion of Wnt1 resulted in mild trabecular osteopenia due to decreased osteoblast function. Targeted deletion of Wnt1 in mesenchymal progenitors led to spontaneous fractures due to impaired osteoblast function and increased bone resorption, mimicking the severe OI phenotype in humans with homozygous WNT1 mutations. Importantly, we showed for the first time that Wnt1 signals strictly in a juxtacrine manner to induce osteoblast differentiation and to suppress osteoclastogenesis, in part via canonical Wnt signaling. In conclusion, mesenchymal cell-derived Wnt1, acting in short range, is an essential regulator of bone homeostasis and an intriguing target for therapeutic interventions for bone diseases. © 2019 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fan Wang
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | | | | | - Kenichi Nagano
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental MedicineHarvard UniversityBostonMAUSA
| | | | | | | | - Jemina Lehto
- Institute of BiomedicineUniversity of TurkuTurkuFinland
| | - Anne Roivainen
- Turku PET CentreUniversity of TurkuTurkuFinland
- Turku Center for Disease Modeling (TCDM)University of TurkuTurkuFinland
| | - Fu‐Ping Zhang
- Turku Center for Disease Modeling (TCDM)University of TurkuTurkuFinland
| | - Outi Mäkitie
- Folkhälsan Institute of GeneticsHelsinkiFinland
- Children's HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Roland Baron
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental MedicineHarvard UniversityBostonMAUSA
| | - Riku Kiviranta
- Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of Endocrinology, Division of MedicineUniversity of Turku and Turku University HospitalTurkuFinland
| |
Collapse
|
336
|
Abstract
PURPOSE OF REVIEW The goal of the review is to summarize the current knowledge on the process of chondrocyte-to-osteoblast transdifferentiation during endochondral bone formation and its potential implications in fracture healing and disease. RECENT FINDINGS Lineage tracing experiments confirmed the transdifferentiation of chondrocytes into osteoblasts. More recent studies lead to the discovery of molecules involved in this process, as well as to the hypothesis that these cells may re-enter a stem cell-like phase prior to their osteoblastic differentiation. This review recapitulates the current knowledge regarding chondrocyte transdifferentiating into osteoblasts, the developmental and postnatal events where transdifferentiation appears to be relevant, and the molecules implicated in this process.
Collapse
Affiliation(s)
- Lena Ingeborg Wolff
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University Münster, Munster, Germany
| | - Christine Hartmann
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University Münster, Munster, Germany.
| |
Collapse
|
337
|
He Y, Moqbel SAA, Xu L, Ran J, Ma C, Xu K, Bao J, Jiang L, Chen W, Xiong Y, Wu L. Costunolide inhibits matrix metalloproteinases expression and osteoarthritis via the NF‑κB and Wnt/β‑catenin signaling pathways. Mol Med Rep 2019; 20:312-322. [PMID: 31115524 PMCID: PMC6580033 DOI: 10.3892/mmr.2019.10239] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/17/2019] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease involving cartilage erosion and matrix degradation. Costunolide is a sesquiterpene lactone that has been demonstrated to exert anti-inflammatory activities in a wide variety of cells. The aim of the present study was to investigate the effect of costunolide in OA treatment, using rat chondrocytes and an OA rat model, in which animals were subjected to destabilization of the medial meniscus. The results revealed that costunolide (2–6 µM) had no effect on chondrocyte viability or phenotype maintenance. Costunolide decreased the interleukin (IL)-1β-induced upregulation of matrix metalloproteinases (MMPs), inducible nitric oxide synthase, cyclooxygenase-2 and IL-6, and increased the expression of collagen II and transcription factor SOX-9, which were inhibited by IL-1β. Costunolide significantly decreased p65 phosphorylation induced by IL-1β and the translocation of p65 into the nucleus of rat chondrocytes, as observed by western blot analysis and immunofluorescence staining. In addition, activation of the Wnt/β-catenin signaling pathway was inhibited by costunolide, as demonstrated by the level of activation of β-catenin and the transfer of β-catenin into the nucleus induced by IL-1β. In vivo, cartilage treated with costunolide exhibited attenuated degeneration and lower Mankin scores compared with the OA group. The present study investigated the anti-osteoarthritic effects of costunolide, which exerted anti-inflammatory activities and inhibited MMPs expression. Taken together, these results indicate that costunolide may have a potential value in the treatment of OA.
