1
|
Woo SM, Paek K, Yoon YM, Kim H, Park SI, Kim JA. Development of a BMU-on-a-chip model based on spatiotemporal regulation of cellular interactions in the bone remodeling cycle. Mater Today Bio 2025; 32:101658. [PMID: 40206145 PMCID: PMC11979395 DOI: 10.1016/j.mtbio.2025.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Bone remodeling is essential for maintaining bone homeostasis throughout life by replacing old bone with new tissue. This dynamic process occurs continuously within basic multicellular unit (BMU) through well-coordinated interactions among osteocytes, osteoblasts, and osteoclasts. However, a precise in vitro model that accurately replicates this mechanism has not yet been developed. In this study, we created a human in vitro BMU-modeling chip platform by tri-culturing cells within a chip unit integrated into a tissue culture well plate, enabling high-throughput three-dimensional (3D) cell culture. To establish the tri-culture, human osteoblasts were isolated from human surgical bone samples and differentiated into osteocytes within collagen gel inside the chip unit. Subsequently, osteoblasts and peripheral blood mononuclear cells (PBMCs) containing osteoclast precursors were added to the chip unit. To simulate each phase of the bone remodeling cycle, we optimized the tri-culture process by adjusting the timing and using two types of osteoblasts at different stages of differentiation. The completed tri-culture model successfully mimicked the bone formation phase. When receptor activators of nuclear factor kappa-Β (RANKL) and macrophage colony-stimulating factor (M-CSF) were introduced, the cells exhibited characteristics of the reversal phase, where osteogenic and osteoclastogenic environments coexist. Additionally, using more differentiated osteoblasts within the tri-culture platform induced osteoclast differentiation, resembling the bone resorption phase. Overall, our model effectively replicates each phase of the bone remodeling cycle in BMUs, both spatially and temporally. This advancement not only facilitates the study of the intricate mechanisms of bone remodeling and cellular function but also aids drug development by providing a robust bone model for testing target drugs.
Collapse
Affiliation(s)
- Sang-Mi Woo
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Kyurim Paek
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Yeo Min Yoon
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
| | - Hyang Kim
- Institute of New Horizon Regenerative Medicine, Myongji Hospital, Goyang, 10475, Republic of Korea
| | - Serk In Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jeong Ah Kim
- Center for Scientific Instrumentation, Korea Basic Science Institute, Daejeon, 34133, Republic of Korea
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon, 34113, Republic of Korea
- Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, 06973, Republic of Korea
| |
Collapse
|
2
|
Chen S, Tang M, Yu X, Qian W, Xu Y, Li J, Wu G, Zhang S. A microprotein encoded by LINC00263 promotes breast cancer osteolytic bone metastasis by inducing osteoclastogenesis and inhibiting osteoclast ferroptosis. Oncogene 2025:10.1038/s41388-025-03400-5. [PMID: 40221529 DOI: 10.1038/s41388-025-03400-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Currently, there are no effective prevention or therapeutic methods for breast cancer bone metastasis (BC-BM), which leading to severe skeletal complications and increased mortality. Understanding the mechanisms underlying BC-BM could provide potential strategies for its prevention and treatment. In this study, we identified a new microprotein encoded by lncRNA LINC00263, which we named LINC00263-encoded protein (LINC00263-P), was significantly upregulated in bone metastatic breast cancer tissues and correlated with BC-BM. Overexpression of LINC00263 significantly promoted BC-BM, while treatment with the neutralizing anti-LINC00263-P antibody effectively inhibited BC-BM. Mechanically, the LINC00263-P binds to integrin αvβ3 for activating Src/Syk/Vav-3 axis and yes-associated protein 1 (YAP1) pathway, which enhanced osteoclastogenesis and diminishes ferroptosis in osteoclasts, thereby creating an osteolytic bone metastasis niche that fosters BC-BM. Importantly, treatment with angoroside C, an active component from the traditional Chinese medicine Scrophulariae Radix extract, effectively blocked the binding of LINC00263-P to αvβ3, thereby inhibiting abnormal osteoclastogenesis and preventing BC-BM. These findings highlight the crucial role of microprotein LINC00263-P in disrupting bone homeostasis and propose a potential molecular mechanism of BC-BM.
