1
|
Dallas SL, Veno PA, Tiede-Lewis LM, Moore D. Live Cell Imaging of Bone Cell and Organ Cultures. Methods Mol Biol 2025; 2885:479-518. [PMID: 40448777 DOI: 10.1007/978-1-0716-4306-8_25] [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] [Indexed: 06/02/2025]
Abstract
Over the past two decades, there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, which vary considerably in different laboratories, we have focused on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory. We also provide detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- School of Dentistry/Department of Oral and Craniofacial Sciences, University of Missouri, Kansas City, Kansas City, MO, USA.
| | - Patricia A Veno
- School of Dentistry/Department of Oral and Craniofacial Sciences, University of Missouri, Kansas City, Kansas City, MO, USA
| | - LeAnn M Tiede-Lewis
- School of Dentistry/Department of Oral and Craniofacial Sciences, University of Missouri, Kansas City, Kansas City, MO, USA
- Division of STEM, Metropolitan Community College, Kansas City, MO, USA
| | - David Moore
- School of Dentistry/Department of Oral and Craniofacial Sciences, University of Missouri, Kansas City, Kansas City, MO, USA
| |
Collapse
|
2
|
Riquelme MA, Wang X, Acosta FM, Zhang J, Chavez J, Gu S, Zhao P, Xiong W, Zhang N, Li G, Srinivasan S, Ma C, Rao MK, Sun LZ, Zhang N, An Z, Jiang JX. Antibody-activation of connexin hemichannels in bone osteocytes with ATP release suppresses breast cancer and osteosarcoma malignancy. Cell Rep 2024; 43:114377. [PMID: 38889005 PMCID: PMC11380445 DOI: 10.1016/j.celrep.2024.114377] [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: 10/05/2023] [Revised: 05/02/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Bone tissue represents the most frequent site of cancer metastasis. We developed a hemichannel-activating antibody, Cx43-M2. Cx43-M2, directly targeting osteocytes in situ, activates osteocytic hemichannels and elevates extracellular ATP, thereby inhibiting the growth and migration of cultured breast and osteosarcoma cancer cells. Cx43-M2 significantly decreases breast cancer metastasis, osteosarcoma growth, and osteolytic activity, while improving survival rates in mice. The antibody's inhibition of breast cancer and osteosarcoma is dose dependent in both mouse and human cancer metastatic models. Furthermore, Cx43-M2 enhances anti-tumor immunity by increasing the population and activation of tumor-infiltrating immune-promoting effector T lymphocytes, while reducing immune-suppressive regulatory T cells. Our results suggest that the Cx43-M2 antibody, by activating Cx43 hemichannels and facilitating ATP release and purinergic signaling, transforms the cancer microenvironment from a supportive to a suppressive state. Collectively, our study underscores the potential of Cx43-M2 as a therapeutic for treating breast cancer bone metastasis and osteosarcoma.
Collapse
Affiliation(s)
- Manuel A Riquelme
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Xuewei Wang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Francisca M Acosta
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jingruo Zhang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Jeffery Chavez
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Sumin Gu
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Peng Zhao
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Wei Xiong
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ningyan Zhang
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Guo Li
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Saranya Srinivasan
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Chaoyu Ma
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Lu-Zhe Sun
- Cell Systems and Anatomy, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA
| | - Nu Zhang
- Immunology & Molecular Genetics, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA; South Texas Veterans Health Care System, San Antonio, TX 78229, USA
| | - Zhiqiang An
- The Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Jean X Jiang
- Departments of Biochemistry and Structural Biology, Microbiology, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| |
Collapse
|
3
|
Zheng L, Zhou D, Ju F, Liu Z, Yan C, Dong Z, Chen S, Deng L, Chan S, Deng J, Zhang X. Oscillating Fluid Flow Activated Osteocyte Lysate-Based Hydrogel for Regulating Osteoblast/Osteoclast Homeostasis to Enhance Bone Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204592. [PMID: 37017573 PMCID: PMC10214251 DOI: 10.1002/advs.202204592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 02/23/2023] [Indexed: 05/27/2023]
Abstract
As major regulators on bone formation/resorption in response to mechanical stimuli, osteocytes have shown great promise for restoring bone injury. However, due to the unmanageable and unabiding cell functions in unloading or diseased environments, the efficacy of osteogenic induction by osteocytes has been enormously limited. Herein, a facile method of oscillating fluid flow (OFF) loading for cell culture is reported, which enables osteocytes to initiate only osteogenesis and not the osteolysis process. After OFF loading, multiple and sufficient soluble mediators are produced in osteocytes, and the collected osteocyte lysates invariably induce robust osteoblastic differentiation and proliferation while restraining osteoclast generation and activity under unloading or pathological conditions. Mechanistic studies confirm that elevated glycolysis and activation of the ERK1/2 and Wnt/β-catenin pathways are the major contributors to the initiation of osteoinduction functions induced by osteocytes. Moreover, an osteocyte lysate-based hydrogel is designed to establish a stockpile of "active osteocytes" to sustainably deliver bioactive proteins, resulting in accelerated healing through regulation of endogenous osteoblast/osteoclast homeostasis.
Collapse
Affiliation(s)
- Liyuan Zheng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Disheng Zhou
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Feier Ju
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Zixuan Liu
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Chenzhi Yan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Zhaoxia Dong
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Shuna Chen
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Lizhi Deng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Szehoi Chan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| | - Junjie Deng
- Joint Centre of Translational MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiang325000P. R. China
- Joint Centre of Translational MedicineWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000P. R. China
- Zhejiang Engineering Research Center for Tissue Repair MaterialsWenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325000P. R. China
| | - Xingding Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory DiseasesSchool of Medicine, Shenzhen Campus of Sun Yat‐Sen UniversitySun Yat‐sen UniversityShenzhen518106P. R. China
| |
Collapse
|
4
|
Nepal AK, van Essen HW, Reijnders CMA, Lips P, Bravenboer N. Mechanical loading modulates phosphate related genes in rat bone. PLoS One 2023; 18:e0282678. [PMID: 36881582 PMCID: PMC9990935 DOI: 10.1371/journal.pone.0282678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Mechanical loading determines bone mass and bone structure, which involves many biochemical signal molecules. Of these molecules, Mepe and Fgf23 are involved in bone mineralization and phosphate homeostasis. Thus, we aimed to explore whether mechanical loading of bone affects factors of phosphate homeostasis. We studied the effect of mechanical loading of bone on the expression of Fgf23, Mepe, Dmp1, Phex, Cyp27b1, and Vdr. Twelve-week old female rats received a 4-point bending load on the right tibia, whereas control rats were not loaded. RT-qPCR was performed on tibia mRNA at 4, 5, 6, 7 or 8 hours after mechanical loading for detection of Mepe, Dmp1, Fgf23, Phex, Cyp27b1, and Vdr. Immunohistochemistry was performed to visualise FGF23 protein in tibiae. Serum FGF23, phosphate and calcium levels were measured in all rats. Four-point bending resulted in a reduction of tibia Fgf23 gene expression by 64% (p = 0.002) and a reduction of serum FGF23 by 30% (p<0.001), six hours after loading. Eight hours after loading, Dmp1 and Mepe gene expression increased by 151% (p = 0.007) and 100% (p = 0.007). Mechanical loading did not change Phex, Cyp27b1, and Vdr gene expression at any time-point. We conclude that mechanical loading appears to provoke both a paracrine as well as an endocrine response in bone by modulating factors that regulate bone mineralization and phosphate homeostasis.
Collapse
Affiliation(s)
- Ashwini Kumar Nepal
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Hubertus W. van Essen
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Christianne M. A. Reijnders
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paul Lips
- Department of Internal Medicine, Endocrine Section, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
5
|
Zhao D, Hua R, Riquelme MA, Cheng H, Guda T, Xu H, Gu S, Jiang JX. Osteocytes regulate bone anabolic response to mechanical loading in male mice via activation of integrin α5. Bone Res 2022; 10:49. [PMID: 35851577 PMCID: PMC9293884 DOI: 10.1038/s41413-022-00222-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Physical mechanical stimulation can maintain and even increase bone mass. Here, we report an important role of osteocytic integrin α5 in regulating the anabolic response of bone to mechanical loading using an Itga5 conditional gene knockout (cKO) mouse model. Integrin α5 gene deletion increased apoptotic osteocytes and reduced cortical anabolic responses to tibial compression including decreased endosteal osteoblasts and bone formation, and increased endosteal osteoclasts and bone resorption, contributing to the decreased bone area fraction and biomechanical properties, leading to an enlarged bone marrow area in cKO mice. Similar disruption of anabolic responses to mechanical loading was also detected in cKO trabecular bone. Moreover, integrin α5 deficiency impeded load-induced Cx43 hemichannel opening, and production and release of PGE2, an anabolic factor, resulting in attenuated effects of the loading on catabolic sclerostin (SOST) reduction and anabolic β-catenin increase. Together, this study shows an indispensable role of integrin α5 in osteocytes in the anabolic action of mechanical loading on skeletal tissue through activation of hemichannels and PGE2-evoked gene expression. Integrin α5 could act as a potential new therapeutic target for bone loss, especially in the elderly population with impeded mechanical sensitivity.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Hongyun Cheng
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas, San Antonio, TX, USA
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
6
|
Zhang J, Griesbach J, Ganeyev M, Zehnder AK, Zeng P, Schädli GN, Leeuw AD, Lai Y, Rubert M, Mueller R. Long-term mechanical loading is required for the formation of 3D bioprinted functional osteocyte bone organoids. Biofabrication 2022; 14. [PMID: 35617929 DOI: 10.1088/1758-5090/ac73b9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/26/2022] [Indexed: 11/11/2022]
Abstract
Mechanical loading has been shown to influence various osteogenic responses of bone-derived cells and bone formation in vivo. However, the influence of mechanical stimulation on the formation of bone organoid in vitro is not clearly understood. Here, 3D bioprinted human mesenchymal stem cells (hMSCs)-laden graphene oxide composite scaffolds were cultured in a novel cyclic-loading bioreactors for up to 56 days. Our results showed that mechanical loading from day 1 (ML01) significantly increased organoid mineral density, organoid stiffness, and osteoblast differentiation compared with non-loading and mechanical loading from day 21. Importantly, ML01 stimulated collagen I maturation, osteocyte differentiation, lacunar-canalicular network formation and YAP expression on day 56. These finding are the first to reveal that long-term mechanical loading is required for the formation of 3D bioprinted functional osteocyte bone organoids. Such 3D bone organoids may serve as a human-specific alternative to animal testing for the study of bone pathophysiology and drug screening.
