1
|
Bhattarai G, An JH, Rijal S, Lee J, Kim J, Kook SH, Lee JC, Cho ES. Supplemental magnesium gluconate recovers osteoblastic Wntless ablation-induced degenerative bone complications. J Bone Miner Metab 2025:10.1007/s00774-025-01599-7. [PMID: 40186045 DOI: 10.1007/s00774-025-01599-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION Although numerous studies have highlighted the involvement of Wnt-mediated signaling in Mg ion-enhanced bone healing, whether Wnt-stimulated signaling is essential for the Mg ion-triggered bone repair and mass accrual is not yet completely understood. MATERIALS AND METHODS We generated Wlsfl/fl wild-type (WT) control and their corresponding mutant (MT), Col2.3-Cre;Wlsfl/fl mice of osteoblastic Wntless (Wls) ablation and explored how supplemental magnesium gluconate (MgG) affects bone mass accrual and defected bone healing in relation to the Wls ablation. RESULTS Osteoblastic Wls ablation impaired bone mass accrual and bone healing along with age-related degenerative complications in bone marrow (BM) and BM cells. Oral supplementation of WT mice with MgG did not change natural bone mass accrual, but enhanced regenerative bone healing in femoral defects and the functionalities of BM cells. Supplemental MgG suppressed the Wls ablation-related bone loss and also stimulated new bone formation in the defects of MT mice. The MgG-induced beneficial effects in the MT mice were orchestrated with its potencies to ameliorate senescence, oxidative damage, and functional loss of BM and BM adherent cells, as well as to stimulate osteogenic activity. CONCLUSION This study demonstrates that supplemental MgG is able to improve bone homeostatic maintenance by recovering age-related degenerative complications even at the lack of osteoblastic Wnt-stimulated signaling.
Collapse
Affiliation(s)
- Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Ju-Hyeon An
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Shankar Rijal
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Junil Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Junhyeok Kim
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Sung-Ho Kook
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, South Korea
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea.
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, 54896, South Korea.
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, 54896, South Korea.
| |
Collapse
|
2
|
Xi Y, Jiang Q, Dai W, Chen C, Wang Y, Miao X, Lai K, Jiang Z, Yang G, Wang Y. SP7 transcription factor ameliorates bone defect healing in low-density lipoprotein receptor-related protein 5 (LRP5)-dependent osteoporosis mice. J Zhejiang Univ Sci B 2025; 26:254-268. [PMID: 40082204 PMCID: PMC11906391 DOI: 10.1631/jzus.b2300531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/30/2023] [Indexed: 03/16/2025]
Abstract
Loss-of-function variants of low-density lipoprotein receptor-related protein 5 (LRP5) can lead to reduced bone formation, culminating in diminished bone mass. Our previous study reported transcription factor osterix (SP7)-binding sites on the LRP5 promoter and its pivotal role in upregulating LRP5 expression during implant osseointegration. However, the potential role of SP7 in ameliorating LRP5-dependent osteoporosis remained unknown. In this study, we used mice with a conditional knockout (cKO) of LRP5 in mature osteoblasts, which presented decreased osteogenesis. The in vitro experimental results showed that SP7 could promote LRP5 expression, thereby upregulating the osteogenic markers such as alkaline phosphatase (ALP), Runt-related transcription factor 2 (Runx2), and β-catenin (P<0.05). For the in vivo experiment, the SP7 overexpression virus was injected into a bone defect model of LRP5 cKO mice, resulting in increased bone mineral density (BMD) (P<0.001) and volumetric density (bone volume (BV)/total volume (TV)) (P<0.001), and decreased trabecular separation (Tb.Sp) (P<0.05). These data suggested that SP7 could ameliorate bone defect healing in LRP5 cKO mice. Our study provides new insights into potential therapeutic opportunities for ameliorating LRP5-dependent osteoporosis.
Collapse
Affiliation(s)
- Yue Xi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Qifeng Jiang
- School of Stomatology, Zhejiang University, Hangzhou 310058, China
| | - Wei Dai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Chaozhen Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Yang Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Xiaoyan Miao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Kaichen Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China. ,
| |
Collapse
|
3
|
Abdullah Sani N, Kamaruddin NA, Soelaiman IN, Pang KL, Chin KY, Ramli ESM. Palm Tocotrienol Activates the Wnt3a/β-Catenin Signaling Pathway, Protecting MC3T3-E1 Osteoblasts from Cellular Damage Caused by Dexamethasone and Promoting Bone Formation. Biomedicines 2025; 13:243. [PMID: 39857826 PMCID: PMC11762645 DOI: 10.3390/biomedicines13010243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Background and aim: Prolonged glucocorticoid (GC) treatment increases oxidative stress, triggers apoptosis of osteoblasts, and contributes to osteoporosis. Tocotrienol, as an antioxidant, could protect the osteoblasts and preserve bone quality under glucocorticoid treatment. From this study, we aimed to determine the effects of tocotrienol on MC3T3-E1 murine pre-osteoblastic cells treated with GC. Methods: MC3T3-E1 cells were exposed to dexamethasone (150 µM), with or without palm tocotrienol (PTT; 0.25, 0.5, and 1 µg/mL). Cell viability was measured by the MTS assay. Alizarin Red staining was performed to detect calcium deposits. Cellular alkaline phosphatase activity was measured to evaluate osteogenic activity. The expression of osteoblastic differentiation markers was measured by an enzyme-linked immunoassay. Results: Enhanced matrix mineralization was observed in the cells treated with 0.5 µg/mL PTT, especially on day 18 (p < 0.05). The expression of Wnt3a, β-catenin, collagen 1α1, alkaline phosphatase, osteocalcin, low-density lipoprotein receptor-related protein 6, and runt-related transcription factor-2 were significantly increased in the PTT-treated groups compared to the vehicle control group, especially at 0.5 µg/mL of PTT (p < 0.05) and on day 6 of treatment. Conclusions: PTT maintains the osteogenic activity of the dexamethasone-treated osteoblasts by promoting their differentiation.
Collapse
Affiliation(s)
- Norfarahin Abdullah Sani
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (N.A.S.); (N.A.K.)
| | - Nur Aqilah Kamaruddin
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (N.A.S.); (N.A.K.)
| | - Ima Nirwana Soelaiman
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.N.S.); (K.-L.P.); (K.-Y.C.)
| | - Kok-Lun Pang
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.N.S.); (K.-L.P.); (K.-Y.C.)
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Subang Jaya 46150, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.N.S.); (K.-L.P.); (K.-Y.C.)
| | - Elvy Suhana Mohd Ramli
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (N.A.S.); (N.A.K.)
| |
Collapse
|
4
|
Zhang X, Gao X, Xu J, Zhang Z, Lin T, Zhang X, Kang X. The role of lncRNA and miRNA on the effects of occurrence and development of osteosarcoma. Int Immunopharmacol 2025; 144:113726. [PMID: 39615111 DOI: 10.1016/j.intimp.2024.113726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/15/2024]
Abstract
Osteosarcoma is a common primary malignant bone tumor with a high incidence in children and adolescents, with high invasiveness and lung metastases. Even after traditional surgical excision, chemoradiotherapy, and comprehensive treatment, the survival rate of patients is still low, and the prognosis is not ideal. As an important part of non-coding RNA family, lncRNA and miRNA have significant regulatory effects on the growth, proliferation, metastasis and apoptosis of osteosarcoma cells. Therefore, exploring the roles of lncRNAs and miRNAs in the occurrence and development of osteosarcoma is of great help for the subsequent diagnosis, treatment, and prognosis of osteosarcoma. This paper mainly reviews the current research progress on the effects and mechanisms of lncRNAs and miRNAs on osteosarcoma cells, in order to provide new ideas for future research on the development process, treatment methods, and prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Zhuoya Zhang
- The First Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Tingtong Lin
- The Second Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Xueyan Zhang
- Institute of Biochemistry and Molecular Biology and School of Basic Medical Sciences, Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Xin Kang
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China.
| |
Collapse
|
5
|
Kobayashi Y, Iwamoto R, He Z, Udagawa N. Wnt family members regulating osteogenesis and their origins. J Bone Miner Metab 2025; 43:39-45. [PMID: 39283365 PMCID: PMC11954845 DOI: 10.1007/s00774-024-01554-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/27/2024] [Indexed: 04/01/2025]
Abstract
Wnt signaling plays an important role in the regulation of bone metabolism. Wnt activates the β-catenin-mediated canonical pathway and β-catenin-independent non-canonical pathway. When Wnt ligands bind to the co-receptors low density lipoprotein receptor-related protein (Lrp)5 or Lrp6, and a seven-transmembrane receptor frizzled, the canonical pathway is activated. On the other hand, when Wnt ligands bind to the receptor complex consisting of the co-receptor receptor tyrosine kinase-like orphan receptor (Ror)1 and Ror2 or Ryk and frizzled, the non-canonical pathway is activated. An analysis of loss-of-function and gain-of-function mutations in molecules involved in Wnt signaling (ligands, receptors, and inhibitors) has revealed the mechanisms by which Wnt signaling regulates bone metabolism. In this review, based on transcriptome analyses of Wnt expression in bone tissues including single cell RNA sequence analysis and previous literatures, we herein introduce and discussed the latest findings on the mechanisms by which Wnt ligand mutations impair bone metabolism, especially bone formation.
Collapse
Affiliation(s)
- Yasuhiro Kobayashi
- Department of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Hirooka Gohara, Shiojiri, Nahano, 399-0781, Japan.
| | - Rina Iwamoto
- Department of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Hirooka Gohara, Shiojiri, Nahano, 399-0781, Japan
| | - Zhifeng He
- Department of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Hirooka Gohara, Shiojiri, Nahano, 399-0781, Japan
| | - Nobuyuki Udagawa
- Department of Hard Tissue Research, Institute for Oral Science, Matsumoto Dental University, 1780 Hirooka Gohara, Shiojiri, Nahano, 399-0781, Japan
- Department of Biochemistry, Matsumoto Dental University, 1780 Hirooka Gohara, Shiojiri, Nagano, 399-0781, Japan
| |
Collapse
|
6
|
Bhattarai G, Shrestha SK, Rijal S, Kook SH, Lee JC. Supplemental Magnesium Gluconate Enhances Scaffold-Mediated New Bone Formation and Natural Bone Healing by Angiogenic- and Wnt Signal-Associated Osteogenic Activation. J Biomed Mater Res A 2025; 113:e37812. [PMID: 39462850 DOI: 10.1002/jbm.a.37812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Local implantation or supplementation of magnesium gluconate (MgG) is being investigated as an effective approach to bone repair. Although studies have highlighted the possible mechanisms in Mg ion-stimulated new bone formation, the role of MgG in healing bone defects and the signaling mechanisms are not yet completely understood. In this study, we explored how supplemental MgG has bone-specific beneficial effects by delivering MgG locally and orally in animal models. We fabricated MgG-incorporated (CMC-M) and -free chitosan (CMC) scaffolds with good microstructures and biocompatible properties. Implantation with CMC-M enhanced bone healing in rat model of mandible defects, compared with CMC, by activating Wnt signals and Wnt-related osteogenic and angiogenic molecules. Oral supplementation with MgG also stimulated bone healing in mouse model of femoral defects along with the increases in Wnt3a and angiogenic and osteogenic factors. Supplemental MgG did not alter nature bone accrual and bone marrow (BM) microenvironment in adult mouse model, but enhanced the functioning of BM stromal cells (BMSCs). Furthermore, MgG directly stimulated the induction of Wnt signaling-, angiogenesis-, and osteogenesis-related molecules in cultures of BMSCs, as well as triggered the migration of endothelial cells. These results suggest that supplemental MgG improves bone repair in a way that is synergistically enhanced by Wnt signal-associated angiogenic and osteogenic molecules. Overall, this study indicates that supplemental MgG might ameliorate oxidative damage in the BM, improve the functionality of BM stem cells, and maintain BM-microenvironmental homeostasis.