Collapse
Affiliation(s)
- Yuzhe He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Safwat Adel Abdo Moqbel
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Langhai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Chiyuan Ma
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Kai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Jiapeng Bao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Weiping Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Yan Xiong
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
338
|
Moon YH, Lim W, Jeong BC. Transmembrane protein 64 modulates prostate tumor progression by regulating Wnt3a secretion. Oncol Lett 2019; 18:283-290. [PMID: 31289498 PMCID: PMC6540479 DOI: 10.3892/ol.2019.10324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
Wnt3a is a glycosylated ligand that activates the β-catenin-dependent signaling pathway. Wnt signaling is also important in the prostate tumor microenvironment, and Wnt proteins secreted by the tumor stroma promote resistance to therapy. Bioactive Wnt3a production requires a number of dedicated factors in the secretory cell, but their coordinated functions are not fully understood. We previously reported transmembrane protein 64 (Tmem64) as a novel regulator of the Wnt/β-catenin signaling pathway, which is correlated with β-catenin regulation. In the present study, the role of Tmem64 in prostate cancer cells was investigated by modulating Wnt3a secretion. Overexpression of Tmem64 inhibited Wnt3a secretion and Lef/Tcf-sensitive transcription. By contrast, a Tmem64 mutation deleting the protein's transmembrane region restored Wnt3a secretion. Notably, Tmem64 protein and mRNA in PC3 cells were significantly overexpressed compared with that observed in LNCaP and DU145 cells. In a mouse metastasis model intracardially injected with PC3 cells, Tmem64 expression was downregulated in the metastatic spine and mandible lesions compared with in the primary injection regions. However, Wnt3a was strongly expressed in the metastatic spine and mandible lesions. Collectively, these findings suggest that Tmem64 is involved in the metastatic progression of prostate cancer cells by regulating Wnt3a secretion.
Collapse
Affiliation(s)
- Yeon Hee Moon
- Department of Dental Hygiene, Chodang University, Muangun, Jeollanamdo 58530, Republic of Korea
| | - Wonbong Lim
- Department of Orthopaedic Surgery, Chosun University Hospital, Donggu, Gwangju 61453, Republic of Korea.,Laboratory of Orthopaedic Research, Department of Orthopaedic Surgery, Chosun University Hospital, Donggu, Gwangju 61453, Republic of Korea.,Department of Premedical Program, School of Medicine, Chosun University, Donggu, Gwangju 61452, Republic of Korea
| | - Byung-Chul Jeong
- Department of Orthopaedic Surgery, Chosun University Hospital, Donggu, Gwangju 61453, Republic of Korea.,Laboratory of Orthopaedic Research, Department of Orthopaedic Surgery, Chosun University Hospital, Donggu, Gwangju 61453, Republic of Korea
| |
Collapse
|
339
|
Xu L, Shunmei E, Lin S, Hou Y, Lin W, He W, Wang H, Li G. Sox11-modified mesenchymal stem cells accelerate cartilage defect repair in SD rats. Cell Tissue Res 2019; 376:247-255. [PMID: 30617615 DOI: 10.1007/s00441-018-02979-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 12/13/2018] [Indexed: 10/27/2022]
Abstract
Cartilage has a limited capacity to heal. Previously, we have shown that overexpression of Sox11 in rMSCs (Rat Mesenchymal Stem Cells) by lentivirus-mediated gene transfer leads to enhanced tri-lineage differentiation and accelerated bone formation in fracture model of rats. We observed that the fracture repair in the rats that received Sox11-modified rMSCs injection proceeded through an endochondral ossification process much faster than those in the control groups. However, the detailed role of Sox11 in rMSCs chondrogenic differentiation, as well as cartilage defect, is still not clearly clarified. Therefore, this study tests the hypothesis that Sox11 promotes chondrogenesis and cartilage defect repair by regulating β-catenin. Sox11 was transduced into rMSCs using lentiviruses. The expression levels of β-catenin and its downstream genes were evaluated by quantitative RT-PCR. The transcriptional activation of β-catenin was proved by dual-luciferase reporter assay and co-immunoprecipitation was performed to evaluate Sox11-β-catenin interaction. In addition, a cartilage defect model in SD rats was used to evaluate the cartilage regeneration ability of Sox11-modified rMSCs in vivo. We found that Sox11 transcriptionally activated β-catenin expression and discovered the core promoter region (from - 242 to - 1414) of β-catenin gene for Sox11 binding. In addition, Sox11 might regulate β-catenin at the post-transcriptional level by protein-protein interaction. Finally, using a cartilage defect model in rats, we found Sox11-modified rMSCs could improve cartilage regeneration. Taken together, our study shows that Sox11 is an important regulator of chondrogenesis and Sox11-modified rMSCs may have clinical implication for accelerating cartilage defect healing.