Collapse
Affiliation(s)
- Jianhua Zhang
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Julia Griesbach
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8093, SWITZERLAND
| | - Marsel Ganeyev
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Anna-Katharina Zehnder
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Peng Zeng
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Gian Nutal Schädli
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Anke de Leeuw
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8092, SWITZERLAND
| | - Yuxiao Lai
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, Shenzhen, 518055, CHINA
| | - Marina Rubert
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8093, SWITZERLAND
| | - Ralph Mueller
- ETH Zurich Department of Health Sciences and Technology, Leopold-Ruzicka-Weg 4, Zurich, Zürich, 8093, SWITZERLAND
| |
Collapse
|
7
|
Wang K, Ren Y, Lin S, Jing Y, Ma C, Wang J, Yuan XB, Han X, Zhao H, Wang Z, Zheng M, Xiao Y, Chen L, Olsen BR, Feng JQ. Osteocytes but not osteoblasts directly build mineralized bone structures. Int J Biol Sci 2021; 17:2430-2448. [PMID: 34326685 PMCID: PMC8315029 DOI: 10.7150/ijbs.61012] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/21/2021] [Indexed: 02/05/2023] Open
Abstract
Bone-forming osteoblasts have been a cornerstone of bone biology for more than a century. Most research toward bone biology and bone diseases center on osteoblasts. Overlooked are the 90% of bone cells, called osteocytes. This study aims to test the hypothesis that osteocytes but not osteoblasts directly build mineralized bone structures, and that defects in osteocytes lead to the onset of hypophosphatemia rickets. The hypothesis was tested by developing and modifying multiple imaging techniques, including both in vivo and in vitro models plus two types of hypophosphatemia rickets models (Dmp1-null and Hyp, Phex mutation mice), and Dmp1-Cre induced high level of β-catenin models. Our key findings were that osteocytes (not osteoblasts) build bone similar to the construction of a high-rise building, with a wire mesh frame (i.e., osteocyte dendrites) and cement (mineral matrices secreted from osteocytes), which is a lengthy and slow process whose mineralization direction is from the inside toward the outside. When osteoblasts fail to differentiate into osteocytes but remain highly active in Dmp-1-null or Hyp mice, aberrant and poor bone mineralization occurs, caused by a sharp increase in Wnt-β-catenin signaling. Further, the constitutive expression of β-catenin in osteocytes recaptures a similar osteomalacia phenotype as shown in Dmp1 null or Hyp mice. Thus, we conclude that osteocytes directly build bone, and osteoblasts with a short life span serve as a precursor to osteocytes, which challenges the existing dogma.
Collapse
Affiliation(s)
- Ke Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Yinshi Ren
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219 USA
| | - Shuxian Lin
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai, 200092, China
| | - Yan Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Chi Ma
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, TX 75219 USA
| | - Jun Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - X Baozhi Yuan
- Angitia Biopharmaceuticals, Guangzhou, 510000, China
| | - Xianglong Han
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hu Zhao
- Department of Restorative Dentistry, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Zheng Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Minghao Zheng
- Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Perth, 6009, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, 4059, Australia
| | - Lin Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Bjorn Reino Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| |
Collapse
|
8
|
Riquelme MA, Gu S, Hua R, Jiang JX. Mechanotransduction via the coordinated actions of integrins, PI3K signaling and Connexin hemichannels. Bone Res 2021; 9:8. [PMID: 33531460 PMCID: PMC7854719 DOI: 10.1038/s41413-020-00126-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/25/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Mechanical loading opens connexin 43 (Cx43) hemichannels (HCs), leading to the release of bone anabolic molecules, such as prostaglandins, from mechanosensitive osteocytes, which is essential for bone formation and remodeling. However, the mechanotransduction mechanism that activates HCs remains elusive. Here, we report a unique pathway by which mechanical signals are effectively transferred between integrin molecules located in different regions of the cell, resulting in HC activation. Both integrin α5 and αV were activated upon mechanical stimulation via either fluid dropping or flow shear stress (FSS). Inhibition of integrin αV activation or ablation of integrin α5 prevented HC opening on the cell body when dendrites were mechanically stimulated, suggesting mechanical transmission from the dendritic integrin αV to α5 in the cell body during HC activation. In addition, HC function was compromised in vivo, as determined by utilizing an antibody blocking αV activation and α5-deficient osteocyte-specific knockout mice. Furthermore, inhibition of integrin αV activation, but not that of α5, attenuated activation of the phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) signaling pathway upon mechanical loading, and the inhibition of PI3K/AKT activation blocked integrin α5 activation and HC opening. Moreover, HC opening was blocked only by an anti-integrin αV antibody at low but not high FSS levels, suggesting that dendritic αV is a more sensitive mechanosensor than α5 for activating HCs. Together, these results reveal a new molecular mechanism of mechanotransduction involving the coordinated actions of integrins and PI3K/AKT in osteocytic dendritic processes and cell bodies that leads to HC opening and the release of key bone anabolic factors.
Collapse
Affiliation(s)
- Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, 78229-3900, USA.
| |
Collapse
|
9
|
Abstract
The aim of this review was to compile a list of tools currently available to study bone cells and in particular osteocytes. As the interest (and importance) in osteocyte biology has greatly expanded over the past decade, new tools and techniques have become available to study these elusive cells, RECENT FINDINGS: Osteocytes are the main orchestrators of bone remodeling. They control both osteoblasts and osteoclast activities via cell-to cell communication or through secreted factors. Osteocytes are also the mechanosensors of the bone and they orchestrate skeletal adaptation to loads. Recent discoveries have greatly expanded our knowledge and understanding of these cells and new models are now available to further uncover the functions of osteocytes. Novel osteocytic cell lines, primary cultures, and 3D scaffolds are now available to investigators to further unravel the functions and roles of these cells.
Collapse
Affiliation(s)
- Paola Divieti Pajevic
- Translational Dental Medicine, Boston University Henry M. Goldman School of Dental Medicine, 700 Albany Street, W201E, Boston, MA, 02118, USA.
| |
Collapse
|
10
|
Sato T, Verma S, Andrade CDC, Omeara M, Campbell N, Wang JS, Cetinbas M, Lang A, Ausk BJ, Brooks DJ, Sadreyev RI, Kronenberg HM, Lagares D, Uda Y, Pajevic PD, Bouxsein ML, Gross TS, Wein MN. A FAK/HDAC5 signaling axis controls osteocyte mechanotransduction. Nat Commun 2020; 11:3282. [PMID: 32612176 PMCID: PMC7329900 DOI: 10.1038/s41467-020-17099-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 06/11/2020] [Indexed: 11/13/2022] Open
Abstract
Osteocytes, cells ensconced within mineralized bone matrix, are the primary skeletal mechanosensors. Osteocytes sense mechanical cues by changes in fluid flow shear stress (FFSS) across their dendritic projections. Loading-induced reductions of osteocytic Sclerostin (encoded by Sost) expression stimulates new bone formation. However, the molecular steps linking mechanotransduction and Sost suppression remain unknown. Here, we report that class IIa histone deacetylases (HDAC4 and HDAC5) are required for loading-induced Sost suppression and bone formation. FFSS signaling drives class IIa HDAC nuclear translocation through a signaling pathway involving direct HDAC5 tyrosine 642 phosphorylation by focal adhesion kinase (FAK), a HDAC5 post-translational modification that controls its subcellular localization. Osteocyte cell adhesion supports FAK tyrosine phosphorylation, and FFSS triggers FAK dephosphorylation. Pharmacologic FAK catalytic inhibition reduces Sost mRNA expression in vitro and in vivo. These studies demonstrate a role for HDAC5 as a transducer of matrix-derived cues to regulate cell type-specific gene expression.
Collapse
Affiliation(s)
- Tadatoshi Sato
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Shiv Verma
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | | | - Maureen Omeara
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Nia Campbell
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Jialiang S. Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Murat Cetinbas
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Audrey Lang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Brandon J. Ausk
- 0000000122986657grid.34477.33Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA USA
| | - Daniel J. Brooks
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA ,Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology and Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Henry M. Kronenberg
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Fibrosis Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Yuhei Uda
- 0000 0004 1936 7558grid.189504.1Translational Dental Medicine, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA USA
| | - Paola Divieti Pajevic
- 0000 0004 1936 7558grid.189504.1Translational Dental Medicine, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA USA
| | - Mary L. Bouxsein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA ,Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Ted S. Gross
- 0000000122986657grid.34477.33Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA USA
| | - Marc N. Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA ,grid.66859.34Broad Institute of Harvard and MIT, Cambridge, MA USA
| |
Collapse
|
11
|
Abstract
Over the past two decades there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point, but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, which may vary in different laboratories, we focus on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory. We also provide detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA.
| | - Patricia A Veno
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA
| | - LeAnn M Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA
| |
Collapse
|
12
|
Maurel DB, Matsumoto T, Vallejo JA, Johnson ML, Dallas SL, Kitase Y, Brotto M, Wacker MJ, Harris MA, Harris SE, Bonewald LF. Characterization of a novel murine Sost ER T2 Cre model targeting osteocytes. Bone Res 2019; 7:6. [PMID: 30820362 PMCID: PMC6382861 DOI: 10.1038/s41413-018-0037-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/25/2018] [Accepted: 10/19/2018] [Indexed: 12/28/2022] Open
Abstract
Transgenic mice are widely used to delete or overexpress genes in a cell specific manner to advance knowledge of bone biology, function and disease. While numerous Cre models exist to target gene recombination in osteoblasts and osteoclasts, few target osteocytes specifically, particularly mature osteocytes. Our goal was to create a spatial and temporal conditional Cre model using tamoxifen to induce Cre activity in mature osteocytes using a Bac construct containing the 5' and 3' regions of the Sost gene (Sost ERT2 Cre). Four founder lines were crossed with the Ai9 Cre reporter mice. One founder line showed high and specific activity in mature osteocytes. Bones and organs were imaged and fluorescent signal quantitated. While no activity was observed in 2 day old pups, by 2 months of age some osteocytes were positive as osteocyte Cre activity became spontaneous or 'leaky' with age. The percentage of positive osteocytes increased following tamoxifen injection, especially in males, with 43% to 95% positive cells compared to 19% to 32% in females. No signal was observed in any bone surface cell, bone marrow, nor in muscle with or without tamoxifen injection. No spontaneous signal was observed in any other organ. However, with tamoxifen injection, a few positive cells were observed in kidney, eye, lung, heart and brain. All other organs, 28 in total, were negative with tamoxifen injection. However, with age, a muscle phenotype was apparent in the Sost-ERT2 Cre mice. Therefore, although this mouse model may be useful for targeting gene deletion or expression to mature osteocytes, the muscle phenotype may restrict the use of this model to specific applications and should be considered when interpreting data.