Collapse
Affiliation(s)
- Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Saroj Kumar Shrestha
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Shankar Rijal
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
7
|
Davidson RK, Corry K, Orlofsky A, Li P, Russell CE, Zhang A, Moraes de Lima Perini M, Priddy CN, Nguyen AV, Li J. Loss of STAT3 in osteoblasts has detrimental and sexually dimorphic effects on skeletal development. PLoS One 2024; 19:e0315078. [PMID: 39689092 PMCID: PMC11651548 DOI: 10.1371/journal.pone.0315078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024] Open
Abstract
Studies with genetically modified mice have implicated the transcriptional regulator STAT3 as a key modulator of bone development. STAT3-OKO knockout mouse lines were generated in two genetic backgrounds, pure C57BL/6 (STAT3-OKO-BL) and mixed C57BL/6, CD1 (STAT3-OKO-M). Both lines exhibited defective postnatal bone development resulting in reduced body weight and shortened femurs that displayed low bone mineral density as well as cortical widening and thinning in the diaphyseal region. Remarkably, each of these defects displayed sexual dimorphism that was dependent on genetic background: the phenotype was entirely male-specific in STAT3-OKO-M but not in STAT3-OKO-BL, in which defects were similar in both sexes. However, both lines exhibited a male-specific bone defect in mineralization, and also in bone mechanical properties related to bone quality, such as yield stress and ultimate stress. On the other hand, bone mechanical properties such as ultimate force, that may reflect density and macrostructure rather than bone quality, showed male-specific defects only in STAT3-OKO-M. These findings suggest that STAT3 may regulate multiple sex-dependent mechanisms in bone development that control either mineralization or bone accrual, and that the sex-dependence of at least some of these mechanisms is affected by genetic background. Finally, we used CRISPR/Cas9 to generate STAT3-deficient preosteoblastic cells from immortalized wild-type bone marrow stem cells and showed that the defective osteoblastic differentiation of STAT3-ablated cells was associated with reduced gene expression of Wnt3a and Wnt5a, consistent with other studies that identify Wnt signaling pathways as potential effector mechanisms for STAT3-mediated regulation of bone development.
Collapse
Affiliation(s)
- Rebecca K. Davidson
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| | - Kylie Corry
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| | - Amos Orlofsky
- Department of Biological Sciences and Geology, the City University of New York-Queensborough Community College, Bayside, New York, United States of America
| | - Ping Li
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Caleb E. Russell
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| | - Amy Zhang
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| | | | - Carlie N. Priddy
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| | - Andrew V. Nguyen
- Department of Biological Sciences and Geology, the City University of New York-Queensborough Community College, Bayside, New York, United States of America
| | - Jiliang Li
- Department of Biology, Indiana University Indianapolis, Indianapolis, Indiana, United States of America
| |
Collapse
|
8
|
Azarkina K, Gromova E, Malashicheva A. "A Friend Among Strangers" or the Ambiguous Roles of Runx2. Biomolecules 2024; 14:1392. [PMID: 39595568 PMCID: PMC11591759 DOI: 10.3390/biom14111392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The transcription factor Runx2 plays a crucial role in regulating osteogenic differentiation and skeletal development. This factor not only controls the expression of genes involved in bone formation, but also interacts with signaling pathways such as the Notch pathway, which are essential for body development. However, studies have produced conflicting results regarding the relationship between Runx2 and the Notch pathway. Some studies suggest a synergistic interaction between these molecules, while others suggest an inhibitory one, for example, the interplay between Notch signaling, Runx2, and vitamin D3 in osteogenic differentiation and bone remodeling. The findings suggest a complex relationship between Notch signaling and osteogenic differentiation, with ongoing research needed to clarify the mechanisms involved and resolve existing contradictions regarding role of Notch in this process. Additionally, there is increasing evidence of contradictory roles for Runx2 in various tissues and organs, both under normal conditions and in pathological states. This diversity of roles makes Runx2 a potential therapeutic target, offering new directions for research. In this review, we have discussed the mechanisms of osteogenic differentiation and the important role of Runx2 in this process. We have also examined its relationship with different signaling pathways. However, there are still many uncertainties and inconsistencies in our current understanding of these interactions. Additionally, given that Runx2 is also involved in numerous other events in various tissues, we have tried to comprehensively examine its functions outside the skeletal system.
Collapse
Affiliation(s)
| | | | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, 194064 Saint-Petersburg, Russia
| |
Collapse
|
9
|
Meng F, Zhu P, Ren X, Wang L, Ding D, Yan J, Zhang Y, Yang SY, Ning B. Cardamonin inhibits osteogenic differentiation by downregulating Wnt/beta-catenin signaling and alleviates subchondral osteosclerosis in osteoarthritic mice. J Orthop Res 2024; 42:1933-1942. [PMID: 38520666 DOI: 10.1002/jor.25842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/27/2024] [Accepted: 03/09/2024] [Indexed: 03/25/2024]
Abstract
Osteoarthritis (OA) is a common degenerative joint disease, and subchondral osteosclerosis is an important pathological change that occurs in its late stages. Cardamonin (CD) is a natural flavonoid isolated from Alpinia katsumadai that has anti-inflammatory activity. The objectives of this study were to investigate the therapeutic effects and potential mechanism of CD in regulating OA subchondral osteosclerosis at in vivo and in vitro settings. Eight-week-old male C57BL/6J mice were randomly divided into four groups: sham operation, anterior cruciate ligament transection (ACLT)-induced OA model, low-dose and high-dose CD treated ACLT-OA model groups. Histological assessment and immunohistochemical examinations for chondrocyte metabolism-related markers metalloproteinase-13, ADAMTS-4, Col II, and Sox-9 were performed. Microcomputed tomography was used to assess the sclerosis indicators in subchondral bone. Further, MC3T3-E1 (a mouse calvarial preosteoblast cell line) cells were treated with various concentrations of CD to reveal the influence and potential molecular pathways of CD in osteogenic differentiations. Animal studies suggested that CD alleviated the pathological changes in OA mice such as maintaining integrity and increasing the thickness of hyaline cartilage, decreasing the thickness of calcified cartilage, decreasing the Osteoarthritis Research Society International score, regulating articular cartilage metabolism, and inhibiting subchondral osteosclerosis. In vitro investigation indicated that CD inhibited alkaline phosphatase expression and production of calcium nodules during osteogenic differentiation of MC3T3-E1 cells. In addition, CD inhibited the expression of osteogenic differentiation-related indicators and Wnt/β-catenin pathway-related proteins. In conclusion, CD inhibits osteogenic differentiation by downregulating Wnt/β-catenin signaling and alleviating subchondral osteosclerosis in a mouse model of OA.
Collapse
Affiliation(s)
- Fanding Meng
- Department of Hand and Foot Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Pengchong Zhu
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoli Ren
- Department of Orthopedic Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Limei Wang
- Faculty of Preclinical Medicine, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Dong Ding
- Department of Orthopedic Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
| | - Jiangbo Yan
- The 3rd Orthopedic Ward, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ying Zhang
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shang-You Yang
- Department of Orthopaedic Surgery, University of Kansas School of Medicine Wichita, Wichita, Kansas, USA
| | - Bin Ning
- Department of Orthopedic Surgery, Jinan Central Hospital, Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
10
|
Ozgurel SU, Reyes Fernandez PC, Chanpaisaeng K, Fleet JC. Male Lrp5A214V mice maintain high bone mass during dietary calcium restriction by altering the vitamin D endocrine system. J Bone Miner Res 2024; 39:315-325. [PMID: 38477773 PMCID: PMC11240165 DOI: 10.1093/jbmr/zjae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/20/2023] [Accepted: 01/10/2024] [Indexed: 03/14/2024]
Abstract
Environmental factors and genetic variation individually impact bone. However, it is not clear how these factors interact to influence peak bone mass accrual. Here we tested whether genetically programmed high bone formation driven by missense mutations in the Lrp5 gene (Lrp5A214V) altered the sensitivity of mice to an environment of inadequate dietary calcium (Ca) intake. Weanling male Lrp5A214V mice and wildtype littermates (control) were fed AIN-93G diets with 0.125%, 0.25%, 0.5% (reference, basal), or 1% Ca from weaning until 12 weeks of age (ie, during bone growth). Urinary Ca, serum Ca, Ca regulatory hormones (PTH, 1,25 dihydroxyvitamin D3 (1,25(OH)2D3)), bone parameters (μCT, ash), and renal/intestinal gene expression were analyzed. As expected, low dietary Ca intake negatively impacted bones and Lrp5A214V mice had higher bone mass and ash content. Although bones of Lrp5A214V mice have more matrix to mineralize, their bones were not more susceptible to low dietary Ca intake. In control mice, low dietary Ca intake exerted expected effects on serum Ca (decreased), PTH (increased), and 1,25(OH)2D3 (increased) as well as their downstream actions (ie, reducing urinary Ca, increasing markers of intestinal Ca absorption). In contrast, Lrp5A214V mice had elevated serum Ca with a normal PTH response but a blunted 1,25(OH)2D3 response to low dietary Ca that was reflected in the renal 1,25(OH)2D3 producing/degrading enzymes, Cyp27b1 and Cyp24a1. Despite elevated serum Ca in Lrp5A214V mice, urinary Ca was not elevated. Despite an abnormal serum 1,25(OH)2D3 response to low dietary Ca, intestinal markers of Ca absorption (Trpv6, S100g mRNA) were elevated in Lrp5A214V mice and responded to low Ca intake. Collectively, our data indicate that the Lrp5A214V mutation induces changes in Ca homeostasis that permit mice to retain more Ca and support their high bone mass phenotype.
Collapse
Affiliation(s)
- Serra Ucer Ozgurel
- Department of Nutritional Sciences, University of Texas, Austin, TX 78723, United States
| | - Perla C Reyes Fernandez
- Department of Physical Therapy, Indiana University –Purdue University, Indianapolis, IN 46202, United States
| | - Krittikan Chanpaisaeng
- National Center for Genetic Engineering and Biotechnology, Pathum Thani 12120, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - James C Fleet
- Department of Nutritional Sciences, University of Texas, Austin, TX 78723, United States
| |
Collapse
|
11
|
Xu C, Wang Y, Ni H, Yao M, Cheng L, Lin X. The role of orphan G protein-coupled receptors in pain. Heliyon 2024; 10:e28818. [PMID: 38590871 PMCID: PMC11000026 DOI: 10.1016/j.heliyon.2024.e28818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024] Open
Abstract
G protein-coupled receptors (GPCRs), which form the largest family of membrane protein receptors in humans, are highly complex signaling systems with intricate structures and dynamic conformations and locations. Among these receptors, a specific subset is referred to as orphan GPCRs (oGPCRs) and has garnered significant interest in pain research due to their role in both central and peripheral nervous system function. The diversity of GPCR functions is attributed to multiple factors, including allosteric modulators, signaling bias, oligomerization, constitutive signaling, and compartmentalized signaling. This review primarily focuses on the recent advances in oGPCR research on pain mechanisms, discussing the role of specific oGPCRs including GPR34, GPR37, GPR65, GPR83, GPR84, GPR85, GPR132, GPR151, GPR160, GPR171, GPR177, and GPR183. The orphan receptors among these receptors associated with central nervous system diseases are also briefly described. Understanding the functions of these oGPCRs can contribute not only to a deeper understanding of pain mechanisms but also offer a reference for discovering new targets for pain treatment.
Collapse
Affiliation(s)
- Chengfei Xu
- Department of Anesthesiology, The Third People's Hospital of Bengbu, Bengbu, 233000, PR China
| | - Yahui Wang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, PR China
| | - Huadong Ni
- Department of Anesthesiology and Pain Research Center, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, PR China
| | - Ming Yao
- Department of Anesthesiology and Pain Research Center, Affiliated Hospital of Jiaxing University, Jiaxing, 314000, PR China
| | - Liang Cheng
- Department of Anesthesiology, The Third People's Hospital of Bengbu, Bengbu, 233000, PR China
| | - Xuewu Lin
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, PR China
| |
Collapse
|
12
|
Choppa VSR, Liu G, Tompkins YH, Kim WK. Altered Osteogenic Differentiation in Mesenchymal Stem Cells Isolated from Compact Bone of Chicken Treated with Varying Doses of Lipopolysaccharides. Biomolecules 2023; 13:1626. [PMID: 38002308 PMCID: PMC10669906 DOI: 10.3390/biom13111626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Persistent inflammation biologically alters signaling molecules and ultimately affects osteogenic differentiation, including in modern-day broilers with unique physiology. Lipopolysaccharides (LPS) are Gram-negative bacterial components that activate cells via transmembrane receptor activation and other molecules. Previous studies have shown several pathways associated with osteogenic inductive ability, but the pathway has yet to be deciphered, and data related to its dose-dependent effect are limited. Primary mesenchymal stem cells (MSCs) were isolated from the bones of day-old broiler chickens, and the current study focused on the dose-dependent variation (3.125 micrograms/mL to 50 micrograms/mL) in osteogenic differentiation and the associated biomarkers in primary MSCs. The doses in this study were determined using a cell viability (MTT) assay. The study revealed that osteogenic differentiation varied with dose, and the cells exposed to higher doses of LPS were viable but lacked differentiating ability. However, this effect became transient with lower doses, and this phenotypic character was observed with differential staining methods like Alizarin Red, Von Kossa, and alkaline phosphatase. The data from this study revealed that LPS at varying doses had a varying effect on osteogenic differentiation via several pathways acting simultaneously during bone development.