Collapse
Affiliation(s)
- Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Jichang Road, Baiyun District, Guangzhou, People's Republic of China
| | - E Shunmei
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, People's Republic of China
| | - Yonghui Hou
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, People's Republic of China
| | - Weiping Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, People's Republic of China
| | - Wei He
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Jichang Road, Baiyun District, Guangzhou, People's Republic of China
| | - Haibin Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.
- Laboratory of Orthopaedics & Traumatology, Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Jichang Road, Baiyun District, Guangzhou, People's Republic of China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, People's Republic of China.
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China.
| |
Collapse
|
340
|
Usami Y, Gunawardena AT, Francois NB, Otsuru S, Takano H, Hirose K, Matsuoka M, Suzuki A, Huang J, Qin L, Iwamoto M, Yang W, Toyosawa S, Enomoto-Iwamoto M. Possible Contribution of Wnt-Responsive Chondroprogenitors to the Postnatal Murine Growth Plate. J Bone Miner Res 2019; 34:964-974. [PMID: 30602070 PMCID: PMC6536347 DOI: 10.1002/jbmr.3658] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022]
Abstract
Active cell proliferation and turnover in the growth plate is essential for embryonic and postnatal bone growth. We performed a lineage tracing of Wnt/β-catenin signaling responsive cells (Wnt-responsive cells) using Axin2CreERT2 ;Rosa26ZsGreen mice and found a novel cell population that resides in the outermost layer of the growth plate facing the Ranvier's groove (RG; the perichondrium adjacent to growth plate). These Wnt-responsive cells rapidly expanded and contributed to formation of the outer growth plate from the neonatal to the growing stage but stopped expanding at the young adult stage when bone longitudinal growth ceases. In addition, a second Wnt-responsive sporadic cell population was localized within the resting zone of the central part of the growth plate during the postnatal growth phase. While it induced ectopic chondrogenesis in the RG, ablation of β-catenin in the Wnt-responsive cells strongly inhibited expansion of their descendants toward the growth plate. These findings indicate that the Wnt-responsive cell population in the outermost layer of the growth plate is a unique cell source of chondroprogenitors involving lateral growth of the growth plate and suggest that Wnt/β-catenin signaling regulates function of skeletal progenitors in a site- and stage-specific manner. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yu Usami
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aruni T Gunawardena
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Noelle B Francois
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Satoru Otsuru
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Orthopaedics, University of Maryland, Baltimore School of Medicine, Baltimore, MD, USA
| | - Hajime Takano
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Katsutoshi Hirose
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masatake Matsuoka
- Department of Orthopaedics, University of Maryland, Baltimore School of Medicine, Baltimore, MD, USA
| | - Akiko Suzuki
- Department of Orthopaedics, University of Maryland, Baltimore School of Medicine, Baltimore, MD, USA
| | - Jiahui Huang
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI, USA
| | - Ling Qin
- Mckay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Masahiro Iwamoto
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Orthopaedics, University of Maryland, Baltimore School of Medicine, Baltimore, MD, USA
| | - Wentian Yang
- Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI, USA
| | - Satoru Toyosawa
- Department of Oral Pathology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Motomi Enomoto-Iwamoto
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Orthopaedics, University of Maryland, Baltimore School of Medicine, Baltimore, MD, USA
| |
Collapse
|
341
|
Tam WL, Luyten FP, Roberts SJ. From skeletal development to the creation of pluripotent stem cell-derived bone-forming progenitors. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0218. [PMID: 29786553 DOI: 10.1098/rstb.2017.0218] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2017] [Indexed: 02/06/2023] Open
Abstract