Collapse
Affiliation(s)
- Delphine B Maurel
- 1Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City School of Dentistry, Kansas City, MO USA.,7Present Address: Pharmaceutical Sciences Department, Universite de Bordeaux, Bio-Tis, INSERM Unité 1026 BioTis, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Tsutomu Matsumoto
- 2Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Julian A Vallejo
- 1Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City School of Dentistry, Kansas City, MO USA
| | - Mark L Johnson
- 1Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City School of Dentistry, Kansas City, MO USA
| | - Sarah L Dallas
- 1Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City School of Dentistry, Kansas City, MO USA
| | - Yukiko Kitase
- 2Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Marco Brotto
- 3Bone-Muscle Collaborative Sciences, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, USA
| | - Michael J Wacker
- 4Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO USA
| | - Marie A Harris
- 5University of Texas Health Science Center, San Antonio, TX USA
| | | | - Lynda F Bonewald
- 1Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City School of Dentistry, Kansas City, MO USA.,2Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN USA.,6Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN USA
| |
Collapse
|
13
|
Clinkenbeard EL, Noonan ML, Thomas JC, Ni P, Hum JM, Aref M, Swallow EA, Moe SM, Allen MR, White KE. Increased FGF23 protects against detrimental cardio-renal consequences during elevated blood phosphate in CKD. JCI Insight 2019; 4:123817. [PMID: 30830862 DOI: 10.1172/jci.insight.123817] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/14/2019] [Indexed: 12/23/2022] Open
Abstract
The phosphaturic hormone FGF23 is elevated in chronic kidney disease (CKD). The risk of premature death is substantially higher in the CKD patient population, with cardiovascular disease (CVD) as the leading mortality cause at all stages of CKD. Elevated FGF23 in CKD has been associated with increased odds for all-cause mortality; however, whether FGF23 is associated with positive adaptation in CKD is unknown. To test the role of FGF23 in CKD phenotypes, a late osteoblast/osteocyte conditional flox-Fgf23 mouse (Fgf23fl/fl/Dmp1-Cre+/-) was placed on an adenine-containing diet to induce CKD. Serum analysis showed casein-fed Cre+ mice had significantly higher serum phosphate and blood urea nitrogen (BUN) versus casein diet and Cre- genotype controls. Adenine significantly induced serum intact FGF23 in the Cre- mice over casein-fed mice, whereas Cre+ mice on adenine had 90% reduction in serum intact FGF23 and C-terminal FGF23 as well as bone Fgf23 mRNA. Parathyroid hormone was significantly elevated in mice fed adenine diet regardless of genotype, which significantly enhanced midshaft cortical porosity. Echocardiographs of the adenine-fed Cre+ hearts revealed profound aortic calcification and cardiac hypertrophy versus diet and genotype controls. Thus, these studies demonstrate that increased bone FGF23, although associated with poor outcomes in CKD, is necessary to protect against the cardio-renal consequences of elevated tissue phosphate.
Collapse
Affiliation(s)
| | | | | | - Pu Ni
- Department of Medical and Molecular Genetics
| | - Julia M Hum
- Department of Medical and Molecular Genetics
| | | | | | - Sharon M Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Kenneth E White
- Department of Medical and Molecular Genetics.,Department of Anatomy and Cell Biology, and
| |
Collapse
|
14
|
Buskermolen J, van der Meijden K, Furrer R, Mons DJ, van Essen HW, Heijboer AC, Lips P, Jaspers RT, Bravenboer N. Effects of different training modalities on phosphate homeostasis and local vitamin D metabolism in rat bone. PeerJ 2019; 7:e6184. [PMID: 30697476 PMCID: PMC6348094 DOI: 10.7717/peerj.6184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/29/2018] [Indexed: 01/31/2023] Open
Abstract
Objectives Mechanical loading may be an important factor in the regulation of bone derived hormones involved in phosphate homeostasis. This study investigated the effects of peak power and endurance training on expression levels of fibroblast growth factor 23 (FGF23) and 1α-hydroxylase (CYP27b1) in bone. Methods Thirty-eight rats were assigned to six weeks of training in four groups: peak power (PT), endurance (ET), PT followed by ET (PET) or no training (control). In cortical bone, FGF23 was quantified using immunohistochemistry. mRNA expression levels of proteins involved in phosphate and vitamin D homeostasis were quantified in cortical bone and kidney. C-terminal FGF23, 25-hydroxyvitamin D3, parathyroid hormone (PTH), calcium and phosphate concentrations were measured in plasma or serum. Results Neither FGF23 mRNA and protein expression levels in cortical bone nor FGF23 plasma concentrations differed between the groups. In cortical bone, mRNA expression levels of sclerostin (SOST), dental matrix protein 1 (DMP1), phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX) and matrix extracellular phosphoglycoprotein (MEPE) were lower after PT compared to ET and PET. Expression levels of CYP27b1 and vitamin D receptor (VDR) in tibial bone were decreased after PT compared to ET. In kidney, no differences between groups were observed for mRNA expression levels of CYP27b1, 24-hydroxylase (CYP24), VDR, NaPi-IIa cotransporter (NPT2a) and NaPi-IIc cotransporter (NPT2c). Serum PTH concentrations were higher after PT compared to controls. Conclusion After six weeks, none of the training modalities induced changes in FGF23 expression levels. However, PT might have caused changes in local phosphate regulation within bone compared to ET and PET. CYP27b1 and VDR expression in bone was reduced after PT compared to ET, suggesting high intensity peak power training in this rat model is associated with decreased vitamin D signalling in bone.
Collapse
Affiliation(s)
- Joost Buskermolen
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Karen van der Meijden
- Department of Internal Medicine/Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Regula Furrer
- Laboratory for Myology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Dirk-Jan Mons
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Huib W van Essen
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Annemieke C Heijboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul Lips
- Department of Internal Medicine/Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, VU University Amsterdam, Amsterdam, The Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Huang J, Romero-Suarez S, Lara N, Mo C, Kaja S, Brotto L, Dallas SL, Johnson ML, Jähn K, Bonewald LF, Brotto M. Crosstalk between MLO-Y4 osteocytes and C2C12 muscle cells is mediated by the Wnt/β-catenin pathway. JBMR Plus 2017; 1:86-100. [PMID: 29104955 DOI: 10.1002/jbm4.10015] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We examined the effects of osteocyte secreted factors on myogenesis and muscle function. MLO-Y4 osteocyte-like cell conditioned media (CM) (10%) increased ex vivo soleus muscle contractile force by ~25%. MLO-Y4 and primary osteocyte CM (1-10%) stimulated myogenic differentiation of C2C12 myoblasts, but 10% osteoblast CMs did not enhance C2C12 cell differentiation. Since WNT3a and WNT1 are secreted by osteocytes, and the expression level of Wnt3a is increased in MLO-Y4 cells by fluid flow shear stress, both were compared, showing WNT3a more potent than WNT1 in inducing myogenesis. Treatment of C2C12 myoblasts with WNT3a at concentrations as low as 0.5ng/mL mirrored the effects of both primary osteocyte and MLO-Y4 CM by inducing nuclear translocation of β-catenin with myogenic differentiation, suggesting that Wnts might be potential factors secreted by osteocytes that signal to muscle cells. Knocking down Wnt3a in MLO-Y4 osteocytes inhibited the effect of CM on C2C12 myogenic differentiation. Sclerostin (100ng/mL) inhibited both the effects of MLO-Y4 CM and WNT3a on C2C12 cell differentiation. RT-PCR array results supported the activation of the Wnt/β-catenin pathway by MLO-Y4 CM and WNT3a. These results were confirmed by qPCR showing up-regulation of myogenic markers and two Wnt/β-catenin downstream genes, Numb and Flh1. We postulated that MLO-Y4 CM/WNT3a could modulate intracellular calcium homeostasis as the trigger mechanism for the enhanced myogenesis and contractile force. MLO-Y4 CM and WNT3a increased caffeine-induced Ca2+ release from the sarcoplasmic reticulum (SR) of C2C12 myotubes and the expression of genes directly associated with intracellular Ca2+ signaling and homeostasis. Together, these data show that in vitro and ex vivo, osteocytes can stimulate myogenesis and enhance muscle contractile function and suggest that Wnts could be mediators of bone to muscle signaling, likely via modulation of intracellular Ca2+ signaling and the Wnt/β-Catenin pathway.
Collapse
Affiliation(s)
- Jian Huang
- Muscle Biology Research Group-MUBIG, School sof Nursing & Health Studies, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Sandra Romero-Suarez
- Muscle Biology Research Group-MUBIG, School sof Nursing & Health Studies, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Nuria Lara
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, MO, 64108, USA
| | - Chenglin Mo
- Muscle Biology Research Group-MUBIG, School sof Nursing & Health Studies, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Simon Kaja
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri-Kansas City, 2411 Holmes St., Kansas City, MO, 64108, USA
| | - Leticia Brotto
- Muscle Biology Research Group-MUBIG, School sof Nursing & Health Studies, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, MO, 64108, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, MO, 64108, USA
| | - Katharina Jähn
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, MO, 64108, USA
| | - Lynda F Bonewald
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, MO, 64108, USA
| | - Marco Brotto
- Muscle Biology Research Group-MUBIG, School sof Nursing & Health Studies, University of Missouri- Kansas City, 2464 Charlotte Street, Kansas City, MO 64108, USA
| |
Collapse
|
16
|
Tamaki H, Yotani K, Ogita F, Hayao K, Nakagawa K, Sugawara K, Kirimoto H, Onishi H, Kasuga N, Yamamoto N. Electrical Stimulation of Denervated Rat Skeletal Muscle Ameliorates Bone Fragility and Muscle Loss in Early-Stage Disuse Musculoskeletal Atrophy. Calcif Tissue Int 2017; 100:420-430. [PMID: 28213864 DOI: 10.1007/s00223-017-0250-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
Abstract
We tested whether daily muscle electrical stimulation (ES) can ameliorate the decrease in cortical bone strength as well as muscle and bone geometric and material properties in the early stages of disuse musculoskeletal atrophy. 7-week-old male F344 rats were randomly divided into three groups: age-matched control group (Cont); a sciatic denervation group (DN); and a DN + direct electrical stimulation group (DN + ES). Denervated tibialis anterior (TA) muscle in the DN + ES group received ES with 16 mA at 10 Hz for 30 min/day, 6 days/week. Micro CT, the three-point bending test, and immunohistochemistry were used to characterize cortical bone mechanical, structural, and material properties of tibiae. TA muscle in the DN + ES group showed significant improvement in muscle mass and myofiber cross-sectional area relative to the DN group. Maximal load and stiffness of tibiae, bone mineral density estimated by micro CT, and immunoreactivity of DMP1 in the cortical bone tissue were also significantly greater in the DN + ES group than in the DN group. These results suggest that daily ES-induced muscle contraction treatment reduced the decrease in muscle mass and cortical bone strength in early-stage disuse musculoskeletal atrophy and is associated with a beneficial effect on material properties such as mineralization of cortical bone tissue.
Collapse
Affiliation(s)
- Hiroyuki Tamaki
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan.
| | - Kengo Yotani
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, Kanoya, Kagoshima, 891-2393, Japan
| | - Futoshi Ogita
- National Institute of Fitness and Sports in Kanoya, 1 Shiromizu, Kanoya, Kagoshima, 891-2393, Japan
| | - Keishi Hayao
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan
| | - Kouki Nakagawa
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan
| | - Kazuhiro Sugawara
- Tohoku Fukushi University, 1‑8‑1 Kunimi, Aoba‑ku, Sendai, Miyagi, 981‑8522, Japan
| | - Hikari Kirimoto
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan
| | - Hideaki Onishi
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan
| | - Norikatsu Kasuga
- Aichi University of Education, 1 Hirosawa, Igaya, Kariya, Aichi, 448-8542, Japan
| | - Noriaki Yamamoto
- Institute for Human Movement and Medical Sciences, Niigata University of Health and Welfare, 1398 Shimami, Kita-ku, Niigata, 950-3198, Japan
- Niigata Rehabilitation Hospital, 761 Kisaki, Kita-ku, Niigata, 950-3304, Japan
| |
Collapse
|
17
|
Abstract
When normal physiologic functions go awry, disorders and disease occur. This is universal; even for the osteocyte, a cell embedded within the mineralized matrix of bone. It was once thought that this cell was simply a placeholder in bone. Within the last decade, the number of studies of osteocytes has increased dramatically, leading to the discovery of novel functions of these cells. With the discovery of novel physiologic functions came the discoveries of how these cells can also be responsible for not only bone diseases and disorders, but also those of the kidney, heart, and potentially muscle.