Collapse
Affiliation(s)
| | | | | | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30605, USA; (V.S.R.C.); (G.L.); (Y.H.T.)
| |
Collapse
|
13
|
Schneider RF, Gunter HM, Salewski I, Woltering JM, Meyer A. Growth dynamics and molecular bases of evolutionary novel jaw extensions in halfbeaks and needlefishes (Beloniformes). Mol Ecol 2023; 32:5798-5811. [PMID: 37750351 DOI: 10.1111/mec.17143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023]
Abstract
Evolutionary novelties-derived traits without clear homology found in the ancestors of a lineage-may promote ecological specialization and facilitate adaptive radiations. Examples for such novelties include the wings of bats, pharyngeal jaws of cichlids and flowers of angiosperms. Belonoid fishes (flying fishes, halfbeaks and needlefishes) feature an astonishing diversity of extremely elongated jaw phenotypes with undetermined evolutionary origins. We investigate the development of elongated jaws in a halfbeak (Dermogenys pusilla) and a needlefish (Xenentodon cancila) using morphometrics, transcriptomics and in situ hybridization. We confirm that these fishes' elongated jaws are composed of distinct base and novel 'extension' portions. These extensions are morphologically unique to belonoids, and we describe the growth dynamics of both bases and extensions throughout early development in both studied species. From transcriptomic profiling, we deduce that jaw extension outgrowth is guided by populations of multipotent cells originating from the anterior tip of the dentary. These cells are shielded from differentiation, but proliferate and migrate anteriorly during the extension's allometric growth phase. Cells left behind at the tip leave the shielded zone and undergo differentiation into osteoblast-like cells, which deposit extracellular matrix with both bone and cartilage characteristics that mineralizes and thereby provides rigidity. Such bone has characteristics akin to histological observations on the elongated 'kype' process on lower jaws of male salmon, which may hint at common conserved regulatory underpinnings. Future studies will evaluate the molecular pathways that govern the anterior migration and proliferation of these multipotent cells underlying the belonoids' evolutionary novel jaw extensions.
Collapse
Affiliation(s)
- Ralf F Schneider
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Marine Ecology, GEOMAR, Kiel, Germany
| | - Helen M Gunter
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Inken Salewski
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Joost M Woltering
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Axel Meyer
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
14
|
Diegel CR, Kramer I, Moes C, Foxa GE, McDonald MJ, Madaj ZB, Guth S, Liu J, Harris JL, Kneissel M, Williams BO. Inhibiting WNT secretion reduces high bone mass caused by Sost loss-of-function or gain-of-function mutations in Lrp5. Bone Res 2023; 11:47. [PMID: 37612291 PMCID: PMC10447437 DOI: 10.1038/s41413-023-00278-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 05/09/2023] [Accepted: 07/03/2023] [Indexed: 08/25/2023] Open
Abstract
Proper regulation of Wnt signaling is critical for normal bone development and homeostasis. Mutations in several Wnt signaling components, which increase the activity of the pathway in the skeleton, cause high bone mass in human subjects and mouse models. Increased bone mass is often accompanied by severe headaches from increased intracranial pressure, which can lead to fatality and loss of vision or hearing due to the entrapment of cranial nerves. In addition, progressive forehead bossing and mandibular overgrowth occur in almost all subjects. Treatments that would provide symptomatic relief in these subjects are limited. Porcupine-mediated palmitoylation is necessary for Wnt secretion and binding to the frizzled receptor. Chemical inhibition of porcupine is a highly selective method of Wnt signaling inhibition. We treated three different mouse models of high bone mass caused by aberrant Wnt signaling, including homozygosity for loss-of-function in Sost, which models sclerosteosis, and two strains of mice carrying different point mutations in Lrp5 (equivalent to human G171V and A214V), at 3 months of age with porcupine inhibitors for 5-6 weeks. Treatment significantly reduced both trabecular and cortical bone mass in all three models. This demonstrates that porcupine inhibition is potentially therapeutic for symptomatic relief in subjects who suffer from these disorders and further establishes that the continued production of Wnts is necessary for sustaining high bone mass in these models.
Collapse
Affiliation(s)
- Cassandra R Diegel
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Ina Kramer
- Diseases of Aging and Regenerative Medicine, Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Charles Moes
- Diseases of Aging and Regenerative Medicine, Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Gabrielle E Foxa
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Mitchell J McDonald
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Zachary B Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA
| | - Sabine Guth
- Diseases of Aging and Regenerative Medicine, Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Jun Liu
- Oncology, Novartis Institutes for Biomedical Research, San Diego, CA, 92121, USA
| | - Jennifer L Harris
- Oncology, Novartis Institutes for Biomedical Research, San Diego, CA, 92121, USA
| | - Michaela Kneissel
- Diseases of Aging and Regenerative Medicine, Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Bart O Williams
- Department of Cell Biology, Van Andel Institute, 333 Bostwick Ave., NE, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
15
|
Choppa VSR, Kim WK. A Review on Pathophysiology, and Molecular Mechanisms of Bacterial Chondronecrosis and Osteomyelitis in Commercial Broilers. Biomolecules 2023; 13:1032. [PMID: 37509068 PMCID: PMC10377700 DOI: 10.3390/biom13071032] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Modern day broilers have a great genetic potential to gain heavy bodyweights with a huge metabolic demand prior to their fully mature ages. Moreover, this made the broilers prone to opportunistic pathogens which may enter the locomotory organs under stress causing bacterial chondronecrosis and osteomyelitis (BCO). Such pathogenic colonization is further accelerated by microfractures and clefts that are formed in the bones due to rapid growth rate of the broilers along with ischemia of blood vessels. Furthermore, there are several pathways which alter bone homeostasis like acute phase response, and intrinsic and extrinsic cell death pathways. In contrast, all the affected birds may not exhibit clinical lameness even with the presence of lameness associated factors causing infection. Although Staphylococcus, E. coli, and Enterococcus are considered as common bacterial pathogens involved in BCO, but there exist several other non-culturable bacteria. Any deviation from maintaining a homeostatic environment in the gut might lead to bacterial translocation through blood followed by proliferation of pathogenic bacteria in respective organs including bones. It is important to alleviate dysbiosis of the blood which is analogous to dysbiosis in the gut. This can be achieved by supplementing pro, pre, and synbiotics which helps in providing a eubiotic environment abating the bacterial translocation that was studied to the incidence of BCO. This review focused on potential and novel biomarkers, pathophysiological mechanism, the economic significance of BCO, immune mechanisms, and miscellaneous factors causing BCO. In addition, the role of gut microbiomes along with their diversity and cell culture models from compact bones of chicken in better understanding of BCO were explored.
Collapse
Affiliation(s)
| | - Woo Kyun Kim
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
16
|
Otani S, Ohnuma M, Ito K, Matsushita Y. Cellular dynamics of distinct skeletal cells and the development of osteosarcoma. Front Endocrinol (Lausanne) 2023; 14:1181204. [PMID: 37229448 PMCID: PMC10203529 DOI: 10.3389/fendo.2023.1181204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Bone contributes to the maintenance of vital biological activities. At the cellular level, multiple types of skeletal cells, including skeletal stem and progenitor cells (SSPCs), osteoblasts, chondrocytes, marrow stromal cells, and adipocytes, orchestrate skeletal events such as development, aging, regeneration, and tumorigenesis. Osteosarcoma (OS) is a primary malignant tumor and the main form of bone cancer. Although it has been proposed that the cellular origins of OS are in osteogenesis-related skeletal lineage cells with cancer suppressor gene mutations, its origins have not yet been fully elucidated because of a poor understanding of whole skeletal cell diversity and dynamics. Over the past decade, the advent and development of single-cell RNA sequencing analyses and mouse lineage-tracing approaches have revealed the diversity of skeletal stem and its lineage cells. Skeletal stem cells (SSCs) in the bone marrow endoskeletal region have now been found to efficiently generate OS and to be robust cells of origin under p53 deletion conditions. The identification of SSCs may lead to a more limited redefinition of bone marrow mesenchymal stem/stromal cells (BM-MSCs), and this population has been thought to contain cells from which OS originates. In this mini-review, we discuss the cellular diversity and dynamics of multiple skeletal cell types and the origin of OS in the native in vivo environment in mice. We also discuss future challenges in the study of skeletal cells and OS.
Collapse
Affiliation(s)
- Shohei Otani
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mizuho Ohnuma
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Clinical Oral Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
17
|
Lav R, Krivanek J, Anthwal N, Tucker AS. Wnt signaling from Gli1-expressing apical stem/progenitor cells is essential for the coordination of tooth root development. Stem Cell Reports 2023; 18:1015-1029. [PMID: 36931279 PMCID: PMC10147554 DOI: 10.1016/j.stemcr.2023.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023] Open
Abstract
Stem cell regulation plays a crucial role during development and homeostasis. Here, an essential source of Wnts from Gli1+ stem/progenitor cells was identified in the murine molar. Loss of Wnt production in Gli1+ apical stem/progenitor cells led to loss of Axin2 at the root apex, mis-regulation of SOX9, loss of BMP and Hh signaling, and truncation of root development. In the absence of Wnt signals, the root epithelium lost its integrity and epithelial identity. This phenotype could be partially mimicked by loss of Sox9 in the Gli1 population. Stabilization of Wnt signaling in the apical papilla led to rapid unordered differentiation of hard tissues and fragmentation of the epithelial root sheath. Wnt signaling from Gli1+ stem/progenitor cells, therefore, orchestrates root development, coordinating mesenchymal and epithelial interactions via SOX9 to regulate stem/progenitor cell expansion and differentiation. Our results demonstrate that disparate stem/progenitor cell populations are unified in their fundamental signaling interactions.
Collapse
Affiliation(s)
- Rupali Lav
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK.
| |
Collapse
|
18
|
Abstract
Intercellular communication by Wnt proteins governs many essential processes during development, tissue homeostasis and disease in all metazoans. Many context-dependent effects are initiated in the Wnt-producing cells and depend on the export of lipidated Wnt proteins. Although much focus has been on understanding intracellular Wnt signal transduction, the cellular machinery responsible for Wnt secretion became better understood only recently. After lipid modification by the acyl-transferase Porcupine, Wnt proteins bind their dedicated cargo protein Evi/Wntless for transport and secretion. Evi/Wntless and Porcupine are conserved transmembrane proteins, and their 3D structures were recently determined. In this Review, we summarise studies and structural data highlighting how Wnts are transported from the ER to the plasma membrane, and the role of SNX3-retromer during the recycling of its cargo receptor Evi/Wntless. We also describe the regulation of Wnt export through a post-translational mechanism and review the importance of Wnt secretion for organ development and cancer, and as a future biomarker.
Collapse
Affiliation(s)
- Lucie Wolf
- German Cancer Research Center (DKFZ), Division of Signalling and Functional Genomics and Heidelberg University, BioQuant and Department of Cell and Molecular Biology, 69120 Heidelberg, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division of Signalling and Functional Genomics and Heidelberg University, BioQuant and Department of Cell and Molecular Biology, 69120 Heidelberg, Germany
| |
Collapse
|
19
|
Marini F, Giusti F, Palmini G, Brandi ML. Role of Wnt signaling and sclerostin in bone and as therapeutic targets in skeletal disorders. Osteoporos Int 2023; 34:213-238. [PMID: 35982318 DOI: 10.1007/s00198-022-06523-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/01/2022] [Indexed: 01/24/2023]
Abstract
UNLABELLED Wnt signaling and its bone tissue-specific inhibitor sclerostin are key regulators of bone homeostasis. The therapeutic potential of anti-sclerostin antibodies (Scl-Abs), for bone mass recovery and fragility fracture prevention in low bone mass phenotypes, has been supported by animal studies. The Scl-Ab romosozumab is currently used for osteoporosis treatment. INTRODUCTION Wnt signaling is a key regulator of skeletal development and homeostasis; germinal mutations affecting genes encoding components, inhibitors, and enhancers of the Wnt pathways were shown to be responsible for the development of rare congenital metabolic bone disorders. Sclerostin is a bone tissue-specific inhibitor of the Wnt/β-catenin pathway, secreted by osteocytes, negatively regulating osteogenic differentiation and bone formation, and promoting osteoclastogenesis and bone resorption. PURPOSE AND METHODS Here, we reviewed current knowledge on the role of sclerostin and Wnt pathways in bone metabolism and skeletal disorders, and on the state of the art of therapy with sclerostin-neutralizing antibodies in low-bone-mass diseases. RESULTS Various in vivo studies on animal models of human low-bone-mass diseases showed that targeting sclerostin to recover bone mass, restore bone strength, and prevent fragility fracture was safe and effective in osteoporosis, osteogenesis imperfecta, and osteoporosis pseudoglioma. Currently, only treatment with romosozumab, a humanized monoclonal anti-sclerostin antibody, has been approved in human clinical practice for the treatment of osteoporosis, showing a valuable capability to increase BMD at various skeletal sites and reduce the occurrence of new vertebral, non-vertebral, and hip fragility fractures in treated male and female osteoporotic patients. CONCLUSIONS Preclinical studies demonstrated safety and efficacy of therapy with anti-sclerostin monoclonal antibodies in the preservation/restoration of bone mass and prevention of fragility fractures in low-bone-mass clinical phenotypes, other than osteoporosis, to be validated by clinical studies for their approved translation into prevalent clinical practice.