Bone has many functions. It is responsible for protecting the underlying soft organs, it allows locomotion, houses the bone marrow and stores minerals such as calcium and phosphate. Upon damage, bone tissue can efficiently repair itself. However, healing is hampered if the defect exceeds a critical size and/or is in compromised conditions. The isolation or generation of bone-forming progenitors has applicability to skeletal repair and may be used in tissue engineering approaches. Traditionally, bone engineering uses osteochondrogenic stem cells, which are combined with scaffold materials and growth factors. Despite promising preclinical data, limited translation towards the clinic has been observed to date. There may be several reasons for this including the lack of robust cell populations with favourable proliferative and differentiation capacities. However, perhaps the most pertinent reason is the failure to produce an implant that can replicate the developmental programme that is observed during skeletal repair. Pluripotent stem cells (PSCs) can potentially offer a solution for bone tissue engineering by providing unlimited cell sources at various stages of differentiation. In this review, we summarize key embryonic signalling pathways in bone formation coupled with PSC differentiation strategies for the derivation of bone-forming progenitors.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Wai Long Tam
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Frank P Luyten
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 bus 813, 3000 Leuven, Belgium
| | - Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, Herestraat 49 Box 813, 3000 Leuven, Belgium .,Bone Therapeutic Area, UCB Pharma, 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| |
Collapse
|
342
|
Sarmah S, Curtis C, Mahin J, Farrell M, Engler TA, Sanchez-Felix MV, Sato M, Ma YL, Chu S, Marrs JA. The Glycogen Synthase Kinase-3β Inhibitor LSN 2105786 Promotes Zebrafish Fin Regeneration. Biomedicines 2019; 7:biomedicines7020030. [PMID: 31010223 PMCID: PMC6630808 DOI: 10.3390/biomedicines7020030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/12/2019] [Accepted: 04/14/2019] [Indexed: 11/16/2022] Open
Abstract
The Wnt pathway has been shown to regulate bone homeostasis and to influence some bone disease states. We utilized a zebrafish model system to study the effects of a synthetic, orally bioavailable glycogen synthase kinase-3β (GSK3β) inhibitor LSN 2105786, which activates Wnt signaling during bone healing and embryogenesis. GSK3β inhibitor treatment was used to phenocopy GSK3β morpholino oligonucleotide (MO) knockdown in zebrafish embryos. Human and zebrafish synthetic mRNA injection were similarly effective at rescue of GSK3β MO knockdown. During caudal fin regeneration, bony rays are the first structure to differentiate in zebrafish fins, providing a useful model to study bone healing. Caudal fin regeneration experiments were conducted using various concentrations of a GSK3β inhibitor, examining duration and concentration dependence on regenerative outgrowth. Experiments revealed continuous low concentration (4-5 nM) treatment to be more effective at increasing regeneration than intermittent dosing. Higher concentrations inhibited fin growth, perhaps by excessive stimulation of differentiation programs. Increased Wnt responsive gene expression and differentiation were observed in response to GSK3b inhibitor treatment. Activating Wnt signaling also increased cell proliferation and osteoblast differentiation in fin regenerates. Together, these data indicate that bone healing in zebrafish fin regeneration was improved by activating Wnt signaling using GSK3b inhibitor treatment. In addition, caudal fin regeneration is useful to evaluate dose-dependent pharmacological efficacy in bone healing, various dosing regimens and possible toxicological effects of compounds.
Collapse
Affiliation(s)
- Swapnalee Sarmah
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Courtney Curtis
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Jennifer Mahin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | - Mark Farrell
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| | | | - Manuel V Sanchez-Felix
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
- Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA.
| | - Masahiko Sato
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
| | | | - Shaoyou Chu
- Lilly Research Laboratories, Indianapolis, IN 46225, USA.