Collapse
Affiliation(s)
- Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, VanNuys Medical Science Building, MS 5055, 635 Barnhill Drive, Indianapolis, IN 46202, USA; Department of Anatomy and Cell Biology, VanNuys Medical Science Building, MS 5035, Indianapolis, IN 46202, USA; Department of Orthopaedic Surgery, 1120 West Michigan Street, Suite 600, Indianapolis, IN 46202, USA.
| |
Collapse
|
18
|
Suswillo RFL, Javaheri B, Rawlinson SCF, Dowthwaite GP, Lanyon LE, Pitsillides AA. Strain uses gap junctions to reverse stimulation of osteoblast proliferation by osteocytes. Cell Biochem Funct 2017; 35:56-65. [PMID: 28083967 PMCID: PMC5299599 DOI: 10.1002/cbf.3245] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022]
Abstract
Identifying mechanisms by which cells of the osteoblastic lineage communicate in vivo is complicated by the mineralised matrix that encases osteocytes, and thus, vital mechanoadaptive processes used to achieve load-bearing integrity remain unresolved. We have used the coculture of immunomagnetically purified osteocytes and primary osteoblasts from both embryonic chick long bone and calvariae to examine these mechanisms. We exploited the fact that purified osteocytes are postmitotic to examine both their effect on proliferation of primary osteoblasts and the role of gap junctions in such communication. We found that chick long bone osteocytes significantly increased basal proliferation of primary osteoblasts derived from an identical source (tibiotarsi). Using a gap junction inhibitor, 18β-glycyrrhetinic acid, we also demonstrated that this osteocyte-related increase in osteoblast proliferation was not reliant on functional gap junctions. In contrast, osteocytes purified from calvarial bone failed to modify basal proliferation of primary osteoblast, but long bone osteocytes preserved their proproliferative action upon calvarial-derived primary osteoblasts. We also showed that coincubated purified osteocytes exerted a marked inhibitory action on mechanical strain-related increases in proliferation of primary osteoblasts and that this action was abrogated in the presence of a gap junction inhibitor. These data reveal regulatory differences between purified osteocytes derived from functionally distinct bones and provide evidence for 2 mechanisms by which purified osteocytes communicate with primary osteoblasts to coordinate their activity.
Collapse
Affiliation(s)
| | - Behzad Javaheri
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Simon C F Rawlinson
- Institute of Dentistry, Barts & The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gary P Dowthwaite
- Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Lance E Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | |
Collapse
|
19
|
Histological evidence that metformin reverses the adverse effects of diabetes on orthodontic tooth movement in rats. J Mol Histol 2016; 48:73-81. [DOI: 10.1007/s10735-016-9707-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
|
20
|
Qin C, D’Souza R, Feng J. Dentin Matrix Protein 1 (DMP1): New and Important Roles for Biomineralization and Phosphate Homeostasis. J Dent Res 2016; 86:1134-41. [DOI: 10.1177/154405910708601202] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Previously, non-collagenous matrix proteins, such as DMP1, were viewed with little biological interest. The last decade of research has increased our understanding of DMP1, as it is now widely recognized that this protein is expressed in non-mineralized tissues, as well as in cancerous lesions. Protein chemistry studies have shown that the full length of DMP1, as a precursor, is cleaved into two distinct forms: the C-terminal and N-terminal fragments. Functional studies have demonstrated that DMP1 is essential in the maturation of odontoblasts and osteoblasts, as well as in mineralization via local and systemic mechanisms. The identification of DMP1 mutations in humans has led to the discovery of a novel disease: autosomal-recessive hypophosphatemic rickets. Furthermore, the regulation of phosphate homeostasis by DMP1 through FGF23, a newly identified hormone that is released from bone and targeted in the kidneys, sets a new direction for research that associates biomineralization with phosphate regulation.
Collapse
Affiliation(s)
- C. Qin
- Department of Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA
| | - R. D’Souza
- Department of Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA
| | - J.Q. Feng
- Department of Biomedical Sciences, Texas A&M Health Science Center, Baylor College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, USA
| |
Collapse
|
21
|
Roeder E, Matthews BG, Kalajzic I. Visual reporters for study of the osteoblast lineage. Bone 2016; 92:189-195. [PMID: 27616604 PMCID: PMC5056847 DOI: 10.1016/j.bone.2016.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/24/2022]
Abstract
Advancing our understanding of osteoblast biology and differentiation is critical to elucidate the pathological mechanisms responsible for skeletal diseases such as osteoporosis. Histology and histomorphometry, the classical methods to study osteoblast biology, identify osteoblasts based on their location and morphology and ability to mineralize matrix, but do not clearly define their stage of differentiation. Introduction of visual transgenes into the cells of osteoblast lineage has revolutionized the field and resulted in a paradigm shift that allowed for specific identification and isolation of subpopulations within the osteoblast lineage. Knowledge acquired from the studies based on GFP transgenes has allowed for more precise interpretation of studies analyzing targeted overexpression or deletion of genes in the osteoblast lineage. Here, we provide a condensed overview of the currently available promoter-fluorescent reporter transgenic mice that have been generated and evaluated to varying extents. We cover different stages of the lineage as transgenes have been utilized to identify osteoprogenitors, pre-osteoblasts, osteoblasts, or osteocytes. We show that each of these promoters present with advantages and disadvantages. The studies based on the use of these reporter mice have improved our understanding of bone biology. They constitute attractive models to target osteoblasts and help to understand their cell biology.
Collapse
Affiliation(s)
- Emilie Roeder
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Brya G Matthews
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Pathophysiology, University of Osijek, Osijek, Croatia.
| |
Collapse
|
22
|
Murshid SA. The role of osteocytes during experimental orthodontic tooth movement: A review. Arch Oral Biol 2016; 73:25-33. [PMID: 27653146 DOI: 10.1016/j.archoralbio.2016.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 09/05/2016] [Accepted: 09/06/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To explore the types of orthodontic force-induced mechanical stimuli that regulate osteocyte function. DESIGN In orthodontics, a tooth can be moved through the alveolar bone when an appropriate orthodontic force is applied. These mechanical loads stimulate cells within the bone tissue around the tooth. These cellular responses lead to bone resorption on the side of the tooth where the pressure has been applied and bone deposition on the side of the tooth experiencing tension. Recently, osteocytes were identified to function as mechano-sensory cells in bone tissue that direct bone resorption and bone formation. Based on recent literature, the proposed function of osteocytes during orthodontic tooth movement is explored with better understanding. RESULTS Several stimuli regulating osteocyte function have been highlighted, and their potential roles in events initiating osteocyte sensing of orthodontic force have been explored in detail. The most popular hypotheses for osteocyte response include stress-induced bone matrix deformation/microcrack formation and fluid-flow shear stress. CONCLUSIONS Understanding osteocyte function under mechanical stress may have profound implications in future orthodontic treatments.
Collapse
Affiliation(s)
- Sakhr A Murshid
- Department of Pedodontics, Orthodontics and Preventive Dentistry, Faculty of Dentistry, Thamar University, Thamar City, Yemen.
| |
Collapse
|
23
|
Clinkenbeard EL, Cass TA, Ni P, Hum JM, Bellido T, Allen MR, White KE. Conditional Deletion of Murine Fgf23: Interruption of the Normal Skeletal Responses to Phosphate Challenge and Rescue of Genetic Hypophosphatemia. J Bone Miner Res 2016; 31:1247-57. [PMID: 26792657 PMCID: PMC4891276 DOI: 10.1002/jbmr.2792] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 01/12/2023]
Abstract
The transgenic and knockout (KO) animals involving Fgf23 have been highly informative in defining novel aspects of mineral metabolism, but are limited by shortened lifespan, inability of spatial/temporal FGF23 control, and infertility of the global KO. To more finely test the role of systemic and genetic influences in FGF23 production, a mouse was developed that carried a floxed ("f")-Fgf23 allele (exon 2 floxed) which demonstrated in vivo recombination when bred to global-Cre transgenic mice (eIIa-cre). Mice homozygous for the recombined allele ("Δ") had undetectable serum intact FGF23, elevated serum phosphate (p < 0.05), and increased kidney Cyp27b1 mRNA (p < 0.05), similar to global Fgf23-KO mice. To isolate cellular FGF23 responses during phosphate challenge, Fgf23(Δ/f) mice were mated with early osteoblast type Iα1 collagen 2.3-kb promoter-cre mice (Col2.3-cre) and the late osteoblast/early osteocyte Dentin matrix protein-1-cre (Dmp1-cre). Fgf23(Δ/f) /Col2.3-cre(+) and Fgf23(Δ/f) /Dmp1-cre(+) exhibited reduced baseline serum intact FGF23 versus controls. After challenge with high-phosphate diet Cre(-) mice had 2.1-fold to 2.5-fold increased serum FGF23 (p < 0.01), but Col2.3-cre(+) mice had no significant increase, and Dmp1-cre(+) mice had only a 37% increase (p < 0.01) despite prevailing hyperphosphatemia in both models. The Fgf23(Δ/f) /Col2.3-cre was bred onto the Hyp (murine X-linked hypophosphatemia [XLH] model) genetic background to test the contribution of osteoblasts and osteocytes to elevated FGF23 and Hyp disease phenotypes. Whereas Hyp mice maintained inappropriately elevated FGF23 considering their marked hypophosphatemia, Hyp/Fgf23(Δ/f) /Col2.3-cre(+) mice had serum FGF23 <4% of Hyp (p < 0.01), and this targeted restriction normalized serum phosphorus and ricketic bone disease. In summary, deleting FGF23 within early osteoblasts and osteocytes demonstrated that both cell types contribute to baseline circulating FGF23 concentrations, and that targeting osteoblasts/osteocytes for FGF23 production can modify systemic responses to changes in serum phosphate concentrations and rescue the Hyp genetic syndrome. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Erica L. Clinkenbeard
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Taryn A. Cass
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pu Ni
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julia M. Hum
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew R. Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kenneth E. White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
24
|
Osteocytic connexin hemichannels suppress breast cancer growth and bone metastasis. Oncogene 2016; 35:5597-5607. [PMID: 27041582 PMCID: PMC5050050 DOI: 10.1038/onc.2016.101] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/13/2016] [Accepted: 01/25/2016] [Indexed: 12/15/2022]
Abstract
Although the skeleton is one of predominant sites for breast cancer metastasis, why breast cancer cells often become dormant after homing to bone is not well understood. Here, we reported an intrinsic self-defense mechanism of bone cells against breast cancer cells: a critical role of connexin (Cx) 43 hemichannels in osteocytes in the suppression of breast cancer bone metastasis. Cx43 hemichannels allow passage of small molecules between the intracellular and extracellular environments. The treatment of bisphosphonate drugs, either alendronate (ALN) or zoledronic acid (ZOL), opened Cx43 hemichannels in osteocytes. Conditioned media (CM) collected from MLO-Y4 osteocyte cells treated with bisphosphonates inhibited the anchorage-independent growth, migration and invasion of MDA-MB-231 human breast cancer cells and Py8119 mouse mammary carcinoma cells and this inhibitory effect was attenuated with Cx43(E2), a specific hemichannel blocking antibody. The opening of osteocytic Cx43 hemichannels by mechanical stimulation had similar inhibitory effects on breast cancer cells and this inhibition was attenuated by Cx43(E2) antibody as well. These inhibitory effects on cancer cells were mediated by ATP released from osteocyte Cx43 hemichannels. Furthermore, both Cx43 osteocyte-specific knockout mice and osteocyte-specific Δ130–136 transgenic mice with impaired Cx43 gap junctions and hemichannels showed significantly increased tumor growth and attenuated the inhibitory effect of ZOL. However, R76W transgenic mice with functional hemichannels but not gap junctions in osteocytes did not display a significant difference. Together, our studies establish the specific inhibitory role of osteocytic Cx43 hemichannels, and exploiting the activity of this channel could serve as a de novo therapeutic strategy.