Collapse
Affiliation(s)
- Suwen Chen
- Department of Oncology, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Oncobiology, Department of Basic Medical Sciences, Shantou University Medical College, Shantou, Guangdong, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Miaoling Tang
- Department of Oncology, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Oncobiology, Department of Basic Medical Sciences, Shantou University Medical College, Shantou, Guangdong, China
| | - Xuexin Yu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wanying Qian
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yingru Xu
- Molecular Diagnosis and Gene Testing Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Geyan Wu
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| | - Shuxia Zhang
- Department of Oncobiology, Department of Basic Medical Sciences, Shantou University Medical College, Shantou, Guangdong, China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
3
|
Barin IAR, da Silva JC, Ramos RF, Lima SMDF, Cantuária APDC, Silva PAO, Dantas EMGL, Martins DCM, de Oliveira NG, Martínez OF, de Almeida JA, Ramada MHS, Franco OL, Rezende TMB. Antibiofilm, regenerative and bone homeostasis potential of the synergistic association of synoeca-MP peptide with chlorhexidine in oral cavity opportunistic infections. Arch Oral Biol 2025; 172:106177. [PMID: 39889438 DOI: 10.1016/j.archoralbio.2025.106177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE Synoeca-MP is an antimicrobial peptide that belongs to the class of defense peptides, known for their antimicrobial and immunomodulatory properties. To evaluate in vitro the association between synoeca-MP peptide and chlorhexidine, regarding their antimicrobial and antibiofilm activities, saliva stability, effect on tissue repair, bone resorption processes, and mineralized matrix formation. DESIGN Initially, the minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and antibiofilm concentration were determined. The synergism and degradation of synoeca-MP and chlorhexidine in human saliva were assessed. Furthermore, biocompatibility was evaluated using MTT assays, hemolytic assays, and proliferation and migration assays of periodontal ligament cells. Finally, bone homeostasis was evaluated through osteoclastogenesis assays, alkaline phosphatase determination, and mineralized matrix formation assay with SaOs-2 and ligament cells. RESULTS The antimicrobial and antibiofilm activity against the tested microorganisms was confirmed. Low synergistic concentrations of the synoeca-MP and chlorhexidine combination inhibited tested microorganisms. The association of these molecules remained stable in healthy saliva. Nevertheless, it degraded as the severity of periodontal disease increased. Additionally, lower synergistic concentrations of the combination were not cytotoxic to human cells, promoted the proliferation and migration of ligament cells, inhibited osteoclastogenesis, and increased mineral matrix formation of ligament cells and SaOs-2. CONCLUSIONS Synoeca-MP and chlorhexidine combination shows potential for oral diseases treatment, as evidenced by its antimicrobial activity, regenerative potential, saliva stability, and bone homeostasis. It may be particularly effective for opportunistic oral infections and in conjunction with mechanical therapy.
Collapse
Affiliation(s)
- Ingrid Aquino Reichert Barin
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil
| | - Johnny Carvalho da Silva
- Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil
| | - Raquel Figuerêdo Ramos
- Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil
| | - Stella Maris de Freitas Lima
- Curso de Odontologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil; Curso de Odontologia, Centro Universitário do Planalto Central Aparecido dos Santos, Gama, Brasília, Distrito Federal, Brazil
| | - Ana Paula de Castro Cantuária
- Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil
| | - Poliana Amanda Oliveira Silva
- Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil
| | | | - Danilo César Mota Martins
- Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil; Curso de Odontologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil
| | - Nelson Gomes de Oliveira
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil
| | - Osmel Fleitas Martínez
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil
| | - Jeeser Alves de Almeida
- Laboratório de Pesquisa em Exercício e Nutrição na Saúde e Rendimento Esportivo - PENSARE, Universidade Federal do Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | - Marcelo Henrique Soller Ramada
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil
| | - Octávio Luiz Franco
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil; Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul, Brazil
| | - Taia Maria Berto Rezende
- Pós-graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, QS 07 Lote 01, Brasília, Distrito Federal, Brazil; Pós-graduação em Ciências da Saúde, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil; Pós-graduação em Odontologia, Faculdade de Ciências de Saúde, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil; Departamento de Odontologia, Universidade de Brasília, Campus Darcy Ribeiro s/n - Asa Norte, Brasília, Distrito Federal, Brazil.