- Molecular Templates, Austin, TX 78729, USA.
| | - James A Marrs
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
343
|
Diederichs S, Tonnier V, März M, Dreher SI, Geisbüsch A, Richter W. Regulation of WNT5A and WNT11 during MSC in vitro chondrogenesis: WNT inhibition lowers BMP and hedgehog activity, and reduces hypertrophy. CELLULAR AND MOLECULAR LIFE SCIENCES : CMLS 2019. [PMID: 30980110 DOI: 10.1007/s00018‐019‐03099‐0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Re-directing mesenchymal stromal cell (MSC) chondrogenesis towards a non-hypertrophic articular chondrocyte-(AC)-like phenotype is important for improving articular cartilage neogenesis to enhance clinical cartilage repair strategies. This study is the first to demonstrate that high levels of non-canonical WNT5A followed by WNT11 and LEF1 discriminated MSC chondrogenesis from AC re-differentiation. Moreover, β-catenin seemed incompletely silenced in differentiating MSCs, which altogether suggested a role for WNT signaling in hypertrophic MSC differentiation. WNT inhibition with the small molecule IWP-2 supported MSC chondrogenesis according to elevated proteoglycan deposition and reduced the characteristic upregulation of BMP4, BMP7 and their target ID1, as well as IHH and its target GLI1 observed during endochondral differentiation. Along with the pro-hypertrophic transcription factor MEF2C, multiple hypertrophic downstream targets including IBSP and alkaline phosphatase activity were reduced by IWP-2, demonstrating that WNT activity drives BMP and hedgehog upregulation, and MSC hypertrophy. WNT inhibition almost matched the strong anti-hypertrophic capacity of pulsed parathyroid hormone-related protein application, and both outperformed suppression of BMP signaling with dorsomorphin, which also reduced cartilage matrix deposition. Yet, hypertrophic marker expression under IWP-2 remained above AC level, and in vivo mineralization and ectopic bone formation were reduced but not eliminated. Overall, the strong anti-hypertrophic effects of IWP-2 involved inhibition but not silencing of pro-hypertrophic BMP and IHH pathways, and more advanced silencing of WNT activity as well as combined application of IHH or BMP antagonists should next be considered to install articular cartilage neogenesis from human MSCs.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Veronika Tonnier
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie März
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Geisbüsch
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
344
|
The ERK MAPK Pathway Is Essential for Skeletal Development and Homeostasis. Int J Mol Sci 2019; 20:ijms20081803. [PMID: 31013682 PMCID: PMC6514701 DOI: 10.3390/ijms20081803] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) are a family of protein kinases that function as key signal transducers of a wide spectrum of extracellular stimuli, including growth factors and pro-inflammatory cytokines. Dysregulation of the extracellular signal-regulated kinase (ERK) MAPK pathway is associated with human skeletal abnormalities including Noonan syndrome, neurofibromatosis type 1, and cardiofaciocutaneous syndrome. Here, we demonstrate that ERK activation in osteoprogenitors is required for bone formation during skeletal development and homeostasis. Deletion of Mek1 and Mek2, kinases upstream of ERK MAPK, in osteoprogenitors (Mek1OsxMek2−/−), resulted in severe osteopenia and cleidocranial dysplasia (CCD), similar to that seen in humans and mice with impaired RUNX2 function. Additionally, tamoxifen-induced deletion of Mek1 and Mek2 in osteoprogenitors in adult mice (Mek1Osx-ERTMek2−/−) significantly reduced bone mass. Mechanistically, this corresponded to decreased activation of osteoblast master regulators, including RUNX2, ATF4, and β-catenin. Finally, we identified potential regulators of osteoblast differentiation in the ERK MAPK pathway using unbiased phospho-mass spectrometry. These observations demonstrate essential roles of ERK activation in osteogenesis and bone formation.