Collapse
|
25
|
Parajuli A, Liu C, Li W, Gu X, Lai X, Pei S, Price C, You L, Lu XL, Wang L. Bone's responses to mechanical loading are impaired in type 1 diabetes. Bone 2015; 81:152-160. [PMID: 26183251 PMCID: PMC4640966 DOI: 10.1016/j.bone.2015.07.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 07/09/2015] [Accepted: 07/10/2015] [Indexed: 12/14/2022]
Abstract
Diabetes adversely impacts many organ systems including the skeleton. Clinical trials have revealed a startling elevation in fracture risk in diabetic patients. Bone fractures can be life threatening: nearly 1 in 6 hip fracture patients die within one year. Because physical exercise is proven to improve bone properties and reduce fracture risk in non-diabetic subjects, we tested its efficacy in type 1 diabetes. We hypothesized that diabetic bone's response to anabolic mechanical loading would be attenuated, partially due to impaired mechanosensing of osteocytes under hyperglycemia. Heterozygous C57BL/6-Ins2(Akita)/J (Akita) male and female diabetic mice and their age- and gender-matched wild-type (WT) C57BL/6J controls (7-month-old, N=5-7 mice/group) were subjected to unilateral axial ulnar loading with a peak strain of 3500 με at 2 Hz and 3 min/day for 5 days. The Akita female mice, which exhibited a relatively normal body weight and a mild 40% elevation of blood glucose level, responded with increased bone formation (+6.5% in Ct.B.Ar, and 4 to 36-fold increase in Ec.BFR/BS and Ps.BFR/BS), and the loading effects, in terms of changes of static and dynamic indices, did not differ between Akita and WT females (p ≥ 0.1). However, loading-induced anabolic effects were greatly diminished in Akita males, which exhibited reduced body weight, severe hyperglycemia (+230%), diminished bone formation (ΔCt.B.Ar: 0.003 vs. 0.030 mm(2), p=0.005), and suppressed periosteal bone appositions (ΔPs.BFR/BS, p=0.02). Hyperglycemia (25 mM glucose) was further found to impair the flow-induced intracellular calcium signaling in MLO-Y4 osteocytes, and significantly inhibited the flow-induced downstream responses including reduction in apoptosis and sRANKL secretion and PGE2 release. These results, along with previous findings showing adverse effects of hyperglycemia on osteoblasts and mesenchymal stem cells, suggest that failure to maintain normal glucose levels may impair bone's responses to mechanical loading in diabetics.
Collapse
Affiliation(s)
- Ashutosh Parajuli
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Chao Liu
- Department of Mechanical and Industrial Engineering, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| | - Wen Li
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Xiaoyu Gu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Xiaohan Lai
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Shaopeng Pei
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada.
| | - X Lucas Lu
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA; Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Liyun Wang
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA; Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
26
|
Kamel-ElSayed SA, Tiede-Lewis LM, Lu Y, Veno PA, Dallas SL. Novel approaches for two and three dimensional multiplexed imaging of osteocytes. Bone 2015; 76:129-40. [PMID: 25794783 PMCID: PMC4591054 DOI: 10.1016/j.bone.2015.02.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 01/20/2023]
Abstract
Although osteocytes have historically been viewed as quiescent cells, it is now clear that they are highly active cells in bone and play key regulatory roles in diverse skeletal functions, including mechanotransduction, phosphate homeostasis and regulation of osteoblast and osteoclast activity. Three dimensional imaging of embedded osteocytes and their dendritic connections within intact bone specimens can be quite challenging and many of the currently available methods are actually imaging the lacunocanalicular network rather than the osteocytes themselves. With the explosion of interest in the field of osteocyte biology, there is an increased need for reliable ways to image these cells in live and fixed bone specimens. Here we report the development of reproducible methods for 2D and 3D imaging of osteocytes in situ using multiplexed imaging approaches in which the osteocyte cell membrane, nucleus, cytoskeleton and extracellular matrix can be imaged simultaneously in various combinations. We also present a new transgenic mouse line expressing a membrane targeted-GFP variant selectively in osteocytes as a novel tool for in situ imaging of osteocytes and their dendrites in fixed or living bone specimens. These methods have been multiplexed with a novel method for labeling of the lacunocanalicular network using fixable dextran, which enables aspects of the osteocyte cell structure and lacunocanalicular system to be simultaneously imaged. The application of these comprehensive approaches for imaging of osteocytes in situ should advance research into osteocyte biology and function in health and disease.
Collapse
Affiliation(s)
- Suzan A Kamel-ElSayed
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, 615 E. 25th Street, Kansas City, MO 64108, USA; Biomedical Sciences Department, Oakland University William Beaumont School of Medicine, 414 O'Dowd Hall, Rochester MI, 48309, USA; Medical Physiology Department, Assiut University School of Medicine, 71516 Asyut, Egypt
| | - LeAnn M Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, 615 E. 25th Street, Kansas City, MO 64108, USA
| | - Yongbo Lu
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, 615 E. 25th Street, Kansas City, MO 64108, USA; Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry, 3302 Gaston Ave., Dallas, TX 75246, USA
| | - Patricia A Veno
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, 615 E. 25th Street, Kansas City, MO 64108, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, 615 E. 25th Street, Kansas City, MO 64108, USA.
| |
Collapse
|
27
|
Characterization of Fam20C expression in odontogenesis and osteogenesis using transgenic mice. Int J Oral Sci 2015; 7:89-94. [PMID: 25537657 PMCID: PMC4817552 DOI: 10.1038/ijos.2014.67] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2013] [Indexed: 11/23/2022] Open
Abstract
Our previous studies have demonstrated that Fam20C promotes differentiation and mineralization of odontoblasts, ameloblasts, osteoblasts and osteocytes during tooth and bone development. Ablation of the Fam20C gene inhibits bone and tooth growth by increasing fibroblast growth factor 23 in serum and causing hypophosphatemia in conditional knockout mice. However, control and regulation of the expression of Fam20C are still unknown. In this study, we generated a transgenic reporter model which expresses green fluorescence protein (GFP) driven by the Fam20C promoter. Recombineering was used to insert a 16 kb fragment of the mouse Fam20C gene (containing the 15 kb promoter and 1.1 kb of exon 1) into a pBluescript SK vector with the topaz variant of GFP and a bovine growth hormone polyadenylation sequence. GFP expression was subsequently evaluated by histomorphometry on cryosections from E14 to adult mice. Fluorescence was evident in the bone and teeth as early as E17.5. The GFP signal was maintained stably in odontoblasts and osteoblasts until 4 weeks after birth. The expression of GFP was significantly reduced in teeth, alveolar bone and muscle by 8 weeks of age. We also observed colocalization of the GFP signal with the Fam20C antibody in postnatal 1- and 7-day-old animals. Successful generation of Fam20C-GFP transgenic mice will provide a unique model for studying Fam20C gene expression and the biological function of this gene during odontogenesis and osteogenesis.
Collapse
|
28
|
Xu H, Gu S, Riquelme MA, Burra S, Callaway D, Cheng H, Guda T, Schmitz J, Fajardo RJ, Werner SL, Zhao H, Shang P, Johnson ML, Bonewald LF, Jiang JX. Connexin 43 channels are essential for normal bone structure and osteocyte viability. J Bone Miner Res 2015; 30:436-48. [PMID: 25270829 PMCID: PMC4333056 DOI: 10.1002/jbmr.2374] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 08/28/2014] [Accepted: 10/01/2014] [Indexed: 11/10/2022]
Abstract
Connexin (Cx) 43 serves important roles in bone function and development. Targeted deletion of Cx43 in osteoblasts or osteocytes leads to increased osteocyte apoptosis, osteoclast recruitment, and reduced biomechanical properties. Cx43 forms both gap junction channels and hemichannels, which mediate the communication between adjacent cells or between cell and extracellular environments, respectively. Two transgenic mouse models driven by a DMP1 promoter with the overexpression of dominant negative Cx43 mutants were generated to dissect the functional contribution of Cx43 gap junction channels and hemichannels in osteocytes. The R76W mutant blocks the gap junction channel, but not the hemichannel function, and the Δ130-136 mutant inhibits activity of both types of channels. Δ130-136 mice showed a significant increase in bone mineral density compared to wild-type (WT) and R76W mice. Micro-computed tomography (µCT) analyses revealed a significant increase in total tissue and bone area in midshaft cortical bone of Δ130-136 mice. The bone marrow cavity was expanded, whereas the cortical thickness was increased and associated with increased bone formation along the periosteal area. However, there is no significant alteration in the structure of trabecular bone. Histologic sections of the midshaft showed increased apoptotic osteocytes in Δ130-136, but not in WT and R76W, mice which correlated with altered biomechanical and estimated bone material properties. Osteoclasts were increased along the endocortical surface in both transgenic mice with a greater effect in Δ130-136 mice that likely contributed to the increased marrow cavity. Interestingly, the overall expression of serum bone formation and resorption markers were higher in R76W mice. These findings suggest that osteocytic Cx43 channels play distinctive roles in the bone; hemichannels play a dominant role in regulating osteocyte survival, endocortical bone resorption, and periosteal apposition, and gap junction communication is involved in the process of bone remodeling.