| |
Collapse
|
4
|
Lang J, Ding A, Henninger E, Reese S, Helzer K, Hazelberg X, de Diego CS, Kerr S, Sethakorn N, Bootsma M, Zhao S, Beebe D. Live Cell Sorting of Differentiated Primary Human Osteoclasts Allows Generation of Transcriptomic Signature Matrix. RESEARCH SQUARE 2025:rs.3.rs-6157400. [PMID: 40235499 PMCID: PMC11998790 DOI: 10.21203/rs.3.rs-6157400/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Osteoclasts are specialized cells that degrade the bone matrix to create space for bone regeneration. During tumorigenesis, cancer cells metastasize to bone by disrupting bone's natural remodeling cycle. However, the mechanisms underlying critical bone-tumor interactions are poorly understood due to challenges in isolating osteoclasts from human bone. Thus, the conventional method to obtain osteoclasts for in vitro studies is via the differentiation of peripheral blood monocytes, which results in mixed cultures containing progenitor cells and osteoclasts of varying maturity and nuclearity. Presently, we hypothesized that the transcriptomic signatures of mature, multinucleated osteoclasts are distinct from osteoclasts with fewer nuclei. We established a live cell biomarker expression-based sorting protocol to allow purification of mature osteoclasts while maintaining viability and function. We observed that mature, multinucleated osteoclasts were transcriptomically distinct from those with fewer nuclei and that mature osteoclasts showed higher expression of genes that are associated with osteoclast fusion and function.
Collapse
|
5
|
Bishop RT, Delgado-Calle J, Reagan MR, Marino S. 2D and 3D In Vitro Co-culture for Cancer and Bone Cell Interaction Studies. Methods Mol Biol 2025; 2885:113-141. [PMID: 40448759 DOI: 10.1007/978-1-0716-4306-8_7] [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
Co-culture assays are used to study the mutual interaction between cells in vitro. This chapter describes 2D and 3D co-culture systems used to study cell-cell signaling crosstalk between cancer cells and bone marrow adipocytes, osteoblasts, osteoclasts, and osteocytes. The chapter provides a step-by-step guide to the most used cell culture techniques, functional assays, and gene expression.
Collapse
Affiliation(s)
- Ryan T Bishop
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Research Center and Institute, Tampa, FL, USA
| | - Jesus Delgado-Calle
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, Little Rock, AR, USA
| | - Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, Scarborough, ME, USA
| | - Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
6
|
Haghpanah Z, Mondal D, Momenbeitollahi N, Mohsenkhani S, Zarshenas K, Jin Y, Watson M, Willett T, Gorbet M. In vitro evaluation of bone cell response to novel 3D-printable nanocomposite biomaterials for bone reconstruction. J Biomed Mater Res A 2024; 112:1725-1739. [PMID: 38619300 DOI: 10.1002/jbm.a.37719] [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: 11/29/2023] [Revised: 02/24/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024]
Abstract
Critically-sized segmental bone defects represent significant challenges requiring grafts for reconstruction. 3D-printed synthetic bone grafts are viable alternatives to structural allografts if engineered to provide appropriate mechanical performance and osteoblast/osteoclast cell responses. Novel 3D-printable nanocomposites containing acrylated epoxidized soybean oil (AESO) or methacrylated AESO (mAESO), polyethylene glycol diacrylate, and nanohydroxyapatite (nHA) were produced using masked stereolithography. The effects of volume fraction of nHA and methacrylation of AESO on interactions of differentiated MC3T3-E1 osteoblast (dMC3T3-OB) and differentiated RAW264.7 osteoclast cells with 3D-printed nanocomposites were evaluated in vitro and compared with a control biomaterial, hydroxyapatite (HA). Higher nHA content and methacrylation significantly improved the mechanical properties. All nanocomposites supported dMC3T3-OB cells' adhesion and proliferation. Higher amounts of nHA enhanced cell adhesion and proliferation. mAESO in the nanocomposites resulted in greater adhesion, proliferation, and activity at day 7 compared with AESO nanocomposites. Excellent osteoclast-like cells survival, defined actin rings, and large multinucleated cells were only observed on the high nHA fraction (30%) mAESO nanocomposite and the HA control. Thus, mAESO-based nanocomposites containing higher amounts of nHA have better interactions with osteoblast-like and osteoclast-like cells, comparable with HA controls, making them a potential future alternative graft material for bone defect repair.