Collapse
Affiliation(s)
- Francesca Marini
- Fondazione FIRMO Onlus, Italian Foundation for the Research on Bone Diseases, Via San Gallo 123, 50129, Florence, Italy
| | - Francesca Giusti
- Donatello Bone Clinic, Villa Donatello Hospital, Sesto Fiorentino, Florence, Italy
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Fondazione FIRMO Onlus, Italian Foundation for the Research on Bone Diseases, Via San Gallo 123, 50129, Florence, Italy.
- Donatello Bone Clinic, Villa Donatello Hospital, Sesto Fiorentino, Florence, Italy.
| |
Collapse
|
20
|
Wnt Signaling in the Development of Bone Metastasis. Cells 2022; 11:cells11233934. [PMID: 36497192 PMCID: PMC9739050 DOI: 10.3390/cells11233934] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Wnt signaling occurs through evolutionarily conserved pathways that affect cellular proliferation and fate decisions during development and tissue maintenance. Alterations in these highly regulated pathways, however, play pivotal roles in various malignancies, promoting cancer initiation, growth and metastasis and the development of drug resistance. The ability of cancer cells to metastasize is the primary cause of cancer mortality. Bone is one of the most frequent sites of metastases that generally arise from breast, prostate, lung, melanoma or kidney cancer. Upon their arrival to the bone, cancer cells can enter a long-term dormancy period, from which they can be reactivated, but can rarely be cured. The activation of Wnt signaling during the bone metastasis process was found to enhance proliferation, induce the epithelial-to-mesenchymal transition, promote the modulation of the extracellular matrix, enhance angiogenesis and immune tolerance and metastasize and thrive in the bone. Due to the complexity of Wnt pathways and of the landscape of this mineralized tissue, Wnt function during metastatic progression within bone is not yet fully understood. Therefore, we believe that a better understanding of these pathways and their roles in the development of bone metastasis could improve our understanding of the disease and may constitute fertile ground for potential therapeutics.
Collapse
|
21
|
Vlashi R, Zhang X, Wu M, Chen G. Wnt signaling: essential roles in osteoblast differentiation, bone metabolism and therapeutic implications for bone and skeletal disorders. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.07.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
22
|
Sharma AR, Lee YH, Gankhuyag B, Chakraborty C, Lee SS. Effect of Alumina Particles on the Osteogenic Ability of Osteoblasts. J Funct Biomater 2022; 13:jfb13030105. [PMID: 35997443 PMCID: PMC9397023 DOI: 10.3390/jfb13030105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Biomaterials are used as implants for bone and dental disabilities. However, wear particles from the implants cause osteolysis following total joint arthroplasty (TJA). Ceramic implants are considered safe and elicit a minimal response to cause periprosthetic osteolysis. However, few reports have highlighted the adverse effect of ceramic particles such as alumina (Al2O3) on various cell types. Hence, we aimed to investigate the effect of Al2O3 particles on osteoprogenitors. A comparative treatment of Al2O3, Ti, and UHMWPE particles to osteoprogenitors at a similar concentration of 200 μg/mL showed that only Al2O3 particles were able to suppress the early and late differentiation markers of osteoprogenitors, including collagen synthesis, alkaline phosphatase (ALP) activity and mRNA expression of Runx2, OSX, Col1α, and OCN. Al2O3 particles even induced inflammation and activated the NFkB signaling pathway in osteoprogenitors. Moreover, bone-forming signals such as the WNT/β-catenin signaling pathway were inhibited by the Al2O3 particles. Al2O3 particles were found to induce the mRNA expression of WNT/β-catenin signaling antagonists such as DKK2, WIF, and sFRP1 several times in osteoprogenitors. Taken together, this study highlights a mechanistic view of the effect of Al2O3 particles on osteoprogenitors and suggests therapeutic targets such as NFĸB and WNT signaling pathways for ceramic particle-induced osteolysis.
Collapse
Affiliation(s)
- Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon 24252, Korea; (A.R.S.); (Y.-H.L.); (B.G.)
| | - Yeon-Hee Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon 24252, Korea; (A.R.S.); (Y.-H.L.); (B.G.)
| | - Buyankhishig Gankhuyag
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon 24252, Korea; (A.R.S.); (Y.-H.L.); (B.G.)
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Rd, Kolkata 700126, India;
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon 24252, Korea; (A.R.S.); (Y.-H.L.); (B.G.)
- Correspondence:
| |
Collapse
|
23
|
Gu R, Zhang S, Saha SK, Ji Y, Reynolds K, McMahon M, Sun B, Islam M, Trainor PA, Chen Y, Xu Y, Chai Y, Burkart-Waco D, Zhou CJ. Single-cell transcriptomic signatures and gene regulatory networks modulated by Wls in mammalian midline facial formation and clefts. Development 2022; 149:dev200533. [PMID: 35781558 PMCID: PMC9382898 DOI: 10.1242/dev.200533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/21/2022] [Indexed: 07/24/2023]
Abstract
Formation of highly unique and complex facial structures is controlled by genetic programs that are responsible for the precise coordination of three-dimensional tissue morphogenesis. However, the underlying mechanisms governing these processes remain poorly understood. We combined mouse genetic and genomic approaches to define the mechanisms underlying normal and defective midfacial morphogenesis. Conditional inactivation of the Wnt secretion protein Wls in Pax3-expressing lineage cells disrupted frontonasal primordial patterning, cell survival and directional outgrowth, resulting in altered facial structures, including midfacial hypoplasia and midline facial clefts. Single-cell RNA sequencing revealed unique transcriptomic atlases of mesenchymal subpopulations in the midfacial primordia, which are disrupted in the conditional Wls mutants. Differentially expressed genes and cis-regulatory sequence analyses uncovered that Wls modulates and integrates a core gene regulatory network, consisting of key midfacial regulatory transcription factors (including Msx1, Pax3 and Pax7) and their downstream targets (including Wnt, Shh, Tgfβ and retinoic acid signaling components), in a mesenchymal subpopulation of the medial nasal prominences that is responsible for midline facial formation and fusion. These results reveal fundamental mechanisms underlying mammalian midfacial morphogenesis and related defects at single-cell resolution.
Collapse
Affiliation(s)
- Ran Gu
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Subbroto Kumar Saha
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Moira McMahon
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Mohammad Islam
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - YiPing Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Ying Xu
- Can-SU Genomic Resource Center, Medical College of Soochow University, Suzhou 215006, China
| | - Yang Chai
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | - Diana Burkart-Waco
- DNA Technologies and Expression Analysis Core, Genome Center, University of California, Davis, California 95616, USA
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children and UC Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
24
|
Zhou D, Wang Y, Gui Y, Fu H, Zhou S, Wang Y, Bastacky SI, Stolz DB, Liu Y. Non-canonical Wnt/calcium signaling is protective against podocyte injury and glomerulosclerosis. Kidney Int 2022; 102:96-107. [PMID: 35341792 PMCID: PMC9232939 DOI: 10.1016/j.kint.2022.02.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
Activation of canonical Wnt signaling has been implicated in podocyte injury and proteinuria. As Wnts are secreted proteins, whether Wnts derived from podocytes are obligatory for promoting proteinuria remains unknown. To address this, we generated conditional knockout mice where Wntless, a cargo receptor protein required for Wnt secretion, was specifically deleted in glomerular podocytes. Mice with podocyte-specific ablation of Wntless (Podo-Wntless-/-) were phenotypically normal. However, after inducing kidney damage with Adriamycin for six days, Podo-Wntless-/- mice developed more severe podocyte injury and albuminuria than their control littermates. Surprisingly, ablation of Wntless resulted in upregulation of β-catenin, accompanied by reduction of nephrin, podocin, podocalyxin, and Wilms tumor 1 proteins. In chronic injury induced by Adriamycin, increased albuminuria, aggravated podocyte lesions and extracellular matrix deposition were evident in Podo-Wntlessl-/- mice, compared to wild type mice. Mechanistically, specific ablation of Wntless in podocytes caused down-regulation of the nuclear factor of activated T cell 1 (NFAT1) and Nemo-like kinase (NLK), key downstream mediators of non-canonical Wnt/calcium signaling. In vitro, knockdown of either NFAT1 or NLK induced β-catenin activation while overexpression of NLK significantly repressed β-catenin induction and largely preserved nephrin in glomerular podocytes. Thus, our results indicate that podocyte-derived Wnts play an important role in protecting podocytes from injury by repressing β-catenin via activating non-canonical Wnt/calcium signaling.
Collapse
Affiliation(s)
- Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Yuanyuan Wang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Department of Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Yuan Gui
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Haiyan Fu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shanshan Zhou
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Donna B Stolz
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Xu Q, Li D, Chen J, Yang J, Yan J, Xia Y, Zhang F, Wang X, Cao H. Crosstalk between the gut microbiota and postmenopausal osteoporosis: Mechanisms and applications. Int Immunopharmacol 2022; 110:108998. [PMID: 35785728 DOI: 10.1016/j.intimp.2022.108998] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 12/14/2022]
Abstract
Postmenopausal osteoporosis (PMO) results from a reduction in bone mass and microarchitectural deterioration in bone tissue due to estrogen deficiency, which may increase the incidence of fragility fractures. The number of people suffering from PMO has increased over the years because of the rapidly aging population worldwide. However, several pharmacological agents for the treatment of PMO have many safety risks and impose a heavy financial burden to patients and society. In recent years, the "gut-bone" axis has been proposed as a new approach in the prevention and treatment of PMO. This paper reviews the relationship between the gut microbiota and PMO, which mainly includes the underlying mechanisms between hormones, immunity, nutrient metabolism, metabolites of the gut microbiota and intestinal permeability, and explores the possible role of the gut microbiota in these processes. Finally, we discuss the therapeutic effects of diet, prebiotics, probiotics, and fecal microbiota transplantation on the gut microbiota.
Collapse
Affiliation(s)
- Qin Xu
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Dan Li
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jing Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Nursing Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Ju Yang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Jiai Yan
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yanping Xia
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Zhang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xuesong Wang
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hong Cao
- Nutrition Department, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China; Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
26
|
Thomas S, Jaganathan BG. Signaling network regulating osteogenesis in mesenchymal stem cells. J Cell Commun Signal 2022; 16:47-61. [PMID: 34236594 PMCID: PMC8688675 DOI: 10.1007/s12079-021-00635-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Osteogenesis is an important developmental event that results in bone formation. Bone forming cells or osteoblasts develop from mesenchymal stem cells (MSCs) through a highly controlled process regulated by several signaling pathways. The osteogenic lineage commitment of MSCs is controlled by cell-cell interactions, paracrine factors, mechanical signals, hormones, and cytokines present in their niche, which activate a plethora of signaling molecules belonging to bone morphogenetic proteins, Wnt, Hedgehog, and Notch signaling. These signaling pathways individually as well as in coordination with other signaling molecules, regulate the osteogenic lineage commitment of MSCs by activating several osteo-lineage specific transcription factors. Here, we discuss the key signaling pathways that regulate osteogenic differentiation of MSCs and the cross-talk between them during osteogenic differentiation. We also discuss how these signaling pathways can be modified for therapy for bone repair and regeneration.