Collapse
Affiliation(s)
- Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xian, China
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Sumin Gu
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Manuel A. Riquelme
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Sirisha Burra
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Danielle Callaway
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Hongyun Cheng
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| | - Teja Guda
- Department of Biomedical Engineering, University of Texas at San Antonio, Texas
| | - James Schmitz
- Department of Orthopedics, University of Texas Health Science Center at San Antonio, Texas
| | - Roberto J. Fajardo
- Department of Orthopedics, University of Texas Health Science Center at San Antonio, Texas
| | - Sherry L. Werner
- Department of Pathology, University of Texas Health Science Center at San Antonio, Texas
| | - Hong Zhao
- Department of Oral Biology, School of Dentistry, University of Missouri, Kansas City, MO
| | - Peng Shang
- School of Life Sciences, Northwestern Polytechnical University, Xian, China
| | - Mark L. Johnson
- Department of Oral Biology, School of Dentistry, University of Missouri, Kansas City, MO
| | - Lynda F. Bonewald
- Department of Oral Biology, School of Dentistry, University of Missouri, Kansas City, MO
| | - Jean X. Jiang
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, Texas
| |
Collapse
|
29
|
Sapir-Koren R, Livshits G. Osteocyte control of bone remodeling: is sclerostin a key molecular coordinator of the balanced bone resorption-formation cycles? Osteoporos Int 2014; 25:2685-700. [PMID: 25030653 DOI: 10.1007/s00198-014-2808-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 07/02/2014] [Indexed: 12/22/2022]
Abstract
Osteocytes, entrapped within a newly mineralized bone matrix, possess a unique cellular identity due to a specialized morphology and a molecular signature. These features endow them to serve as a bone response mechanism for mechanical stress in their microenvironment. Sclerostin, a primarily osteocyte product, is widely considered as a mechanotranduction key molecule whose expression is suppressed by mechanical loading, or it is induced by unloading. This review presents a model suggesting that sclerostin is major mediator for integrating mechanical, local, and hormonal signals, sensed by the osteocytes, in controlling the remodeling apparatus. This central role is achieved through interplay between two opposing mechanisms: (1) unloading-induced high sclerostin levels, which antagonize Wnt-canonical-β-catenin signaling in osteocytes and osteoblasts, permitting simultaneously Wnt-noncanonical and/or other pathways in osteocytes and osteoclasts, directed at bone resorption; (2) mechanical loading results in low sclerostin levels, activation of Wnt-canonical signaling, and bone formation. Therefore, adaptive bone remodeling occurring at a distinct bone compartment is orchestrated by altered sclerostin levels, which regulate the expression of the other osteocyte-specific proteins, such as RANKL, OPG, and proteins encoded by "mineralization-related genes" (DMP1, PHEX, and probably FGF23). For example, under specific terms, sclerostin regulates differential RANKL and OPG production, and creates a dynamic RANKL/OPG ratio, leading either to bone formation or resorption. It also controls the expression of PHEX, DMP1, and most likely FGF23, leading to either bone matrix mineralization or its inhibition. Such opposing up- or down-regulation of remodeling phases allows osteocytes to function as an "external unit", ensuring transition from bone resorption to bone formation.Mini Abstract: The osteocyte network plays a central role in directing bone response either to mechanical loading, or to unloading, leading correspondingly to bone formation or resorption. This review shows a key role of the osteocyte-produced sclerostin as a major mediator of the molecular mechanisms involved in the process of adaptive bone remodeling.
Collapse
Affiliation(s)
- R Sapir-Koren
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | | |
Collapse
|
30
|
Takano-Yamamoto T. Osteocyte function under compressive mechanical force. JAPANESE DENTAL SCIENCE REVIEW 2014. [DOI: 10.1016/j.jdsr.2013.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
31
|
Abstract
Few investigators think of bone as an endocrine gland, even after the discovery that osteocytes produce circulating fibroblast growth factor 23 that targets the kidney and potentially other organs. In fact, until the last few years, osteocytes were perceived by many as passive, metabolically inactive cells. However, exciting recent discoveries have shown that osteocytes encased within mineralized bone matrix are actually multifunctional cells with many key regulatory roles in bone and mineral homeostasis. In addition to serving as endocrine cells and regulators of phosphate homeostasis, these cells control bone remodeling through regulation of both osteoclasts and osteoblasts, are mechanosensory cells that coordinate adaptive responses of the skeleton to mechanical loading, and also serve as a manager of the bone's reservoir of calcium. Osteocytes must survive for decades within the bone matrix, making them one of the longest lived cells in the body. Viability and survival are therefore extremely important to ensure optimal function of the osteocyte network. As we continue to search for new therapeutics, in addition to the osteoclast and the osteoblast, the osteocyte should be considered in new strategies to prevent and treat bone disease.
Collapse
Affiliation(s)
- Sarah L Dallas
- PhD, Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 650 East 25th Street, Kansas City, Missouri 64108.
| | | | | |
Collapse
|
32
|
Preosteocytes/osteocytes have the potential to dedifferentiate becoming a source of osteoblasts. PLoS One 2013; 8:e75204. [PMID: 24040401 PMCID: PMC3765403 DOI: 10.1371/journal.pone.0075204] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 08/11/2013] [Indexed: 01/23/2023] Open
Abstract
Presently there is no clear evidence for the ability of mature osteogenic lineage cells to dedifferentiate. In order to identify and trace mature osteogenic lineage cells, we have utilized transgenic mouse models in which the dentin matrix protein 1 (Dmp1) promoter drives expression of GFP (active marker) or Cre recombinase (historic label) in preosteocytes/osteocytes. In long bone chip outgrowth cultures, in which cells on the bone surface were enzymatically removed, cells with previous activity of the Dmp1 promoter migrated onto plastic and down-regulated Dmp1-GFP expression. Dmp1Cre-labeled cells from these cultures had the potential to re-differentiate into the osteogenic lineage, while the negative population showed evidence of adipogenesis. We observed numerous Dmp1Cre-labeled osteoblasts on the surface of bone chips following their in vivo transplantation. Our data indicate that cells embedded in bone matrix are motile, and once given access to the extra bony milieu will migrate out of their lacunae. This population of cells is phenotypically and functionally heterogeneous in vitro. Once the preosteocytes/osteocytes leave lacunae, they can dedifferentiate, potentially providing an additional source of functional osteoblasts.
Collapse
|
33
|
Abstract
Apoptotic death of osteocytes was recognized over 15 years ago, but its significance for bone homeostasis has remained elusive. A new paradigm has emerged that invokes osteocyte apoptosis as a critical event in the recruitment of osteoclasts to a specific site in response to skeletal unloading, fatigue damage, estrogen deficiency and perhaps in other states where bone must be removed. This is accomplished by yet to be defined signals emanating from dying osteocytes, which stimulate neighboring viable osteocytes to produce osteoclastogenic cytokines. The osteocyte apoptosis caused by chronic glucocorticoid administration does not increase osteoclasts; however, it does negatively impact maintenance of bone hydration, vascularity, and strength.
Collapse
Affiliation(s)
- Robert L Jilka
- Division of Endocrinology & Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, Central Arkansas Veterans Healthcare System, 4301 W. Markham, Slot 587, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
34
|
Kalajzic I, Matthews BG, Torreggiani E, Harris MA, Divieti Pajevic P, Harris SE. In vitro and in vivo approaches to study osteocyte biology. Bone 2013; 54:296-306. [PMID: 23072918 PMCID: PMC3566324 DOI: 10.1016/j.bone.2012.09.040] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/27/2012] [Accepted: 09/27/2012] [Indexed: 11/26/2022]
Abstract
Osteocytes, the most abundant cell population of the bone lineage, have been a major focus in the bone research field in recent years. This population of cells that resides within mineralized matrix is now thought to be the mechanosensory cell in bone and plays major roles in the regulation of bone formation and resorption. Studies of osteocytes had been impaired by their location, resulting in numerous attempts to isolate primary osteocytes and to generate cell lines representative of the osteocytic phenotype. Progress has been achieved in recent years by utilizing in vivo genetic technology and generation of osteocyte directed transgenic and gene deficiency mouse models. We will provide an overview of the current in vitro and in vivo models utilized to study osteocyte biology. We discuss generation of osteocyte-like cell lines and isolation of primary osteocytes and summarize studies that have utilized these cellular models to understand the functional role of osteocytes. Approaches that attempt to selectively identify and isolate osteocytes using fluorescent protein reporters driven by regulatory elements of genes that are highly expressed in osteocytes will be discussed. In addition, recent in vivo studies utilizing overexpression or conditional deletion of various genes using dentin matrix protein (Dmp1) directed Cre recombinase are outlined. In conclusion, evaluation of the benefits and deficiencies of currently used cell lines/genetic models in understanding osteocyte biology underlines the current progress in this field. The future efforts will be directed towards developing novel in vitro and in vivo models that would additionally facilitate in understanding the multiple roles of osteocytes.
Collapse
Affiliation(s)
- Ivo Kalajzic
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, Connecticut 06032, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Klein-Nulend J, Bakker AD, Bacabac RG, Vatsa A, Weinbaum S. Mechanosensation and transduction in osteocytes. Bone 2013; 54:182-90. [PMID: 23085083 DOI: 10.1016/j.bone.2012.10.013] [Citation(s) in RCA: 317] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 01/08/2023]
Abstract
The human skeleton is a miracle of engineering, combining both toughness and light weight. It does so because bones possess cellular mechanisms wherein external mechanical loads are sensed. These mechanical loads are transformed into biological signals, which ultimately direct bone formation and/or bone resorption. Osteocytes, since they are ubiquitous in the mineralized matrix, are the cells that sense mechanical loads and transduce the mechanical signals into a chemical response. The osteocytes then release signaling molecules, which orchestrate the recruitment and activity of osteoblasts or osteoclasts, resulting in the adaptation of bone mass and structure. In this review, we highlight current insights in bone adaptation to external mechanical loading, with an emphasis on how a mechanical load placed on whole bones is translated and amplified into a mechanical signal that is subsequently sensed by the osteocytes.
Collapse
Affiliation(s)
- Jenneke Klein-Nulend
- Department of Oral Cell Biology, ACTA-VU University Amsterdam, Research Institute MOVE, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
36
|
Gibson MP, Zhu Q, Liu Q, D'Souza RN, Feng JQ, Qin C. Loss of dentin sialophosphoprotein leads to periodontal diseases in mice. J Periodontal Res 2013; 48:221-7. [PMID: 22934831 PMCID: PMC3514631 DOI: 10.1111/j.1600-0765.2012.01523.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2012] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVE Dentin sialophosphoprotein (DSPP) and its cleaved products, dentin phosphoprotein (DPP) and dentin sialoprotein (DSP), play important roles in biomineralization. Recently, we observed that DSPP is highly expressed in the alveolar bone and cementum, indicating that this molecule may play an important role in the formation and maintenance of a healthy periodontium, and its deletion may cause increased susceptibility to periodontal diseases. The objective of this investigation was to study the effects of Dspp ablation on periodontal tissues by analyzing Dspp null mice. MATERIAL AND METHODS Newborn to 6-mo-old Dspp null mice were examined, and the 3- and 6-mo-old Dspp null mice were characterized in detail using X-ray radiography, histology and scanning electron microscopy (backscattered as well as resin-infiltrating). Wild-type mice of the same age groups served as the normal controls. RESULTS The Dspp null mice showed significant loss of alveolar bone and cementum, particularly in the furcation and interproximal regions of the molars. The alveolar bone appeared porous while the quantity of cementum was reduced in the apical region. The canalicular systems and osteocytes in the alveolar bone were abnormal, with reduced numbers of canaliculi and altered osteocyte morphology. The loss of alveolar bone and cementum along with the detachment of the periodontal ligaments (PDL) led to the apical migration of the epithelial attachment and formation of periodontal pockets. CONCLUSION Inactivation of DSPP leads to the loss of alveolar bone and cementum and increased susceptibility to bacterial infections in PDL of Dspp null mice. The fact that the loss of DSPP results in periodontal diseases indicates that this molecule plays a vital role in maintaining the health of the periodontium.