Collapse
Affiliation(s)
- Zahra Haghpanah
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Dibakar Mondal
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Nikan Momenbeitollahi
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Sadaf Mohsenkhani
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Kiyoumars Zarshenas
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Yutong Jin
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Michael Watson
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Thomas Willett
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Maud Gorbet
- Department of Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
7
|
Babendreyer A, Kieselhorst J, Rinkens C, Lyashenko AM, Düsterhöft S, Jahr H, Craveiro RB, Wolf M, Ludwig A. Downregulation of the metalloproteinases ADAM10 or ADAM17 promotes osteoclast differentiation. Cell Commun Signal 2024; 22:322. [PMID: 38863060 PMCID: PMC11167776 DOI: 10.1186/s12964-024-01690-y] [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: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Bone resorption is driven through osteoclast differentiation by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-Β ligand (RANKL). We noted that a disintegrin and metalloproteinase (ADAM) 10 and ADAM17 are downregulated at the expression level during osteoclast differentiation of the murine monocytic cell line RAW264.7 in response to RANKL. Both proteinases are well known to shed a variety of single-pass transmembrane molecules from the cell surface. We further showed that inhibitors of ADAM10 or ADAM17 promote osteoclastic differentiation and furthermore enhance the surface expression of receptors for RANKL and M-CSF on RAW264.7 cells. Using murine bone marrow-derived monocytic cells (BMDMCs), we demonstrated that a genetic deficiency of ADAM17 or its required regulator iRhom2 leads to increased osteoclast development in response to M-CSF and RANKL stimulation. Moreover, ADAM17-deficient osteoclast precursor cells express increased levels of the receptors for RANKL and M-CSF. Thus, ADAM17 negatively regulates osteoclast differentiation, most likely through shedding of these receptors. To assess the time-dependent contribution of ADAM10, we blocked this proteinase by adding a specific inhibitor on day 0 of BMDMC stimulation with M-CSF or on day 7 of subsequent stimulation with RANKL. Only ADAM10 inhibition beginning on day 7 increased the size of developing osteoclasts indicating that ADAM10 suppresses osteoclast differentiation at a later stage. Finally, we could confirm our findings in human peripheral blood mononuclear cells (PBMCs). Thus, downregulation of either ADAM10 or ADAM17 during osteoclast differentiation may represent a novel regulatory mechanism to enhance their differentiation process. Enhanced bone resorption is a critical issue in osteoporosis and is driven through osteoclast differentiation by specific osteogenic mediators. The present study demonstrated that the metalloproteinases ADAM17 and ADAM10 critically suppress osteoclast development. This was observed for a murine cell line, for isolated murine bone marrow cells and for human blood cells by either preferential inhibition of the proteinases or by gene knockout. As a possible mechanism, we studied the surface expression of critical receptors for osteogenic mediators on developing osteoclasts. Our findings revealed that the suppressive effects of ADAM17 and ADAM10 on osteoclastogenesis can be explained in part by the proteolytic cleavage of surface receptors by ADAM10 and ADAM17, which reduces the sensitivity of these cells to osteogenic mediators. We also observed that osteoclast differentiation was associated with the downregulation of ADAM10 and ADAM17, which reduced their suppressive effects. We therefore propose that this downregulation serves as a feedback loop for enhancing osteoclast development.