Collapse
Affiliation(s)
- Sachin Thomas
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Bithiah Grace Jaganathan
- Stem Cells and Cancer Biology Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India.
| |
Collapse
|
27
|
Tripathi AK, Rai D, Kothari P, Kushwaha P, Sashidhara KV, Trivedi R. Benzofuran pyran hybrid prevents glucocorticoid induced osteoporosis in mice via modulation of canonical Wnt/β-catenin signaling. Apoptosis 2022; 27:90-111. [PMID: 35107658 PMCID: PMC8808472 DOI: 10.1007/s10495-021-01702-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 11/26/2022]
Abstract
Glucocorticoid induced osteoporosis (GIOP) is the second most leading cause of osteoporosis. We have identified a compound, a benzofuran pyran hybrid compound 4e that has osteogenic potential and we wanted to assess its efficacy in GIOP in male mice. We assessed the effect of dexamethasone and compound 4e on primary osteoblasts using various cell based and immunofluorescence assays. For in vivo studies we administered methylprednisolone and compound 4e as a prophylactic measure in male Balb/c mice for 28 days and then evaluated the effect on bone microarchitecture by microCT, bone formation by histology along with clinically relevant bone markers. Compound 4e preserved osteoblast differentiation as evident by higher ALP positive cells and mineralization in compound treated groups. Compound 4e also increased the expression of osteogenic genes. This compound guarded β-catenin expression both in vitro and in vivo as confirmed by western blot and immunofluorescence assays. This led to the preservation of bone microarchitecture and cortical thickness at 2.5 mg kg−1 and 5 mg kg−1 doses. Further compound 4e enhanced bone formation rate and regulated osteocyte death. The osteogenic potential of compound 4e was reflected by an increased level of serum marker osteocalcin and decreased levels of SOST and CTX-I. Overall, Compound 4e is able to overcome the catabolic effect of dexamethasone on bone by targeting the canonical WNT/β-catenin signaling as evidenced by both in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ashish Kumar Tripathi
- Endocrinology Division, CSIR-Central Drug Research Institute, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
| | - Divya Rai
- Endocrinology Division, CSIR-Central Drug Research Institute, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Priyanka Kothari
- Endocrinology Division, CSIR-Central Drug Research Institute, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
| | - Pragati Kushwaha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Ritu Trivedi
- Endocrinology Division, CSIR-Central Drug Research Institute, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
28
|
Lawson LY, Brodt MD, Migotsky N, Chermside-Scabbo CJ, Palaniappan R, Silva MJ. Osteoblast-Specific Wnt Secretion Is Required for Skeletal Homeostasis and Loading-Induced Bone Formation in Adult Mice. J Bone Miner Res 2022; 37:108-120. [PMID: 34542191 PMCID: PMC8770559 DOI: 10.1002/jbmr.4445] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/10/2021] [Accepted: 08/28/2021] [Indexed: 01/03/2023]
Abstract
Wnt signaling is critical to many aspects of skeletal regulation, but the importance of Wnt ligands in the bone anabolic response to mechanical loading is not well established. Recent transcriptome profiling studies by our laboratory and others show that mechanical loading potently induces genes encoding Wnt ligands, including Wnt1 and Wnt7b. Based on these findings, we hypothesized that mechanical loading stimulates adult bone formation by inducing Wnt ligand expression. To test this hypothesis, we inhibited Wnt ligand secretion in adult (5 months old) mice using a systemic (drug) and a bone-targeted (conditional gene knockout) approach, and subjected them to axial tibial loading to induce lamellar bone formation. Mice treated with the Wnt secretion inhibitor WNT974 exhibited a decrease in bone formation in non-loaded bones as well as a 54% decline in the periosteal bone formation response to tibial loading. Next, osteoblast-specific Wnt secretion was inhibited by dosing 5-month-old Osx-CreERT2; WlsF/F mice with tamoxifen. Within 1 to 2 weeks of Wls deletion, skeletal homeostasis was altered with decreased bone formation and increased resorption, and the anabolic response to loading was reduced 65% compared to control (WlsF/F ). Together, these findings show that Wnt ligand secretion is required for adult bone homeostasis, and furthermore establish a role for osteoblast-derived Wnts in mediating the bone anabolic response to tibial loading. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lisa Y. Lawson
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Michael D. Brodt
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Nicole Migotsky
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| | - Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Ramya Palaniappan
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Matthew J. Silva
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| |
Collapse
|
29
|
Sim HJ, So HS, Poudel SB, Bhattarai G, Cho ES, Lee JC, Kook SH. Osteoblastic Wls Ablation Protects Mice from Total Body Irradiation-Induced Impairments in Hematopoiesis and Bone Marrow Microenvironment. Aging Dis 2022; 14:919-936. [PMID: 37191410 DOI: 10.14336/ad.2022.1026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Ionizing irradiation (IR) causes bone marrow (BM) injury, with senescence and impaired self-renewal of hematopoietic stem cells (HSCs), and inhibiting Wnt signaling could enhance hematopoietic regeneration and survival against IR stress. However, the underlying mechanisms by which a Wnt signaling blockade modulates IR-mediated damage of BM HSCs and mesenchymal stem cells (MSCs) are not yet completely understood. We investigated the effects of osteoblastic Wntless (Wls) depletion on total body irradiation (TBI, 5 Gy)-induced impairments in hematopoietic development, MSC function, and the BM microenvironment using conditional Wls knockout mutant mice (Col-Cre;Wlsfl/fl) and their littermate controls (Wlsfl/fl). Osteoblastic Wls ablation itself did not dysregulate BM frequency or hematopoietic development at a young age. Exposure to TBI at 4 weeks of age induced severe oxidative stress and senescence in the BM HSCs of Wlsfl/fl mice but not in those of the Col-Cre;Wlsfl/fl mice. TBI-exposed Wlsfl/fl mice exhibited greater impairments in hematopoietic development, colony formation, and long-term repopulation than TBI-exposed Col-Cre;Wlsfl/fl mice. Transplantation with BM HSCs or whole BM cells derived from the mutant, but not Wlsfl/fl mice, protected against HSC senescence and hematopoietic skewing toward myeloid cells and enhanced survival in recipients of lethal TBI (10 Gy). Unlike the Wlsfl/fl mice, the Col-Cre;Wlsfl/fl mice also showed radioprotection against TBI-mediated MSC senescence, bone mass loss, and delayed body growth. Our results indicate that osteoblastic Wls ablation renders BM-conserved stem cells resistant to TBI-mediated oxidative injuries. Overall, our findings show that inhibiting osteoblastic Wnt signaling promotes hematopoietic radioprotection and regeneration.
Collapse
|
30
|
Chen T, Gao F, Luo D, Wang S, Zhao Y, Liu S, Huang J, Lin Y, Zhang Z, Huang H, Wan L. Cistanoside A promotes osteogenesis of primary osteoblasts by alleviating apoptosis and activating autophagy through involvement of the Wnt/β-catenin signal pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:64. [PMID: 35282110 PMCID: PMC8848445 DOI: 10.21037/atm-21-6742] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
Background As a phenylethanoid glycoside extracted from Cistanche deserticola, cistanoside A has been shown to have antioxidative effects. In recent years, it has been found to play an important role in osteoporosis. Methods Primary osteoblasts were randomly divided into a cistanoside A (Cis A)-1 group (5 µM), a Cis A-2 group (10 µM), and a Cis A-3 group (20 µM) to screen the optimal dose. Then, cells were treated with Rapamycin (Rapa), 3-MA, Dickkopf-1 (DKK-1), 3MA + Cis A (10 µM), and DKK-1 + Cis A (10 µM). After a certain period of routine culture, Alkaline Phosphatase (ALP) and Alizarin Red S Staining were performed again and the cells were collected for subsequent experiments including immunofluorescence staining, western blotting, transmission electron microscopy, mitochondrial membrane measurement, and Annexin-V-Fluorescein isothiocyanate (Annexin-V-FITC). Results The optimal Cis A dose that preserved osteoblast viability and activated osteogenesis was 10 µM. It appeared that Cis A (10 µM) decreased apoptosis and augmented autophagy via increasing microtubule-associated protein light chain 3 (LC3)-I/II expressions as well as raising Wnt/β-catenin signal pathway activity. The addition of 3-MA further inhibited osteogenic differentiation and suppressed Wnt/β-catenin signal pathway activity to increase apoptosis while reducing autophagy levels. A combination of Cis A and DKK-1 resulted in higher levels of apoptosis but lower levels of autophagy. Conclusions Cis A appears to be a potent inducer of autophagy and inhibitor of apoptosis in primary osteoblasts by working through the Wnt/β-catenin signal pathway, thereby resulting in enhanced osteogenic differentiation.
Collapse
Affiliation(s)
- Tongying Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fenghe Gao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Luo
- Research Laboratory of Spine Degenerative Disease, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shihao Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhua Liu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiachun Huang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanping Lin
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhihai Zhang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongxing Huang
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Wan
- Department of Osteoporosis, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
31
|
Wang LT, Lin MH, Liu KY, Chiou SS, Wang SN, Chai CY, Tseng LW, Chiou HYC, Wang HC, Yokoyama KK, Hsu SH, Huang SK. WLS/wntless is essential in controlling dendritic cell homeostasis via a WNT signaling-independent mechanism. Autophagy 2021; 17:4202-4217. [PMID: 33853474 PMCID: PMC8726611 DOI: 10.1080/15548627.2021.1907516] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
We propose that beyond its role in WNT secretion, WLS/GPR177 (wntless, WNT ligand secretion mediator) acts as an essential regulator controlling protein glycosylation, endoplasmic reticulum (ER) homeostasis, and dendritic cell (DC)-mediated immunity. WLS deficiency in bone marrow-derived DCs (BMDCs) resulted in poor growth and an inability to mount cytokine and T-cell responses in vitro, phenotypes that were irreversible by the addition of exogenous WNTs. In fact, WLS was discovered to integrate a protein complex in N-glycan-dependent and WLS domain-selective manners, comprising ER stress sensors and lectin chaperones. WLS deficiency in BMDCs led to increased ER stress response and macroautophagy/autophagy, decreased calcium efflux from the ER, and the loss of CALR (calreticulin)-CANX (calnexin) cycle, and hence protein hypo-glycosylation. Consequently, DC-specific wls-null mice were unable to develop both Th1-, Th2- and Th17-associated responses in the respective autoimmune and allergic disease models. These results suggest that WLS is a critical chaperone in maintaining ER homeostasis, glycoprotein quality control and calcium dynamics in DCs.Abbreviations: ATF6: activating transcription factor 6; ATG5: autophagy related 5; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; ATP2A1/SERCA1: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1; BALF: bronchoalveolar lavage fluid; BFA: brefeldin A; BMDC: bone marrow-derived dendritic cell; CALR: calreticulin; CANX: calnexin; CCL2/MCP-1: C-C motif chemokine ligand 2; CNS: central nervous system; CT: C-terminal domain; DTT: dithiothreitol; DNAJB9/ERDJ4: DnaJ heat shock protein family (Hsp40) member B9; EAE: experimental autoimmune encephalomyelitis; EIF2A/eIF2α: eukaryotic translation initiation factor 2A; EIF2AK3/PERK: eukaryotic translation initiation factor 2 alpha kinase 3; ERN1/IRE1: endoplasmic reticulum (ER) to nucleus signaling 1; GFP: green fluorescent protein; HSPA5/GRP78/BiP: heat shock protein A5; IFNA: interferon alpha; IFNAR1: interferon alpha and beta receptor subunit 1; IFNB: interferon beta; IFNG/INFγ: interferon gamma; IFNGR2: interferon gamma receptor 2; IL6: interleukin 6; IL10: interleukin 10; IL12A: interleukin 12A; IL23A: interleukin 23 subunit alpha; ITGAX/CD11c: integrin subunit alpha X; ITPR1/InsP3R1: inositol 1,4,5-trisphosphate receptor type 1; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; OVA: ovalbumin; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PLF: predicted lipocalin fold; PPP1R15A/GADD34: protein phosphatase 1 regulatory subunit 15A; RYR1/RyanR1: ryanodine receptor 1, skeletal muscle; SD: signal domain; TGFB/TGF-β: transforming growth factor beta family; Th1: T helper cell type 1; Th17: T helper cell type 17; TM: tunicamycin; TNF/TNF-α: tumor necrosis factor; UPR: unfolded protein response; WLS/wntless: WNT ligand secretion mediator.