Collapse
|
345
|
Liu YB, Lin LP, Zou R, Zhao QH, Lin FQ. Silencing long non-coding RNA MEG3 accelerates tibia fraction healing by regulating the Wnt/β-catenin signalling pathway. J Cell Mol Med 2019; 23:3855-3866. [PMID: 30955246 PMCID: PMC6533481 DOI: 10.1111/jcmm.14229] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 01/21/2019] [Accepted: 01/25/2019] [Indexed: 02/06/2023] Open
Abstract
As fracture healing is related to gene expression, fracture healing is prospected to be implicated in long non‐coding RNAs (lncRNAs). This study focuses on the effects of epigenetic silencing of long non‐coding RNA maternally expressed gene 3 (lncRNA MEG3) on fracture healing by regulating the Wnt/β‐catenin signalling pathway. Genes expressed in fracture were screened using bioinformatics and the subcellular location of MEG3 was determined using FISH. Next, we successfully established tibia fracture (TF) models of C57BL/6J and Col2a1‐ICAT mice and the effect of silencing lncRNA MEG3 on fracture healing was detected after TF mice were treated with phosphate buffer saline (PBS), MEG3 siRNA and scramble siRNA. X‐ray imaging, Safranin‐O/fast green and haematoxylin‐eosin (HE) staining and histomorphometrical and biomechanical analysis were adopted to observe and to detect the fracture healing conditions. Additionally, the positive expression of collagen II and osteocalcin was examined using immunohistochemistry. At last, in the in vitro experiment, the relationship of MEG3 and the Wnt/β‐catenin signalling pathway in fraction healing was investigated. MEG3 was located in the cell nucleus. In addition, it was found that MEG3 and the Wnt/β‐catenin signalling pathway were associated with fraction healing. Moreover, silencing MEG3 was proved to elevate callus area and maximum bending load and to furthermore enhance the recanalization of bone marrow cavity. Finally, MEG3 knockdown elevated levels of Col10a1, Runx2, Osterix, Osteocalcin, Wnt10b and β‐catenin/β‐catenin whereas it reduced p‐GSK‐3β/GSK‐3β levels. Taken together, our data supported that epigenetic silencing of lncRNA MEG3 could promote the tibia fracture healing by activating the Wnt/β‐catenin signalling pathway.
Collapse
Affiliation(s)
- Yu-Bao Liu
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Lu-Pan Lin
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Rui Zou
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Qing-Hua Zhao
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| | - Fu-Qing Lin
- Department of Orthopaedics, Luhe People's Hospital of Nanjing, Nanjing, P.R. China
| |
Collapse
|
346
|
Komori T. Regulation of Proliferation, Differentiation and Functions of Osteoblasts by Runx2. Int J Mol Sci 2019; 20:ijms20071694. [PMID: 30987410 PMCID: PMC6480215 DOI: 10.3390/ijms20071694] [Citation(s) in RCA: 497] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 11/25/2022] Open
Abstract
Runx2 is essential for osteoblast differentiation and chondrocyte maturation. During osteoblast differentiation, Runx2 is weakly expressed in uncommitted mesenchymal cells, and its expression is upregulated in preosteoblasts, reaches the maximal level in immature osteoblasts, and is down-regulated in mature osteoblasts. Runx2 enhances the proliferation of osteoblast progenitors by directly regulating Fgfr2 and Fgfr3. Runx2 enhances the proliferation of suture mesenchymal cells and induces their commitment into osteoblast lineage cells through the direct regulation of hedgehog (Ihh, Gli1, and Ptch1), Fgf (Fgfr2 and Fgfr3), Wnt (Tcf7, Wnt10b, and Wnt1), and Pthlh (Pthr1) signaling pathway genes, and Dlx5. Runx2 heterozygous mutation causes open fontanelle and sutures because more than half of the Runx2 gene dosage is required for the induction of these genes in suture mesenchymal cells. Runx2 regulates the proliferation of osteoblast progenitors and their differentiation into osteoblasts via reciprocal regulation with hedgehog, Fgf, Wnt, and Pthlh signaling molecules, and transcription factors, including Dlx5 and Sp7. Runx2 induces the expression of major bone matrix protein genes, including Col1a1, Spp1, Ibsp, Bglap2, and Fn1, in vitro. However, the functions of Runx2 in differentiated osteoblasts in the expression of these genes in vivo require further investigation.