Collapse
Affiliation(s)
- Aaron Babendreyer
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Julia Kieselhorst
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Cindy Rinkens
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anastasia M Lyashenko
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Holger Jahr
- Institute of Anatomy and Cell Biology, University Hospital RWTH Aachen, Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, Aachen, Germany
| | - Rogerio B Craveiro
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, University Hospital RWTH Aachen, Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, University Hospital RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
8
|
Wirsig K, Bacova J, Richter RF, Hintze V, Bernhardt A. Cellular response of advanced triple cultures of human osteocytes, osteoblasts and osteoclasts to high sulfated hyaluronan (sHA3). Mater Today Bio 2024; 25:101006. [PMID: 38445011 PMCID: PMC10912908 DOI: 10.1016/j.mtbio.2024.101006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Bone remodelling, important for homeostasis and regeneration involves the controlled action of osteoblasts, osteocytes and osteoclasts. The present study established a three-dimensional human in vitro bone model as triple culture with simultaneously differentiating osteocytes and osteoclasts, in the presence of osteoblasts. Since high sulfated hyaluronan (sHA3) was reported as a biomaterial to enhance osteogenesis as well as to dampen osteoclastogenesis, the triple culture was exposed to sHA3 to investigate cellular responses compared to the respective bone cell monocultures. Osteoclast formation and marker expression was stimulated by sHA3 only in triple culture. Osteoprotegerin (OPG) gene expression and protein secretion, but not receptor activator of NF-κB ligand (RANKL) or sclerostin (SOST), were strongly enhanced, suggesting an important role of sHA3 itself in osteoclastogenesis with other targets than indirect modulation of the RANKL/OPG ratio. Furthermore, sHA3 upregulated osteocalcin (BGLAP) in osteocytes and osteoblasts in triple culture, while alkaline phosphatase (ALP) was downregulated.
Collapse
Affiliation(s)
- Katharina Wirsig
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| | - Jana Bacova
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentska 573, 53210 Pardubice, Czech Republic
| | - Richard F. Richter
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| | - Vera Hintze
- Max Bergmann Center of Biomaterials, Institute of Material Science, TUD University of Technology, Budapester Str. 27, 01069, Dresden, Germany
| | - Anne Bernhardt
- Centre for Translational Bone, Joint- and Soft Tissue Research, Faculty of Medicine and University Hospital, TUD University of Technology, Fetscherstraße 74, 01307, Dresden, Germany
| |
Collapse
|
9
|
Zhang Z, Meng Y, Lin T, Zhang Z, Tao Z, Yin H, Yang F, Zhou X. Dancr-BRG1 regulates Nfatc1 transcription and Pgc1β-dependent metabolic shifts in osteoclastogenesis. Proc Natl Acad Sci U S A 2024; 121:e2313656121. [PMID: 38252822 PMCID: PMC10835043 DOI: 10.1073/pnas.2313656121] [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: 08/09/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Long non-coding RNA (lncRNA) serves as a vital regulator of bone metabolism, but its role in pathologically overactive osteoclast differentiation remains elusive. Here, we identify lncRNA Dancr (Differentiation Antagonizing Non-protein Coding RNA) as a critical suppressor of osteoclastogenesis and bone resorption, which is down-regulated in response to estrogen deficiency. Global or osteoclast-specific Dancr Knockout mice display significant trabecular bone deterioration and enhanced osteoclast activity, but minimal alteration of bone formation. Moreover, the bone-targeted delivery of Dancr by Adeno-associated viral remarkably attenuates ovariectomy-induced osteopenia in mice. Mechanistically, Dancr establishes a direct interaction with Brahma-related gene 1 to prevent its binding and preserve H3K27me3 enrichment at the nuclear factor of activated T cells 1 and proliferator-activated receptor gamma coactivator 1-beta promoters, thereby maintaining appropriate expression of osteoclastic genes and metabolic programs during osteoclastogenesis. These results demonstrate that Dancr is a key molecule maintaining proper osteoclast differentiation and bone homeostasis under physiological conditions, and Dancr overexpression constitutes a potential strategy for treating osteoporosis.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
- Department of Orthopedic rehabilitation, Qingdao Special Servicemen Recuperation Center of People's Liberation Army Navy, Qingdao266000, China
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Tao Lin
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Zhanrong Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Zhengbo Tao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
| | - Haozan Yin
- Department of Medical Genetics, Second Military Medical University (Naval Medical University), Shanghai200433, China
| | - Fu Yang
- Department of Medical Genetics, Second Military Medical University (Naval Medical University), Shanghai200433, China
- Key Laboratory of Biological Defense, Ministry of Education, Shanghai200433, China
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai200003, China
- Translational research center of orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai201600, China
| |
Collapse
|
10
|
Kwack KH, Zhang L, Kirkwood KL. In vitro osteoclastogenesis assessment using murine myeloid-derived suppressor cells. Methods Cell Biol 2023; 184:133-147. [PMID: 38555153 PMCID: PMC11287231 DOI: 10.1016/bs.mcb.2023.04.007] [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] [Indexed: 04/02/2024]