Collapse
Affiliation(s)
- M P Gibson
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, TX, USA
| | | | | | | | | | | |
Collapse
|
37
|
Foster BL, Nagatomo KJ, Nociti FH, Fong H, Dunn D, Tran AB, Wang W, Narisawa S, Millán JL, Somerman MJ. Central role of pyrophosphate in acellular cementum formation. PLoS One 2012; 7:e38393. [PMID: 22675556 PMCID: PMC3366957 DOI: 10.1371/journal.pone.0038393] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 05/09/2012] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Inorganic pyrophosphate (PP(i)) is a physiologic inhibitor of hydroxyapatite mineral precipitation involved in regulating mineralized tissue development and pathologic calcification. Local levels of PP(i) are controlled by antagonistic functions of factors that decrease PP(i) and promote mineralization (tissue-nonspecific alkaline phosphatase, Alpl/TNAP), and those that increase local PP(i) and restrict mineralization (progressive ankylosis protein, ANK; ectonucleotide pyrophosphatase phosphodiesterase-1, NPP1). The cementum enveloping the tooth root is essential for tooth function by providing attachment to the surrounding bone via the nonmineralized periodontal ligament. At present, the developmental regulation of cementum remains poorly understood, hampering efforts for regeneration. To elucidate the role of PP(i) in cementum formation, we analyzed root development in knock-out ((-/-)) mice featuring PP(i) dysregulation. RESULTS Excess PP(i) in the Alpl(-/-) mouse inhibited cementum formation, causing root detachment consistent with premature tooth loss in the human condition hypophosphatasia, though cementoblast phenotype was unperturbed. Deficient PP(i) in both Ank and Enpp1(-/-) mice significantly increased cementum apposition and overall thickness more than 12-fold vs. controls, while dentin and cellular cementum were unaltered. Though PP(i) regulators are widely expressed, cementoblasts selectively expressed greater ANK and NPP1 along the root surface, and dramatically increased ANK or NPP1 in models of reduced PP(i) output, in compensatory fashion. In vitro mechanistic studies confirmed that under low PP(i) mineralizing conditions, cementoblasts increased Ank (5-fold) and Enpp1 (20-fold), while increasing PP(i) inhibited mineralization and associated increases in Ank and Enpp1 mRNA. CONCLUSIONS Results from these studies demonstrate a novel developmental regulation of acellular cementum, wherein cementoblasts tune cementogenesis by modulating local levels of PP(i), directing and regulating mineral apposition. These findings underscore developmental differences in acellular versus cellular cementum, and suggest new approaches for cementum regeneration.
Collapse
Affiliation(s)
- Brian L Foster
- Laboratory of Oral Connective Tissue Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Osteocytes, the cells residing within the bone matrix and comprising 90% to 95% of the all bone cells, have long been considered quiescent bystander cells compared to the osteoblasts and osteoclasts whose activities cause bone gain and loss, and whose dysfunction lead to growth defects and osteoporosis. However, recent studies show that osteocytes play a crucial, central role in regulating the dynamic nature of bone in all its diverse functions. Osteocytes are now known to be the principal sensors for mechanical loading of bone. They produce the soluble factors that regulate the onset of both bone formation and resorption. Osteocytes regulate local mineral deposition and chemistry at the bone matrix level, and they also function as endocrine cells producing factors that target distant organs such as the kidney to regulate phosphate transport. Osteocytes appear to be the major local orchestrator of many of bone's functions.
Collapse
Affiliation(s)
- Mitchell B Schaffler
- New York Center for Biomedical Engineering, City College of New York, 160 Convent Avenue, New York, NY, 10031, USA.
| | | |
Collapse
|
39
|
Stern AR, Stern MM, Van Dyke ME, Jähn K, Prideaux M, Bonewald LF. Isolation and culture of primary osteocytes from the long bones of skeletally mature and aged mice. Biotechniques 2012; 52:361-73. [PMID: 22668415 PMCID: PMC3612989 DOI: 10.2144/0000113876] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 05/18/2012] [Indexed: 12/20/2022] Open
Abstract
The purpose of this work was to establish a methodology to enable the isolation and study of osteocytes from skeletally mature young (4-month-old) and old (22-month-old) mice. The location of osteocytes deep within bone is ideal for their function as mechanosensors. However, this location makes the observation and study of osteocytes in vivo technically difficult. Osteocytes were isolated from murine long bones through a process of extended collagenase digestions combined with EDTA-based decalcification. A tissue homogenizer was used to reduce the remaining bone fragments to a suspension of bone particles, which were placed in culture to yield an outgrowth of osteocyte-like cells. All of the cells obtained from this outgrowth that displayed an osteocyte-like morphology stained positive for the osteocyte marker E11/GP38. The osteocyte phenotype was further confirmed by a lack of staining for alkaline phosphatase and the absence of collagen1a1 expression. The outgrowth of osteocytes also expressed additional osteocyte-specific genes such as Sost and Mepe. This technique facilitates the isolation of osteocytes from skeletally mature bone. This novel enabling methodology should prove useful in advancing our understanding of the roles mature osteocytes play in bone health and disease.
Collapse
Affiliation(s)
- Amber Rath Stern
- Mechanical Engineering and Oral Biology, University of Missouri Kansas City, Kansas City, MO, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The idea that osteoblasts, or their progenitors, support osteoclast formation by expressing the cytokine receptor activator of NFkB ligand (RANKL) is a widely held tenet of skeletal biology. Two recent studies provide evidence that osteocytes, and not osteoblasts or their progenitors, are the major source of RANKL driving osteoclast formation in cancellous bone. The goal of this review is to highlight the results of these new studies and discuss their implications for our understanding of bone remodeling.
Collapse
Affiliation(s)
- Jinhu Xiong
- Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | |
Collapse
|
41
|
|
42
|
Tu X, Rhee Y, Condon K, Bivi N, Allen MR, Dwyer D, Stolina M, Turner CH, Robling AG, Plotkin LI, Bellido T. Sost downregulation and local Wnt signaling are required for the osteogenic response to mechanical loading. Bone 2012; 50:209-17. [PMID: 22075208 PMCID: PMC3246572 DOI: 10.1016/j.bone.2011.10.025] [Citation(s) in RCA: 347] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/30/2011] [Accepted: 10/25/2011] [Indexed: 01/16/2023]
Abstract
Sclerostin, the Wnt signaling antagonist encoded by the Sost gene, is secreted by osteocytes and inhibits bone formation by osteoblasts. Mechanical stimulation reduces sclerostin expression, suggesting that osteocytes might coordinate the osteogenic response to mechanical force by locally unleashing Wnt signaling. To investigate whether sclerostin downregulation is a pre-requisite for load-induced bone formation, we conducted experiments in transgenic mice (TG) engineered to maintain high levels of SOST expression during mechanical loading. This was accomplished by introducing a human SOST transgene driven by the 8 kb fragment of the DMP1 promoter that also provided osteocyte specificity of the transgene. Right ulnae were subjected to in vivo cyclic axial loading at equivalent strains for 1 min/day at 2 Hz; left ulnae served as internal controls. Endogenous murine Sost mRNA expression measured 24 h after 1 loading bout was decreased by about 50% in TG and wild type (WT) littermates. In contrast, human SOST, only expressed in TG mice, remained high after loading. Mice were loaded on 3 consecutive days and bone formation was quantified 16 days after initiation of loading. Periosteal bone formation in control ulnae was similar in WT and TG mice. Loading induced the expected strain-dependent increase in bone formation in WT mice, resulting from increases in both mineralizing surface (MS/BS) and mineral apposition rate (MAR). In contrast, load-induced bone formation was reduced by 70-85% in TG mice, due to lower MS/BS and complete inhibition of MAR. Moreover, Wnt target gene expression induced by loading in WT mice was absent in TG mice. Thus, downregulation of Sost/sclerostin in osteocytes is an obligatory step in the mechanotransduction cascade that activates Wnt signaling and directs osteogenesis to where bone is structurally needed.
Collapse
Affiliation(s)
- Xiaolin Tu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yumie Rhee
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith Condon
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicoletta Bivi
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew R. Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Denise Dwyer
- Metabolic Research Department, Amgen Inc., Thousand Oaks, CA, USA
| | - Marina Stolina
- Metabolic Research Department, Amgen Inc., Thousand Oaks, CA, USA
| | - Charles H. Turner
- Department of Orthopedic Surgery, Biomechanics and Biomaterials Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G. Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lilian I. Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Teresita Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
- Corresponding author and reprint requests: Teresita Bellido, Ph.D., Department of Anatomy and Cell Biology, and Department of Internal Medicine, Division of Endocrinology, Indiana University School of Medicine, 635 Barnhill Drive, MS5035, Indianapolis, IN 46202, Phone 317-274-7410, Fax 317-278-2040,
| |
Collapse
|
43
|
Abstract
Over the past two decades there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point, but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, we have focused on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory, together with detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- School of Dentistry/Department of Oral Biology, University of Missouri, Kansas City, MO, USA.
| | | |
Collapse
|
44
|
Elefteriou F, Yang X. Genetic mouse models for bone studies--strengths and limitations. Bone 2011; 49:1242-54. [PMID: 21907838 PMCID: PMC3331798 DOI: 10.1016/j.bone.2011.08.021] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/15/2011] [Accepted: 08/18/2011] [Indexed: 11/25/2022]
Abstract
Mice have become a preferred model system for bone research because of their genetic and pathophysiological similarities to humans: a relatively short reproductive period, leading to relatively low cost of maintenance and the availability of the entire mouse genome sequence information. The success in producing the first transgenic mouse line that expressed rabbit β-globin protein in mouse erythrocytes three decades ago marked the beginning of the use of genetically engineered mice as model system to study human diseases. Soon afterward the development of cultured pluripotent embryonic stem cells provided the possibility of gene replacement or gene deletion in mice. These technologies have been critical to identify new genes involved in bone development, growth, remodeling, repair, and diseases, but like many other approaches, they have limitations. This review will introduce the approaches that allow the generation of transgenic mice and global or conditional (tissue-specific and inducible) mutant mice. A list of the various promoters used to achieve bone-specific gene deletion or overexpression is included. The limitations of these approaches are discussed, and general guidelines related to the analysis of genetic mouse models are provided.