Collapse
Affiliation(s)
- Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan.
| |
Collapse
|
347
|
Wang T, Sun CH, Zhong HB, Gong Y, Cui ZK, Xie J, Wang YP, Liang C, Cao HH, Chen XR, Zou ZP, Li SF, Bai XC. N-(3-methoxybenzyl)-(9Z,12Z,15Z)-octadecatrienamide promotes bone formation via the canonical Wnt/β-catenin signaling pathway. Phytother Res 2019; 33:1074-1083. [PMID: 30768733 DOI: 10.1002/ptr.6301] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 12/14/2018] [Accepted: 01/10/2019] [Indexed: 11/07/2022]
Abstract
Osteoporosis is characterized by low bone mineral density and microarchitectural deterioration of bone tissue. N-(3-methoxybenzyl)-(9Z,12Z,15Z)-octadecatrienamide (MBOC) is one of the macamides isolated from Maca (Lepidium meyenii Walp.), a cruciferous plant from the Andes of Peru. In this study, C3H/10T1/2 mesenchymal stem cells were treated with MBOC in osteogenic induction medium. An ovariectomized (OVX) mouse model was used to investigate the effect of 1-month MBOC treatment on the prevention of postmenopausal osteoporosis. Remarkably, trabecular thickness, trabecular number, and bone volume/tissue volume of the distal femoral metaphysis were significantly increased in OVX + MBOC mice compared with OVX mice, as revealed by microcomputed tomography analysis. Trabecular separation was decreased in OVX + MBOC mice compared with OVX mice. Consistently, MBOC increased the levels of osteocalcin and runt-related transcription factor 2 in OVX mice, as well as the expression of runt-related transcription factor 2, osterix, and alkaline phosphatase in C3H/10T1/2 cells. Mechanistically, MBOC activates the canonical Wnt/β-catenin signaling pathway via inhibiting phosphorylation of GSK-3β at Tyr216 and maintaining β-catenin expression. Collectively, the current study demonstrates the robustness of MBOC in the induction of mesenchymal stem cells osteogenic differentiation and consequent bone formation, suggesting that MBOC may be a potentially effective drug to treat postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Ting Wang
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Chun-Han Sun
- Department of Orthopedics, Huizhou First People's Hospital, Huizhou, Guangdong, China
| | - Hao-Bo Zhong
- Department of Orthopedics, Huizhou First People's Hospital, Huizhou, Guangdong, China
| | - Yan Gong
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhong-Kai Cui
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Xie
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Peng Wang
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuang Liang
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - He-He Cao
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao-Rui Chen
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhi-Peng Zou
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Sheng-Fa Li
- Department of Orthopedics, Huizhou First People's Hospital, Huizhou, Guangdong, China
| | - Xiao-Chun Bai
- Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
348
|
Chen H, Ji X, Lee WC, Shi Y, Li B, Abel ED, Jiang D, Huang W, Long F. Increased glycolysis mediates Wnt7b-induced bone formation. FASEB J 2019; 33:7810-7821. [PMID: 30913395 DOI: 10.1096/fj.201900201rr] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Wingless/integrated (Wnt) signaling has emerged as a major mechanism for promoting bone formation and a target pathway for developing bone anabolic agents against osteoporosis. However, the downstream events mediating the potential therapeutic effect of Wnt proteins are not fully understood. Previous studies have indicated that increased glycolysis is associated with osteoblast differentiation in response to Wnt signaling, but direct genetic evidence for the importance of glucose metabolism in Wnt-induced bone formation is lacking. Here, we have generated compound transgenic mice to overexpress Wnt family member 7B (Wnt7b) transiently in the osteoblast lineage of postnatal mice, with or without concurrent deletion of the glucose transporter 1 (Glut1), also known as solute carrier family 2, facilitated glucose transporter member 1. Overexpression of Wnt7b in 1-mo-old mice for 1 wk markedly stimulated bone formation, but the effect was essentially abolished without Glut1, even though transient deletion of Glut1 itself did not affect normal bone accrual. Consistent with the in vivo results, Wnt7b increased Glut1 expression and glucose consumption in the primary culture of osteoblast lineage cells, and deletion of Glut1 diminished osteoblast differentiation in vitro. Thus, Wnt7b promotes bone formation in part through stimulating glucose metabolism in osteoblast lineage cells.-Chen, H., Ji, X., Lee, W.-C., Shi, Y., Li, B., Abel, E. D., Jiang, D., Huang, W., Long, F. Increased glycolysis mediates Wnt7b-induced bone formation.