Abstract
The study of myeloid-derived suppressor cells (MDSCs) has been commonly reported in the context of cancer immunology. MDSCs play a key role in cancer growth and progression by inhibiting both innate and adaptive immunity. In addition to the immunosuppressive function of MDSCs in cancer, a novel function of MDSCs as osteoclast precursors has recently been attracting attention. Because monocytic-MDSCs (M-MDSCs) are derived from the same myeloid lineage as macrophages, which are osteoclast progenitors, M-MDSCs can undergo differentiation into osteoclasts, contributing to bone destruction not only in the cancer microenvironment but also in inflammatory conditions including obesity and osteoarthritis. Herein, we present details of the technique to evaluate osteoclasts in vitro, as well as specific techniques to isolate M-MDSCs and identify them. This protocol can be easily adapted to isolate M-MDSCs from most pathologic conditions for easy evaluation.
Collapse
Affiliation(s)
- Kyu Hwan Kwack
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States; Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea.
| | - Lixia Zhang
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
| | - Keith L Kirkwood
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States; Department of Head & Neck/Plastic & Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
11
|
Babu B, Pawar S, Mittal A, Kolanthai E, Neal CJ, Coathup M, Seal S. Nanotechnology enabled radioprotectants to reduce space radiation-induced reactive oxidative species. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1896. [PMID: 37190884 DOI: 10.1002/wnan.1896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023]
Abstract
Interest in space exploration has seen substantial growth following recent launch and operation of modern space technologies. In particular, the possibility of travel beyond low earth orbit is seeing sustained support. However, future deep space travel requires addressing health concerns for crews under continuous, longer-term exposure to adverse environmental conditions. Among these challenges, radiation-induced health issues are a major concern. Their potential to induce chronic illness is further potentiated by the microgravity environment. While investigations into the physiological effects of space radiation are still under investigation, studies on model ionizing radiation conditions, in earth and micro-gravity conditions, can provide needed insight into relevant processes. Substantial formation of high, sustained reactive oxygen species (ROS) evolution during radiation exposure is a clear threat to physiological health of space travelers, producing indirect damage to various cell structures and requiring therapeutic address. Radioprotection toward the skeletal system components is essential to astronaut health, due to the high radio-absorption cross-section of bone mineral and local hematopoiesis. Nanotechnology can potentially function as radioprotectant and radiomitigating agents toward ROS and direct radiation damage. Nanoparticle compositions such as gold, silver, platinum, carbon-based materials, silica, transition metal dichalcogenides, and ceria have all shown potential as viable radioprotectants to mitigate space radiation effects with nanoceria further showing the ability to protect genetic material from oxidative damage in several studies. As research into space radiation-induced health problems develops, this review intends to provide insights into the nanomaterial design to ameliorate pathological effects from ionizing radiation exposure. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Nanotechnology Approaches to Biology > Cells at the Nanoscale Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Craig J Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Melanie Coathup
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- College of Medicine, Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
12
|
Shimizu K, Kikuta J, Ohta Y, Uchida Y, Miyamoto Y, Morimoto A, Yari S, Sato T, Kamakura T, Oshima K, Imai R, Liu YC, Okuzaki D, Hara T, Motooka D, Emoto N, Inohara H, Ishii M. Single-cell transcriptomics of human cholesteatoma identifies an activin A-producing osteoclastogenic fibroblast subset inducing bone destruction. Nat Commun 2023; 14:4417. [PMID: 37537159 PMCID: PMC10400591 DOI: 10.1038/s41467-023-40094-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 07/12/2023] [Indexed: 08/05/2023] Open
Abstract
Cholesteatoma, which potentially results from tympanic membrane retraction, is characterized by intractable local bone erosion and subsequent hearing loss and brain abscess formation. However, the pathophysiological mechanisms underlying bone destruction remain elusive. Here, we performed a single-cell RNA sequencing analysis on human cholesteatoma samples and identify a pathogenic fibroblast subset characterized by abundant expression of inhibin βA. We demonstrate that activin A, a homodimer of inhibin βA, promotes osteoclast differentiation. Furthermore, the deletion of inhibin βA /activin A in these fibroblasts results in decreased osteoclast differentiation in a murine model of cholesteatoma. Moreover, follistatin, an antagonist of activin A, reduces osteoclastogenesis and resultant bone erosion in cholesteatoma. Collectively, these findings indicate that unique activin A-producing fibroblasts present in human cholesteatoma tissues are accountable for bone destruction via the induction of local osteoclastogenesis, suggesting a potential therapeutic target.