Collapse
Affiliation(s)
- Florent Elefteriou
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| | - Xiangli Yang
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| |
Collapse
|
45
|
Lu Y, Thiagarajan G, Nicolella DP, Johnson ML. Load/strain distribution between ulna and radius in the mouse forearm compression loading model. Med Eng Phys 2011; 34:350-6. [PMID: 21903442 DOI: 10.1016/j.medengphy.2011.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 07/24/2011] [Accepted: 07/26/2011] [Indexed: 10/17/2022]
Abstract
Finite element analysis (FEA) of the mouse forearm compression loading model is used to relate strain distributions with downstream changes in bone formation and responses of bone cells. The objective of this study was to develop two FEA models - the first one with the traditional ulna only and the second one in which both the ulna and radius are included, in order to examine the effect of the inclusion of the radius on the strain distributions in the ulna. The entire mouse forearm was scanned using microCT and images were converted into FEA tetrahedral meshes using a suite of software programs. The performance of both linear and quadratic tetrahedral elements and coarse and fine meshes were studied. A load of 2N was applied to the ulna/radius model and a 1.3N load (based on previous investigations of load sharing between the ulna and radius in rats) was applied to the ulna only model for subsequent simulations. The results showed differences in the cross sectional strain distributions and magnitude within the ulna for the combined ulna/radius model versus the ulna only model. The maximal strain in the combined model occurred about 4mm toward the distal end from the ulna mid-shaft in both models. Results from the FEA model simulations were also compared to experimentally determined strain values. We conclude that inclusion of the radius in FE models to predict strains during in vivo forearm loading increases the magnitude of the estimated ulna strains compared to those predicted from a model of the ulna alone but the distribution was similar. This has important ramifications for future studies to understand strain thresholds needed to activate bone cell responses to mechanical loading.
Collapse
Affiliation(s)
- Yunkai Lu
- Department of Civil and Mechanical Engineering, University of Missouri-Kansas City, Kansas City, MO 64110, United States
| | | | | | | |
Collapse
|
46
|
Powell WF, Barry KJ, Tulum I, Kobayashi T, Harris SE, Bringhurst FR, Pajevic PD. Targeted ablation of the PTH/PTHrP receptor in osteocytes impairs bone structure and homeostatic calcemic responses. J Endocrinol 2011; 209:21-32. [PMID: 21220409 PMCID: PMC3783949 DOI: 10.1530/joe-10-0308] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Parathyroid hormone (PTH) is a major physiologic regulator of calcium, phosphorous, and skeletal homeostasis. Cells of the osteoblastic lineage are key targets of PTH action in bone, and recent evidence suggests that osteocytes might be important in the anabolic effects of PTH. To understand the role of PTH signaling through the PTH/PTHrP receptors (PPR) in osteocytes and to determine the role(s) of these cells in mediating the effects of the hormone, we have generated mice in which PPR expression is specifically ablated in osteocytes. Transgenic mice in which the 10 kb-Dmp1 promoter drives a tamoxifen-inducible Cre-recombinase were mated with animals in which exon 1 of PPR is flanked by lox-P sites. In these animals, osteocyte-selective PPR knockout (Ocy-PPR(cKO) mice) could be induced by administration of tamoxifen. Histological analysis revealed a reduction in trabecular bone and mild osteopenia in Ocy-PPR(cKO) mice. Reduction of trabeculae number and thickness was also detected by micro-computed tomography analysis whereas bone volume fraction (BV/TV%) was unchanged. These findings were associated with an increase in Sost and sclerostin expression. When Ocy-PPR(cKO) mice were subjected to a low-calcium diet to induce secondary hyperparathyroidism, their blood calcium levels were significantly lower than littermate controls. Moreover, PTH was unable to suppress Sost and sclerostin expression in the Ocy-PPR(cKO) animals, suggesting an important role of PTH signaling in osteocytes for proper bone remodeling and calcium homeostasis.
Collapse
Affiliation(s)
- William F. Powell
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Kevin J. Barry
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Irena Tulum
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Tatsuya Kobayashi
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Stephen E. Harris
- Department of Periodontics, University of Texas Health Science Center School of Dentistry, San Antonio, TX
| | - F. Richard Bringhurst
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| | - Paola Divieti Pajevic
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston MA
| |
Collapse
|
47
|
Balic A, Mina M. Identification of secretory odontoblasts using DMP1-GFP transgenic mice. Bone 2011; 48:927-37. [PMID: 21172466 PMCID: PMC3062740 DOI: 10.1016/j.bone.2010.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2010] [Revised: 12/02/2010] [Accepted: 12/12/2010] [Indexed: 10/18/2022]
Abstract
Terminal differentiation of odontoblasts from dental papilla is a long process involving several intermediate steps and changes in the transcriptional profile and expression of proteins secreted by cells in the odontoblast lineage. Transgenic mouse lines in which GFP expression is under the control of tissue- and stage specific promoters have provided powerful experimental tools for identification and isolation of cells at specific stages of differentiation along a lineage. Our previous studies showed utilization of pOBCol3.6GFP and pOBCol2.3GFP animals for identification of odontoblasts at early and late stages of polarization respectively. In the present study we used the DMP1-GFP transgenic animal as an experimental model to examine its expression during the differentiation of odontoblasts from progenitor cells in vivo and in vitro. Our observations showed that DMP1-GFP transgene is first activated in secretory/functional odontoblasts engaged in secretion of predentin and then transiently expressed at high levels in newly differentiated odontoblasts. Expression of DMP1-GFP was down-regulated in highly differentiated odontoblasts. The temporal and spatial pattern of expression of DMP1-GFP transgene closely mimics the expression of endogenous DMP1. This transgenic animal will facilitate studies of gene expression and biological functions in secretory/functional odontoblasts.
Collapse
Affiliation(s)
- Anamaria Balic
- Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|
48
|
Foster BL, Nagatomo KJ, Bamashmous SO, Tompkins KA, Fong H, Dunn D, Chu EY, Guenther C, Kingsley DM, Rutherford RB, Somerman MJ. The progressive ankylosis protein regulates cementum apposition and extracellular matrix composition. Cells Tissues Organs 2011; 194:382-405. [PMID: 21389671 DOI: 10.1159/000323457] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND/AIMS Tooth root cementum is sensitive to modulation of inorganic pyrophosphate (PP(i)), an inhibitor of hydroxyapatite precipitation. Factors increasing PP(i) include progressive ankylosis protein (ANK) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) while tissue nonspecific alkaline phosphatase hydrolyzes PP(i). Studies here aimed to define the role of ANK in root and cementum by analyzing tooth development in Ank knock-out (KO) mice versus wild type. MATERIALS AND METHODS Periodontal development in KO versus control mice was analyzed by histology, histomorphometry, immunohistochemistry, in situ hybridization, electron microscopy, and nanoindentation. Cementoblast cultures were used in vitro to provide mechanistic underpinnings for PP(i) modulation of cell function. RESULTS Over the course of root development, Ank KO cervical cementum became 8- to 12-fold thicker than control cervical cementum. Periodontal ligament width was maintained and other dentoalveolar tissues, including apical cementum, were unaltered. Cervical cementum uncharacteristically included numerous cells, from rapid cementogenesis. Ank KO increased osteopontin and dentin matrix protein 1 gene and protein expression, and markedly increased NPP1 protein expression in cementoblasts but not in other cell types. Conditional ablation of Ank in joints and periodontia confirmed a local role for ANK in cementogenesis. In vitro studies employing cementoblasts indicated that Ank and Enpp1 mRNA levels increased in step with mineral nodule formation, supporting a role for these factors in regulation of cementum matrix mineralization. CONCLUSION ANK, by modulating local PP(i), controls cervical cementum apposition and extracellular matrix. Loss of ANK created a local environment conducive to rapid cementogenesis; therefore, approaches modulating PP(i) in periodontal tissues have potential to promote cementum regeneration.
Collapse
Affiliation(s)
- B L Foster
- Department of Periodontics, University of Washington School of Dentistry, Seattle, Wash. 98195, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Mazzaferro S, Pasquali M, Pirrò G, Rotondi S, Tartaglione L. The bone and the kidney. Arch Biochem Biophys 2010; 503:95-102. [PMID: 20599669 DOI: 10.1016/j.abb.2010.06.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 06/24/2010] [Accepted: 06/26/2010] [Indexed: 12/21/2022]
Abstract
Renal tubular diseases may present with osteopenia, osteoporosis or osteomalacia, as a result of significant derangements in body electrolytes. In case of insufficient synthesis of calcitriol, as in renal failure, the more complex picture of renal osteodystrophy may develop. Hypothetically, also disturbed renal production of BMP-7 and Klotho could cause bone disease. However, the acknowledgment that osteocytes are capable of producing FGF23, a phosphaturic hormone at the same time modulating renal synthesis of calcitriol, indicates that it is also bone that can influence renal function. Importantly, a feed-back mechanism exists between FGF23 and calcitriol synthesis, while Klotho, produced by the kidney, determines activity and selectivity of FGF23. Identification of human diseases linked to disturbed production of FGF23 and Klotho underlines the importance of this new bone-kidney axis. Kidney and bone communicate reciprocally to regulate the sophisticated machinery responsible for divalent ions homeostasis and for osseous or extraosseous mineralisation processes.
Collapse
|
50
|
Kalogeropoulos M, Varanasi SS, Olstad OK, Sanderson P, Gautvik VT, Reppe S, Francis RM, Gautvik KM, Birch MA, Datta HK. Zic1 transcription factor in bone: neural developmental protein regulates mechanotransduction in osteocytes. FASEB J 2010; 24:2893-903. [DOI: 10.1096/fj.09-148908] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michail Kalogeropoulos
- Musculoskeletal Research GroupInstitute of Cellular MedicineThe Medical School Newcastle upon Tyne UK
| | - Satya S. Varanasi
- Musculoskeletal Research GroupInstitute of Cellular MedicineThe Medical School Newcastle upon Tyne UK
| | - Ole K. Olstad
- Department of Clinical ChemistryOslo University Hospital Ullevaal Oslo Norway
| | - Paul Sanderson
- Department of Orthopaedic SurgeryThe Newcastle upon Tyne NHS Foundation Trust Hospitals Newcastle upon Tyne UK
| | - Vigdis T. Gautvik
- Department of Clinical ChemistryLovisenberg Deacon Hospital Oslo Norway
| | - Sjur Reppe
- Department of Clinical ChemistryLovisenberg Deacon Hospital Oslo Norway
| | - Roger M. Francis
- Institute for Ageing and HealthNewcastle University Newcastle upon Tyne UK
| | - Kaare M. Gautvik
- Department of Clinical ChemistryOslo University Hospital Ullevaal Oslo Norway
- Department of Clinical ChemistryLovisenberg Deacon Hospital Oslo Norway
- Institute of Basic Medical SciencesUniversity of Oslo Oslo Norway
| | - Mark A. Birch
- Musculoskeletal Research GroupInstitute of Cellular MedicineThe Medical School Newcastle upon Tyne UK
| | - Harish K. Datta
- Musculoskeletal Research GroupInstitute of Cellular MedicineThe Medical School Newcastle upon Tyne UK
| |
Collapse
|