Collapse
Affiliation(s)
- Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Xing Ji
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Wen-Chih Lee
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Yu Shi
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Boer Li
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| | - E Dale Abel
- Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Dianming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fanxin Long
- Department of Orthopedic Surgery, School of Medicine, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
349
|
Kim SP, Da H, Li Z, Kushwaha P, Beil C, Mei L, Xiong WC, Wolfgang MJ, Clemens TL, Riddle RC. Lrp4 expression by adipocytes and osteoblasts differentially impacts sclerostin's endocrine effects on body composition and glucose metabolism. J Biol Chem 2019; 294:6899-6911. [PMID: 30842262 DOI: 10.1074/jbc.ra118.006769] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/23/2019] [Indexed: 01/10/2023] Open
Abstract
Sclerostin exerts profound local control over bone acquisition and also mediates endocrine communication between fat and bone. In bone, sclerostin's anti-osteoanabolic activity is enhanced by low-density lipoprotein receptor-related protein 4 (Lrp4), which facilitates its interaction with the Lrp5 and Lrp6 Wnt co-receptors. To determine whether Lrp4 similarly affects sclerostin's endocrine function, we examined body composition as well as glucose and fatty acid metabolism in mice rendered deficient of Lrp4 in the adipocyte (AdΔLrp4) or the osteoblast (ObΔLrp4). AdΔLrp4 mice exhibit a reduction in adipocyte hypertrophy and improved glucose and lipid homeostasis, marked by increased glucose and insulin tolerance and reduced serum fatty acids, and mirror the effect of sclerostin deficiency on whole-body metabolism. Indeed, epistasis studies place adipocyte-expressed Lrp4 and sclerostin in the same genetic cascade that regulates adipocyte function. Intriguingly, ObΔLrp4 mice, which exhibit dramatic increases in serum sclerostin, accumulate body fat and develop impairments in glucose tolerance and insulin sensitivity despite development of a high bone mass phenotype. These data indicate that expression of Lrp4 by both the adipocyte and osteoblast is required for normal sclerostin endocrine function and that the impact of sclerostin deficiency on adipocyte physiology is distinct from the effect on osteoblast function.
Collapse
Affiliation(s)
| | - Hao Da
- From the Departments of Orthopedic Surgery and
| | - Zhu Li
- From the Departments of Orthopedic Surgery and
| | | | - Conor Beil
- From the Departments of Orthopedic Surgery and
| | - Lin Mei
- the Department of Neuroscience, Case Western Reserve University Medical School, Cleveland, Ohio 44106, and
| | - Wen-Cheng Xiong
- the Department of Neuroscience, Case Western Reserve University Medical School, Cleveland, Ohio 44106, and
| | - Michael J Wolfgang
- Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Thomas L Clemens
- From the Departments of Orthopedic Surgery and.,the Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201
| | - Ryan C Riddle
- From the Departments of Orthopedic Surgery and .,the Baltimore Veterans Affairs Medical Center, Baltimore, Maryland 21201
| |
Collapse
|
350
|
Jaber FA, Khan NM, Ansari MY, Al-Adlaan AA, Hussein NJ, Safadi FF. Autophagy plays an essential role in bone homeostasis. J Cell Physiol 2019; 234:12105-12115. [PMID: 30820954 DOI: 10.1002/jcp.27071] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/29/2018] [Indexed: 12/20/2022]
Abstract
Autophagy is very critical for multiple cellular processes. Autophagy plays a critical role in bone cell differentiation and function.
Collapse
Affiliation(s)
- Fatima A Jaber
- Department of Biology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazir M Khan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Mohammad Y Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Asaad A Al-Adlaan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazar J Hussein
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Fayez F Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio.,Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio.,Department of Orthopedic Surgery, SUMMA Health System, Akron, Ohio.,Rebecca D. Considine Research Institute Akron Children's Hospital, Akron, Ohio
| |
Collapse
|