Collapse
Affiliation(s)
- Li-Ting Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ming-Hong Lin
- D Department of Microbiology and Immunology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kwei-Yan Liu
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Shyh-Shin Chiou
- Department of Pathology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Hematology-Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shen-Nien Wang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chee-Yin Chai
- Department of Pathology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Wen Tseng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-Ying Clair Chiou
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Medical Education and Research Center, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsueh-Chun Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center of Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shih-Hsien Hsu
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shau-Ku Huang
- Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli County, Taiwan
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
32
|
Study of selected genes of Wnt signaling pathway in relation to the parameters in the bone tissue of the laying hens. Saudi J Biol Sci 2021; 29:2526-2531. [PMID: 35531234 PMCID: PMC9072936 DOI: 10.1016/j.sjbs.2021.12.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/29/2021] [Accepted: 12/12/2021] [Indexed: 11/02/2022] Open
Abstract
The Wnt signaling pathway plays a critical role in almost all aspects of skeletal development and homeostasis. Many studies suggest the importance of this signaling pathway in connection with bone metabolism through many skeletal disorders caused by mutations in Wnt signaling genes. The knowledge gained through targeting this pathway is of great value for skeletal health and diseases, for example of increased bone mass in the case of osteoporosis. Our objective was to focus on the detection of single nucleotide polymorphisms and investigate the associations between possible polymorphisms in selected genes that are part of those signaling pathways and parameters of bones in hens of ISA Brown hybrids (bone breaking strength, length, width, and bone mass). Different regions of the GPR177, ESR1 and RUNX2 genes were studied, using PCR and sequencing, in a total of forty-eight samples for each marker. Thirteen polymorphisms have been discovered in selected regions of studied genes, whereas these polymorphisms were only within the GPR177 gene. Eight of these polymorphisms were synonymous and five were in the intron. The tested regions of the ESR1 and RUNX2 genes were monomorphic. The only statistically significant difference was found within the GPR177 gene (exon 2) and the bone length parameter, in the c.443 + 86G > A polymorphism. However, this polymorphism was found in the intron, and no other one was found within the selected regions to show associations with the observed bone parameters.
Collapse
|
33
|
Xiong L, Pan JX, Guo HH, Mei L, Xiong WC. Parkinson's in the bone. Cell Biosci 2021; 11:190. [PMID: 34740382 PMCID: PMC8569842 DOI: 10.1186/s13578-021-00702-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
Patients with Parkinson’s disease (PD) exhibit systemic deficits, including arthritis and osteoporosis-like symptoms. However, the questions, how the deficits in periphery organs or tissues occur in PD patients, and what are the relationship (s) of the periphery tissue deficits with the brain pathology (e.g., dopamine neuron loss), are at the beginning stage to be investigated. Notice that both PD and osteoporosis are the products of a complex interaction of genetic and environmental risk factors. Genetic mutations in numerous genes have been identified in patients either with recessive or autosomal dominant PD. Most of these PD risk genes are ubiquitously expressed; and many of them are involved in regulation of bone metabolism. Here, we review the functions of the PD risk genes in regulating bone remodeling and homeostasis. The knowledge gaps in our understanding of the bone-to-brain axis in PD development are also outlined.
Collapse
Affiliation(s)
- Lei Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Jin-Xiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Hao-Han Guo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stoke VA Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
34
|
Cheng L, Li Y, Xia Q, Meng M, Ye Z, Tang Z, Feng H, Chen X, Chen H, Zeng X, Luo Y, Dong Q. Enamel matrix derivative (EMD) enhances the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Bioengineered 2021; 12:7033-7045. [PMID: 34587869 PMCID: PMC8806549 DOI: 10.1080/21655979.2021.1971504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
To investigate the EMD's capacity in BMSCs osteogenic differentiation. In vivo and in vitro, BMSCs were treated with EMD, scanning electron microscopy, and Alizarin Red staining were used to detect the changes in the osteogenic ability of BMSCs, and the proliferation ability of BMSCs was evaluated by CCK8. In addition, by adding xav939, a typical inhibitor of Wnt/β-catenin signaling pathway, the regulatory function of Wnt/β-catenin signaling was clarified. The results showed that EMD promote cell proliferation and 25 μg/ml EMD had the most significant effect. Cells inducing osteogenesis for 2 and 3 even 4 weeks, the cell staining is deeper in EMD treated group than that of the control (P < 0.05) by alizarin Red staining, suggesting more mineralization of BMSCs. In vivo implanting the titanium plate wrapped with 25 μg/ml EMD treated-BMSC film into nude mice for 8 weeks, more nodules were formed on the surface of the titanium plate than that the control (P < 0.05). HE showed that there is a little blue-violet immature bone-like tissue block. Besides, the expression of RUNX Family Transcription Factor 2 (Runx2), Osterix, Osteocalcin (OCN), collagen I (COLI), alkaline phosphatase (ALP) and β-catenin were inhibited in xav939 group (P < 0.05); Inversely, all were activated in EMD group (P < 0.05). In conclusion, EMD promoted the proliferation and osteogenic differentiation of BMSCs. EMD's function on BMSCs might be associated with the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Lu Cheng
- Department of Prosthodontics, Guiyang Hospital of Stomatology, Guiyang, Gsuizhou Province, 550002, People's Republic of China
| | - Ying Li
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Qian Xia
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - MaoHua Meng
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - ZhaoYang Ye
- Clinical Research Center, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - ZhengLong Tang
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China.,Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - HongChao Feng
- Department of Oral and Maxillofacial Surgery, Guiyang Hospital of Stomatology, Guiyang, Guizhou Province, 550002, People's Republic of China
| | - Xin Chen
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - HeLin Chen
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Xiao Zeng
- Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| | - Yi Luo
- Department of Prosthodontics, Guiyang Hospital of Stomatology, Guiyang, Gsuizhou Province, 550002, People's Republic of China
| | - Qiang Dong
- Department of Prosthodontics, School of Stomatology, Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China.,Department of Prosthodonticsand Oral Implantology, Stomatological hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550004, People's Republic of China
| |
Collapse
|
35
|
Nilsson KH, Henning P, El Shahawy M, Nethander M, Andersen TL, Ejersted C, Wu J, Gustafsson KL, Koskela A, Tuukkanen J, Souza PPC, Tuckermann J, Lorentzon M, Ruud LE, Lehtimäki T, Tobias JH, Zhou S, Lerner UH, Richards JB, Movérare-Skrtic S, Ohlsson C. RSPO3 is important for trabecular bone and fracture risk in mice and humans. Nat Commun 2021; 12:4923. [PMID: 34389713 PMCID: PMC8363747 DOI: 10.1038/s41467-021-25124-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/19/2021] [Indexed: 11/22/2022] Open
Abstract
With increasing age of the population, countries across the globe are facing a substantial increase in osteoporotic fractures. Genetic association signals for fractures have been reported at the RSPO3 locus, but the causal gene and the underlying mechanism are unknown. Here we show that the fracture reducing allele at the RSPO3 locus associate with increased RSPO3 expression both at the mRNA and protein levels, increased trabecular bone mineral density and reduced risk mainly of distal forearm fractures in humans. We also demonstrate that RSPO3 is expressed in osteoprogenitor cells and osteoblasts and that osteoblast-derived RSPO3 is the principal source of RSPO3 in bone and an important regulator of vertebral trabecular bone mass and bone strength in adult mice. Mechanistic studies revealed that RSPO3 in a cell-autonomous manner increases osteoblast proliferation and differentiation. In conclusion, RSPO3 regulates vertebral trabecular bone mass and bone strength in mice and fracture risk in humans.
Collapse
Affiliation(s)
- Karin H Nilsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Maha El Shahawy
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Faculty of Dentistry, Department of Oral Biology, Minia University, Minia, Egypt
| | - Maria Nethander
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Thomas Levin Andersen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin L Gustafsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Pedro P C Souza
- Innovation in Biomaterials Laboratory, Faculty of Dentistry, Federal University of Goiás, Goiâna, Brazil
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Mattias Lorentzon
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Geriatric Medicine, Sahlgrenska University Hospital, Mölndal, Sweden
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Linda Engström Ruud
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jon H Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, and Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Sirui Zhou
- Department of Medicine, Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Ulf H Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - J Brent Richards
- Department of Medicine, Centre for Clinical Epidemiology, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
- Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
36
|
He J, Ling L, Liu Z, Ren X, Wan L, Tu C, Li Z. Functional interplay between long non-coding RNAs and the Wnt signaling cascade in osteosarcoma. Cancer Cell Int 2021; 21:313. [PMID: 34130697 PMCID: PMC8207720 DOI: 10.1186/s12935-021-02013-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma is a common and highly malignant bone tumor among children, adolescents and young adults. However, the underlying molecular mechanisms remain largely unexplored. LncRNAs are transcripts with no or limited protein-coding capacity in human genomes, and have been demonstrated to play crucial functions in initiation, progression, therapeutic resistance, recurrence and metastasis of tumor. Considerable studies revealed a dysregulated lncRNA expression pattern in osteosarcoma, which may act as oncogenes or suppressors to regulate osteosarcoma progression. Wnt signaling pathway is an important cascade in tumorigenesis by modulation of pleiotropic biological functions including cell proliferation, apoptosis, differentiation, stemness, genetic stability and chemoresistance. Hyperactivation or deficiency of key effectors in Wnt cascade is a common event in many osteosarcoma patients. Recently, increasing evidences have suggested that lncRNAs could interplay with component of Wnt pathway, and thereby contribute to osteosarcoma onset, progression and dissemination. In this review, we briefly summarize Wnt signaling-related lncRNAs in osteosarcoma progression, aiming to gain insights into their underlying crosstalk as well as clinical application in osteosarcoma therapeutic modalities.
Collapse
Affiliation(s)
- Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lin Ling
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Zhongyue Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Xiaolei Ren
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lu Wan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, No 139 Middle Renmin Road, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
37
|
Nilsson KH, Henning P, El Shahawy M, Wu J, Koskela A, Tuukkanen J, Perret C, Lerner UH, Ohlsson C, Movérare-Skrtic S. Osteocyte- and late osteoblast-derived NOTUM reduces cortical bone mass in mice. Am J Physiol Endocrinol Metab 2021; 320:E967-E975. [PMID: 33749332 DOI: 10.1152/ajpendo.00565.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Osteoporosis is a common skeletal disease, with increased risk of fractures. Currently available osteoporosis treatments reduce the risk of vertebral fractures, mainly dependent on trabecular bone, whereas the effect on nonvertebral fractures, mainly dependent on cortical bone, is less pronounced. WNT signaling is a crucial regulator of bone homeostasis, and the activity of WNTs is inhibited by NOTUM, a secreted WNT lipase. We previously demonstrated that conditional inactivation of NOTUM in all osteoblast lineage cells increases the cortical but not the trabecular bone mass. The aim of the present study was to determine if NOTUM increasing cortical bone is derived from osteoblast precursors/early osteoblasts or from osteocytes/late osteoblasts. First, we demonstrated Notum mRNA expression in Dmp1-expressing osteocytes and late osteoblasts in cortical bone using in situ hybridization. We then developed a mouse model with inactivation of NOTUM in Dmp1-expressing osteocytes and late osteoblasts (Dmp1-creNotumflox/flox mice). We observed that the Dmp1-creNotumflox/flox mice displayed a substantial reduction of Notum mRNA in cortical bone, resulting in increased cortical bone mass and decreased cortical porosity in femur but no change in trabecular bone volume fraction in femur or in the lumbar vertebrae L5 in Dmp1-creNotumflox/flox mice as compared with control mice. In conclusion, osteocytes and late osteoblasts are the principal source of NOTUM in cortical bone, and NOTUM derived from osteocytes/late osteoblasts reduces cortical bone mass. These findings demonstrate that inhibition of osteocyte/late osteoblast-derived NOTUM might be an interesting pharmacological target to increase cortical bone mass and reduce nonvertebral fracture risk.NEW & NOTEWORTHY NOTUM produced by osteoblasts is known to regulate cortical bone mass. Our new findings show that NOTUM specifically derived by DMP1-expressing osteocytes and late osteoblasts regulates cortical bone mass and not trabecular bone mass.
Collapse
Affiliation(s)
- Karin H Nilsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maha El Shahawy
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oral Biology, Minia University, Minia, Egypt
| | - Jianyao Wu
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Christine Perret
- Université de Paris, Institut Cochin, INSERM, CNRS, Paris, France
| | - Ulf H Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sofia Movérare-Skrtic
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, Riminucci M. From Stem Cells to Bone-Forming Cells. Int J Mol Sci 2021; 22:ijms22083989. [PMID: 33924333 PMCID: PMC8070464 DOI: 10.3390/ijms22083989] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022] Open
Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
Collapse
Affiliation(s)
- Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA;
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Correspondence:
| |
Collapse
|
39
|
Li C, Pi G, Li F. The Role of Intestinal Flora in the Regulation of Bone Homeostasis. Front Cell Infect Microbiol 2021; 11:579323. [PMID: 33777828 PMCID: PMC7994858 DOI: 10.3389/fcimb.2021.579323] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
Intestinal flora located within the intestinal tract comprises a large number of cells, which are referred to as the second gene pool of the human body and form a complex symbiotic relationship with the host. The knowledge of the complex interaction between the intestinal flora and various life activities of the host is a novel and rapidly expanding field. Recently, many studies are being conducted on the relationship between the intestinal flora and bone homeostasis and indicate that the intestinal flora can regulate bone homeostasis via the host immune, metabolic, and endocrine systems. What’s more, based on several clinical and preclinical pieces of evidence, changing the composition and function of the host intestinal flora through the application of probiotics, prebiotics, and fecal microbiota transplantation is being considered to be a potential novel target for the regulation of bone homeostasis. Here, we searched relevant literature and reviewed the role of the intestinal flora in the regulation of bone homeostasis and its modulating interventions.