Collapse
Affiliation(s)
- Kotaro Shimizu
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan.
| | - Yumi Ohta
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yutaka Uchida
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yu Miyamoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Akito Morimoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shinya Yari
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Takashi Sato
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Takefumi Kamakura
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kazuo Oshima
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Ryusuke Imai
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yu-Chen Liu
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tetsuya Hara
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, 658-8558, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, Higashinada, Kobe, 658-8558, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan.
| |
Collapse
|
13
|
Amadeu de Oliveira F, Tokuhara CK, Veeriah V, Domezi JP, Santesso MR, Cestari TM, Ventura TMO, Matos AA, Dionísio T, Ferreira MR, Ortiz RC, Duarte MAH, Buzalaf MAR, Ponce JB, Sorgi CA, Faccioli LH, Buzalaf CP, de Oliveira RC. The Multifarious Functions of Leukotrienes in Bone Metabolism. J Bone Miner Res 2023; 38:1135-1153. [PMID: 37314430 DOI: 10.1002/jbmr.4867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023]
Abstract
Leukotrienes (LTs) are derived from arachidonic acid metabolism by the 5-lipoxygenase (5-LO) enzyme. The production of LTs is stimulated in the pathogenesis of rheumatoid arthritis (RA), osteoarthritis, and periodontitis, with a relevant contribution to bone resorption. However, its role in bone turnover, particularly the suppression of bone formation by modulating the function of osteoclasts and osteoblasts, remains unclear. We investigated the effects of LTs on bone metabolism and their impact on osteogenic differentiation and osteoclastogenesis using a 5-LO knockout (KO) mouse model. Results from micro-computed tomography (μCT) analysis of femur from 8-week-old 5-LO-deficient mice showed increased cortical bone and medullary region in females and males and decreased trabecular bone in females. In the vertebra, we observed increased marrow area in both females and males 5-LO KO and decreased trabecular bone only in females 5-LO KO. Immunohistochemistry (IHC) analysis showed higher levels of osteogenic markers tissue-nonspecific alkaline phosphatase (TNAP) and osteopontin (OPN) and lower expression of osteoclastogenic marker tartrate-resistant acid phosphatase (TRAP) in the femurs of 5-LO KO mice versus wild-type (WT). Alkaline phosphatase activity and mineralization assay results showed that the 5-LO absence enhances osteoblasts differentiation and mineralization but decreases the proliferation. Alkaline phosphatase (ALP), Bglap, and Sp7 gene expression were higher in 5-LO KO osteoblasts compared to WT cells. Eicosanoids production was higher in 5-LO KO osteoblasts except for thromboxane 2, which was lower in 5-LO-deficient mice. Proteomic analysis identified the downregulation of proteins related to adenosine triphosphate (ATP) metabolism in 5-LO KO osteoblasts, and the upregulation of transcription factors such as the adaptor-related protein complex 1 (AP-1 complex) in long bones from 5-LO KO mice leading to an increased bone formation pattern in 5-LO-deficient mice. We observed enormous differences in the morphology and function of osteoclasts with reduced bone resorption markers and impaired osteoclasts in 5-LO KO compared to WT osteoclasts. Altogether, these results demonstrate that the absence of 5-LO is related to the greater osteogenic profile. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Flávia Amadeu de Oliveira
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- Human Genetics Program, Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Cintia K Tokuhara
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- Human Genetics Program, Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Vimal Veeriah
- Institute for Regenerative Medicine, University of Zürich, Zürich, Switzerland
| | - João Paulo Domezi
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | | | - Tania M Cestari