Collapse
Affiliation(s)
- Chengxiang Li
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guofu Pi
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
40
|
Nygaard R, Yu J, Kim J, Ross DR, Parisi G, Clarke OB, Virshup DM, Mancia F. Structural Basis of WLS/Evi-Mediated Wnt Transport and Secretion. Cell 2021; 184:194-206.e14. [PMID: 33357447 PMCID: PMC7797000 DOI: 10.1016/j.cell.2020.11.038] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/26/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
Wnts are evolutionarily conserved ligands that signal at short range to regulate morphogenesis, cell fate, and stem cell renewal. The first and essential steps in Wnt secretion are their O-palmitoleation and subsequent loading onto the dedicated transporter Wntless/evenness interrupted (WLS/Evi). We report the 3.2 Å resolution cryogenic electron microscopy (cryo-EM) structure of palmitoleated human WNT8A in complex with WLS. This is accompanied by biochemical experiments to probe the physiological implications of the observed association. The WLS membrane domain has close structural homology to G protein-coupled receptors (GPCRs). A Wnt hairpin inserts into a conserved hydrophobic cavity in the GPCR-like domain, and the palmitoleate protrudes between two helices into the bilayer. A conformational switch of highly conserved residues on a separate Wnt hairpin might contribute to its transfer to receiving cells. This work provides molecular-level insights into a central mechanism in animal body plan development and stem cell biology.
Collapse
Affiliation(s)
- Rie Nygaard
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jia Yu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Daniel R Ross
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Giacomo Parisi
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David M Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore; Department of Pediatrics, Duke University School of Medicine, Durham, NC 27705, USA.
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
41
|
Zhou L, Wang J, Zhao J, Kuai F, Yang H. Shikonin promotes osteogenesis and suppresses osteoclastogenesis in vitro. Am J Transl Res 2020; 12:8099-8110. [PMID: 33437384 PMCID: PMC7791499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Shikonin, as a traditional Chinese herbal medicine with a role of anti-cancer, anti-inflammatory, anti-bacterial and other effects. However, there are few studies on the effect of shikonin on osteoporosis. Therefore, the purpose of this study aims to investigate the role and mechanism of shikonin on differentiation of BMSCs and BMMs into osteoblasts and osteoclasts formation. In our study, we treated the cells with different concentrations of shikonin, and then illuminated its effect on oteogenesis and osteoclast differentiation by ALP/alizarin red staining, ALP activity, qRT-PCR, immunofluorescence, Western blot, and TRAP staining. The result showed that shikonin may promote BMSCs differentiate into osteoblasts through the Wnt/β-catenin signaling pathway. At the same time, it may also inhibit the formation of osteoclasts mediated by RANK/RANKL/OPG pathway in vitro. Our research explains excellently the mechanism of shikonin alleviating osteoporosis in vitro, which maybe contributing to the exploration of a new way to prevent osteoporosis.
Collapse
Affiliation(s)
- Liang Zhou
- Department of Orthopedics, Lianshui County People’s HospitalHuai’an 223001, Jiangsu, China
| | - Jiaqian Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Jiali Zhao
- Department of Orthopedics, The Affiliated Huai’an Hospital of Xuzhou Medical University and The Second People’s Hospital of Huai’anHuai’an 223002, Jiangsu, China
| | - Feng Kuai
- Department of Geriatrics, The First People’s Hospital of Yancheng, The Forth Affiliated Hospital of Nantong UniversityYancheng 224001, Jiangsu, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| |
Collapse
|
42
|
Chua K, Virshup DM, Odono EG, Chang KTE, Tan NJH, Hue SSS, Sim AYL, Lee VKM. YJ5 as an immunohistochemical marker of osteogenic lineage. Pathology 2020; 53:229-238. [PMID: 33187685 DOI: 10.1016/j.pathol.2020.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
Overexpression of WLS, an upstream protein in the Wnt pathway, has been implicated in several non-osteogenic tumours. This study represents the first attempt at evaluating WLS expression in various bone and soft tissue tumours using YJ5, a monoclonal antibody specific to WLS, with the aim of elucidating its utility in discerning tumours with aberrant Wnt signalling and as a marker of osteogenic lineage in challenging cases. Tumour tissue sections of 144 bone mass lesions and 63 soft tissue mass lesions were immunostained with the YJ5 antibody following standardised protocols. Subsequent assessment of immunoreactivity segregated cases into one of three groups: absent/weak, moderate, or strong YJ5 immunoreactivity. For the bone tumours, strong YJ5 immunoreactivity was seen in almost all osteosarcomas and chondroblastomas, all osteoblastomas and osteoid osteomas. In contrast, all other cartilaginous tumours, chordomas, aneurysmal bone cysts, chondromyxoid fibromas, most fibrous dysplasias and most giant cell tumours exhibited absent/weak YJ5 immunostaining. For the soft tissue tumours, a more heterogeneous pattern of YJ5 immunoreactivity was observed. Because diffuse and strong YJ5 expression is identified in almost all benign and malignant bone tumours with osteoblastic activity, it can be potentially utilised as an immunohistochemical marker to support osteogenic lineage. If interpreted in the appropriate context, this marker is useful in determining whether a malignant bone tumour is an osteosarcoma, particularly in those subtypes with no or minimal osteoid or unusual morphological features. This marker can also complement SATB2 to denote osteogenic lineage.
Collapse
Affiliation(s)
- Kenon Chua
- Department of Orthopaedic Surgery, Singapore General Hospital, Singapore; Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - David M Virshup
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Eugene G Odono
- Department of Pathology, College of Medicine, University of the Philippines, Manila, Philippines
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Nicholas Jin Hong Tan
- Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Susan Swee-Shan Hue
- Department of Pathology, NUH Advance Molecular Pathology Laboratory, Institute of Molecular and Cellular Biology, Singapore
| | - Arthur Yi Loong Sim
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
43
|
Poudel SB, So HS, Sim HJ, Cho JS, Cho ES, Jeon YM, Kook SH, Lee JC. Osteoblastic Wntless deletion differentially regulates the fate and functions of bone marrow-derived stem cells in relation to age. Stem Cells 2020; 39:103-114. [PMID: 33038284 DOI: 10.1002/stem.3289] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/22/2020] [Indexed: 11/10/2022]
Abstract
Although functional association between Wnt signaling and bone homeostasis has been well described through genetic ablation of Wntless (Wls), the mechanisms of how osteoblastic Wls regulates the fate of bone marrow stromal cells (BMSCs) and hematopoietic stem cells (HSCs) in relation to age are not yet understood. Here, we generated Col2.3-Cre;Wlsfl/fl mice that were free from premature lethality and investigated age-related impacts of osteoblastic Wls deficiency on hematopoiesis, BM microenvironment, and maintenance of BMSCs (also known as BM-derived mesenchymal stem/stromal cells) and HSCs. Ablation of osteoblastic Wls deteriorated BM microenvironment and bone mass accrual along with age-independent effects on functions of BMSCs. Osteoblastic Wls deletion impaired HSC repopulation and progeny with skewing toward myeloid lineage cells only at old stage. As proven by hallmarks of stem cell senescence, osteoblastic Wls ablation differentially induced senescence of BMSCs and HSCs in relation to age without alteration in their BM frequency. Our findings support that deletion of Wls in Col2.3-expressing cells induces senescence of BMSCs and impairs BM microenvironment in age-independent manner. Overall, long-term deterioration in BM microenvironment contributes to age-related HSC senescence with impaired progeny and hematopoiesis, which also suggests possible roles of osteoblastic Wls on the maintenance of BM HSCs.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, New York, USA
| | - Han-Sol So
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Hyun-Jaung Sim
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Joon-Seok Cho
- Department of Medicine-Endocrinology, Gerontology and Metabolism, Stanford University School of Medicine, Stanford, California, USA
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Young-Mi Jeon
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| | - Sung-Ho Kook
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences, Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, South Korea.,Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences (BK21 Program) and School of Dentistry, Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
44
|
Zhou L, Huang Y, Zhao J, Yang H, Kuai F. Oridonin promotes osteogenesis through Wnt/β-catenin pathway and inhibits RANKL-induced osteoclastogenesis in vitro. Life Sci 2020; 262:118563. [PMID: 33038376 DOI: 10.1016/j.lfs.2020.118563] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/23/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
AIMS To study the molecular mechanism of oridonin (ORI) on osteoblast differentiation and osteoclast formation in vitro. MAIN METHODS Rat bone marrow mesenchymal stem cells (BMSCs) were treated with different concentrations of ORI in osteogenic medium (OM). CCK-8 assay and were used to detect the effect on BMSCs viability. Alizarin red staining and ALP activity were used to illuminate the effect of ORI on osteogenic differentiation. Expressions of osteogenic differentiation related genes were detected by real-time quantitative PCR (qRT-PCR), and expressions of osteogenic related proteins were detected by Western blot (WB) and immunofluorescence. Similarly, bone marrow mononuclear cells (BMMs) were treated with different concentrations of ORI. CCK-8 assay and Live/Dead staining were used to detect the effect of ORI on BMMs activity. TRAP staining was used to detect its effect on osteoclast differentiation. Expressions of osteoclast-related genes were detected by qRT-PCR, and expressions of osteoclast-related proteins were detected by WB and immunofluorescence. KEY FINDINGS (1) ORI (2 μM) promoted the ALP activity of BMSCs differentiation into osteoblasts and increased the number of calcium nodules. (2) ORI stimulated the expressions of wnt1, β-catenin and Runx2, but with no significantly effect on p-GSK-3β and GSK-3β. (3) ORI promoted the expression of OPG and inhibited the expression of RANKL. (4) ORI directly/indirectly inhibited the osteoclast formation and expressions of osteoclast-related genes TRAP, NFATc1 and c-Fos. SIGNIFICANCE ORI may promote BMSCs differentiate into osteoblasts through the Wnt/β-catenin signaling pathway. At the same time, it may also inhibit the formation of osteoclasts mediated by RANKL.
Collapse
Affiliation(s)
- Liang Zhou
- Department of Orthopedics, Lianshui county People's Hospital, Huai'an, Jiangsu 223001, China
| | - Yingkang Huang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiali Zhao
- Department of Orthopedics, the Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huai'an, Jiangsu 223002, China
| | - Huilin Yang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| | - Feng Kuai
- Department of Geriatrics, the First People's Hospital of Yancheng, the Forth Affiliated Hospital of Nantong University, Yancheng, Jiangsu 224001, China.
| |
Collapse
|
45
|
Matsuoka K, Bakiri L, Wolff LI, Linder M, Mikels-Vigdal A, Patiño-García A, Lecanda F, Hartmann C, Sibilia M, Wagner EF. Wnt signaling and Loxl2 promote aggressive osteosarcoma. Cell Res 2020; 30:885-901. [PMID: 32686768 PMCID: PMC7608146 DOI: 10.1038/s41422-020-0370-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/22/2020] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is the most frequent primary malignant bone tumor in urgent need of better therapies. Using genetically modified mouse models (GEMMs), we demonstrate that Wnt signaling promotes c-Fos-induced OS formation via the actions of the collagen-modifying enzyme Loxl2. c-Fos/AP-1 directly regulates the expression of the Wnt ligands Wnt7b and Wnt9a in OS cells through promoter binding, and Wnt7b and Wnt9a in turn promote Loxl2 expression in murine and human OS cells through the transcription factors Zeb1 and Zeb2. Concordantly, inhibition of Wnt ligand secretion by inactivating the Wnt-less (Wls) gene in osteoblasts in c-Fos GEMMs either early or in a therapeutic setting reduces Loxl2 expression and progression of OS. Wls-deficient osteosarcomas proliferate less, are less mineralized and are enriched in fibroblastic cells surrounded by collagen fibers. Importantly, Loxl2 inhibition using either the pan-Lox inhibitor BAPN or a specific inducible shRNA reduces OS cell proliferation in vitro and decreases tumor growth and lung colonization in murine and human orthotopic OS transplantation models. Finally, OS development is delayed in c-Fos GEMMs treated with BAPN or with specific Loxl2 blocking antibodies. Congruently, a strong correlation between c-FOS, LOXL2 and WNT7B/WNT9A expression is observed in human OS samples, and c-FOS/LOXL2 co-expression correlates with OS aggressiveness and decreased patient survival. Therefore, therapeutic targeting of Wnt and/or Loxl2 should be considered to potentiate the inadequate current treatments for pediatric, recurrent, and metastatic OS.
Collapse
Affiliation(s)
- Kazuhiko Matsuoka
- Laboratory Genes and Disease, Department of Dermatology, Medical University of Vienna (MUV), Vienna, 1090, Austria
- Genes, Development and Disease Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Latifa Bakiri
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna (MUV), Vienna, 1090, Austria
- Genes, Development and Disease Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Lena I Wolff
- Department of Bone and Skeletal Research, Medical Faculty, Institute of Musculoskeletal Medicine, University of Münster, Münster, 48149, Germany
| | - Markus Linder
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna (MUV), Vienna, 1090, Austria
| | | | - Ana Patiño-García
- Navarra Institute for Health Research(IdISNA) and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Department of Pediatrics, University Clinic of Navarra, Pamplona, 31008, Spain
| | - Fernando Lecanda
- Navarra Institute for Health Research(IdISNA) and Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, 31008, Spain
| | - Christine Hartmann
- Department of Bone and Skeletal Research, Medical Faculty, Institute of Musculoskeletal Medicine, University of Münster, Münster, 48149, Germany
| | - Maria Sibilia
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna (MUV), Vienna, 1090, Austria
| | - Erwin F Wagner
- Laboratory Genes and Disease, Department of Dermatology, Medical University of Vienna (MUV), Vienna, 1090, Austria.
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna (MUV), Vienna, 1090, Austria.
| |
Collapse
|
46
|
Bagchi DP, Li Z, Corsa CA, Hardij J, Mori H, Learman BS, Lewis KT, Schill RL, Romanelli SM, MacDougald OA. Wntless regulates lipogenic gene expression in adipocytes and protects against diet-induced metabolic dysfunction. Mol Metab 2020; 39:100992. [PMID: 32325263 PMCID: PMC7264081 DOI: 10.1016/j.molmet.2020.100992] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Obesity is a key risk factor for many secondary chronic illnesses, including type 2 diabetes and cardiovascular disease. Canonical Wnt/β-catenin signaling is established as an important endogenous inhibitor of adipogenesis. This pathway is operative in mature adipocytes; however, its roles in this context remain unclear due to complexities of Wnt signaling and differences in experimental models. In this study, we used novel cultured cell and mouse models to investigate functional roles of Wnts secreted from adipocytes. METHODS We generated adipocyte-specific Wntless (Wls) knockout mice and cultured cell models to investigate molecular and metabolic consequences of disrupting Wnt secretion from mature adipocytes. To characterize Wls-deficient cultured adipocytes, we evaluated the expression of Wnt target and lipogenic genes and the downstream functional effects on carbohydrate and lipid metabolism. We also investigated the impact of adipocyte-specific Wls deletion on adipose tissues and global glucose metabolism in mice fed normal chow or high-fat diets. RESULTS Many aspects of the Wnt signaling apparatus are expressed and operative in mature adipocytes, including the Wnt chaperone Wntless. Deletion of Wntless in cultured adipocytes results in the inhibition of de novo lipogenesis and lipid monounsaturation, likely through repression of Srebf1 (SREBP1c) and Mlxipl (ChREBP) and impaired cleavage of immature SREBP1c into its active form. Adipocyte-specific Wls knockout mice (Wls-/-) have lipogenic gene expression in adipose tissues and isolated adipocytes similar to that of controls when fed a normal chow diet. However, closer investigation reveals that a subset of Wnts and downstream signaling targets are upregulated within stromal-vascular cells of Wls-/- mice, suggesting that adipose tissues defend loss of Wnt secretion from adipocytes. Interestingly, this compensation is lost with long-term high-fat diet challenges. Thus, after six months of a high-fat diet, Wls-/- mice are characterized by decreased adipocyte lipogenic gene expression, reduced visceral adiposity, and improved glucose homeostasis. CONCLUSIONS Taken together, these studies demonstrate that adipocyte-derived Wnts regulate de novo lipogenesis and lipid desaturation and coordinate the expression of lipogenic genes in adipose tissues. In addition, we report that Wnt signaling within adipose tissues is defended, such that a loss of Wnt secretion from adipocytes is sensed and compensated for by neighboring stromal-vascular cells. With chronic overnutrition, this compensatory mechanism is lost, revealing that Wls-/- mice are resistant to diet-induced obesity, adipocyte hypertrophy, and metabolic dysfunction.
Collapse
Affiliation(s)
- Devika P Bagchi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ziru Li
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Callie A Corsa
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Julie Hardij
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Hiroyuki Mori
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Brian S Learman
- Department of Microbiology and Immunology, University of Buffalo, Buffalo, NY, USA.
| | - Kenneth T Lewis
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Rebecca L Schill
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Steven M Romanelli
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA; Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
47
|
Staehlke S, Haack F, Waldner AC, Koczan D, Moerke C, Mueller P, Uhrmacher AM, Nebe JB. ROS Dependent Wnt/β-Catenin Pathway and Its Regulation on Defined Micro-Pillars-A Combined In Vitro and In Silico Study. Cells 2020; 9:E1784. [PMID: 32726949 PMCID: PMC7464713 DOI: 10.3390/cells9081784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
The physico-chemical surface design of implants influences the surrounding cells. Osteoblasts on sharp-edged micro-topographies revealed an impaired cell phenotype, function and Ca2+ mobilization. The influence of edges and ridges on the Wnt/β-catenin pathway in combination with the cells' stress response has not been clear. Therefore, MG-63 osteoblasts were studied on defined titanium-coated micro-pillars (5 × 5 × 5 µm) in vitro and in silico. MG-63s on micro-pillars indicated an activated state of the Wnt/β-catenin pathway. The β-catenin protein accumulated in the cytosol and translocated into the nucleus. Gene profiling indicated an antagonism mechanism of the transcriptional activity of β-catenin due to an increased expression of inhibitors like ICAT (inhibitor of β-catenin and transcription factor-4). Cells on pillars produced a significant reactive oxygen species (ROS) amount after 1 and 24 h. In silico analyses provided a detailed view on how transcriptional activity of Wnt signaling is coordinated in response to the oxidative stress induced by the micro-topography. Based on a coordinated expression of regulatory elements of the Wnt/β-catenin pathway, MG-63s are able to cope with an increased accumulation of β-catenin on micro-pillars and suppress an unintended target gene expression. Further, β-catenin may be diverted into other signaling pathways to support defense mechanisms against ROS.
Collapse
Affiliation(s)
- Susanne Staehlke
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany; (A.-C.W.); (C.M.); (P.M.); (J.B.N.)
| | - Fiete Haack
- Modeling and Simulation Group, Institute for Visual and Analytic Computing, University of Rostock, Albert-Einstein-Str. 22, 18059 Rostock, Germany; (F.H.); (A.M.U.)
| | - Anna-Christin Waldner
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany; (A.-C.W.); (C.M.); (P.M.); (J.B.N.)
| | - Dirk Koczan
- Institute for Immunology, Core Facility for Microarray Analysis, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany;
| | - Caroline Moerke
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany; (A.-C.W.); (C.M.); (P.M.); (J.B.N.)
| | - Petra Mueller
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany; (A.-C.W.); (C.M.); (P.M.); (J.B.N.)
| | - Adelinde M. Uhrmacher
- Modeling and Simulation Group, Institute for Visual and Analytic Computing, University of Rostock, Albert-Einstein-Str. 22, 18059 Rostock, Germany; (F.H.); (A.M.U.)
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
| | - J. Barbara Nebe
- Department of Cell Biology, Rostock University Medical Center, Schillingallee 69, 18057 Rostock, Germany; (A.-C.W.); (C.M.); (P.M.); (J.B.N.)
- Department Science and Technology of Life, Light and Matter, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
| |
Collapse
|
48
|
Du JH, Lin SX, Wu XL, Yang SM, Cao LY, Zheng A, Wu JN, Jiang XQ. The Function of Wnt Ligands on Osteocyte and Bone Remodeling. J Dent Res 2020; 98:930-938. [PMID: 31282847 DOI: 10.1177/0022034519854704] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone homeostasis is continually maintained by the process of bone remodeling throughout life. Recent studies have demonstrated that Wnt signaling pathways play a fundamental role in the process of bone homeostasis and remodeling. Intracellular Wnt signaling cascades are initially triggered by a Wnt ligand-receptor complex formation. In previous studies, the blocking of Wnt ligands from different osteoblastic differentiation stages could cause defective bone development at an early stage. Osteocytes, the most abundant and long-lived type of bone cell, are a crucial orchestrator of bone remodeling. However, the role of Wnt ligands on osteocyte and bone remodeling remains unclear. In our present study, we found that, besides osteoblasts, osteocytes also express multiple Wnt ligands in the bone environment. Then, we used a Dmp1-Cre mouse line, in which there is expression in a subset of osteoblasts but mainly osteocytes, to study the function of Wnt ligands on osteocyte and bone remodeling in vivo. Furthermore, we explored the role of Wnt ligands on osteocytic mineralization ability, as well as the regulatory function of osteocytes on the process of osteoblastic differentiation and osteoclastic migration and maturity in vitro. We concluded that Wnt proteins play an important regulatory role in 1) the process of perilacunar/canalicular remodeling, as mediated by osteocytes, and 2) the balance of osteogenesis and bone resorption at the bone surface, as mediated by osteoblasts and osteoclasts, at least partly through the canonical Wnt/β-catenin signaling pathway and the OPG/RANKL signaling pathway.
Collapse
Affiliation(s)
- J H Du
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - S X Lin
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.,5 Department of Prosthodontics, School and Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - X L Wu
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - S M Yang
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - L Y Cao
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - A Zheng
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - J N Wu
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - X Q Jiang
- 1 Department of Prosthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,2 National Clinical Research Center for Oral Diseases, Shanghai, China.,3 Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.,4 Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
49
|
Turkkahraman H, Yuan X, Salmon B, Chen CH, Brunski JB, Helms JA. Root resorption and ensuing cementum repair by Wnt/β-catenin dependent mechanism. Am J Orthod Dentofacial Orthop 2020; 158:16-27. [DOI: 10.1016/j.ajodo.2019.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 06/01/2019] [Accepted: 06/01/2019] [Indexed: 02/02/2023]
|
50
|
Lim KE, Hoggatt AM, Bullock WA, Horan DJ, Yokota H, Pavalko FM, Robling AG. Pten deletion in Dmp1-expressing cells does not rescue the osteopenic effects of Wnt/β-catenin suppression. J Cell Physiol 2020; 235:9785-9794. [PMID: 32529635 DOI: 10.1002/jcp.29792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 11/06/2022]
Abstract
Skeletal homeostasis is sensitive to perturbations in Wnt signaling. Beyond its role in the bone, Wnt is a major target for pharmaceutical inhibition in a wide range of diseases, most notably cancers. Numerous clinical trials for Wnt-based candidates are currently underway, and Wnt inhibitors will likely soon be approved for clinical use. Given the bone-suppressive effects accompanying Wnt inhibition, there is a need to expose alternate pathways/molecules that can be targeted to counter the deleterious effects of Wnt inhibition on bone properties. Activation of the Pi3k/Akt pathway via Pten deletion is one possible osteoanabolic pathway to exploit. We investigated whether the osteopenic effects of β-catenin deletion from bone cells could be rescued by Pten deletion in the same cells. Mice carrying floxed alleles for Pten and β-catenin were bred to Dmp1-Cre mice to delete Pten alone, β-catenin alone, or both genes from the late-stage osteoblast/osteocyte population. The mice were assessed for bone mass, density, strength, and formation parameters to evaluate the potential rescue effect of Pten deletion in Wnt-impaired mice. Pten deletion resulted in high bone mass and β-catenin deletion resulted in low bone mass. Compound mutants had bone properties similar to β-catenin mutant mice, or surprisingly in some assays, were further compromised beyond β-catenin mutants. Pten inhibition, or one of its downstream nodes, is unlikely to protect against the bone-wasting effects of Wnt/βcat inhibition. Other avenues for preserving bone mass in the presence of Wnt inhibition should be explored to alleviate the skeletal side effects of Wnt inhibitor-based therapies.
Collapse
Affiliation(s)
- Kyung-Eun Lim
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - April M Hoggatt
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Whitney A Bullock
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Daniel J Horan
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Hiroki Yokota
- Indiana Center for Musculoskeletal Health, Indianapolis, Indiana.,Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana
| | - Frederick M Pavalko
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana
| | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana.,Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana.,Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| |
Collapse
|