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | | | - Adriana A Matos
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
- Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago Dionísio
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Marcel R Ferreira
- Institute of Biosciences, São Paulo State University-UNESP, Botucatu, SP, Brazil
| | - Rafael C Ortiz
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Marco A H Duarte
- Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | | | - José B Ponce
- Department of Medicine, University Center of Adamantina, Adamantina, SP, Brazil
- Department of Medicine, Faculdades de Dracena, Dracena, SP, Brazil
| | - Carlos A Sorgi
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Lucia H Faccioli
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | |
Collapse
|
14
|
Fung SL, Cohen JP, Pashuck ET, Miles CE, Freeman JW, Kohn J. Rational design of poly(peptide-ester) block copolymers for enzyme-specific surface resorption. J Mater Chem B 2023; 11:6621-6633. [PMID: 37358375 PMCID: PMC10519181 DOI: 10.1039/d3tb00265a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Tissue resorption and remodeling are pivotal steps in successful healing and regeneration, and it is important to design biomaterials that are responsive to regenerative processes in native tissue. The cell types responsible for remodeling, such as macrophages in the soft tissue wound environment and osteoclasts in the bone environment, utilize a class of enzymes called proteases to degrade the organic matrix. Many hydrophobic thermoplastics used in tissue regeneration are designed to degrade and resorb passively through hydrolytic mechanisms, leaving the potential of proteolytic-guided degradation underutilized. Here, we report the design and synthesis of a tyrosol-derived peptide-polyester block copolymer where protease-mediated resorption is tuned through changing the chemistry of the base polymer backbone and protease specificity is imparted through incorporation of specific peptide sequences. Quartz crystal microbalance was used to quantify polymer surface resorption upon exposure to various enzymes. Aqueous solubility of the diacids and the thermal properties of the resulting polymer had a significant effect on enzyme-mediated polymer resorption. While peptide incorporation at 2 mol% had little effect on the final thermal and physical properties of the block copolymers, its incorporation improved polymer resorption significantly in a peptide sequence- and protease-specific manner. To our knowledge, this is the first example of a peptide-incorporated linear thermoplastic with protease-specific sensitivity reported in the literature. The product is a modular system for engineering specificity in how polyesters can resorb under physiological conditions, thus providing a potential framework for improving vascularization and integration of biomaterials used in tissue engineering.
Collapse
Affiliation(s)
- Stephanie L Fung
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Jarrod P Cohen
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - E Thomas Pashuck
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Department of Bioengineering, Lehigh University, Bethlehem, PA, 18018, USA
| | - Catherine E Miles
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Joachim Kohn
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
15
|
Anginot A, Nguyen J, Abou Nader Z, Rondeau V, Bonaud A, Kalogeraki M, Boutin A, Lemos JP, Bisio V, Koenen J, Hanna Doumit Sakr L, Picart A, Coudert A, Provot S, Dulphy N, Aurrand-Lions M, Mancini SJC, Lazennec G, McDermott DH, Guidez F, Blin-Wakkach C, Murphy PM, Cohen-Solal M, Espéli M, Rouleau M, Balabanian K. WHIM Syndrome-linked CXCR4 mutations drive osteoporosis. Nat Commun 2023; 14:2058. [PMID: 37045841 PMCID: PMC10097661 DOI: 10.1038/s41467-023-37791-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/07/2023] [Indexed: 04/14/2023] Open
Abstract
WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.
Collapse
Affiliation(s)
- Adrienne Anginot
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julie Nguyen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Zeina Abou Nader
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Vincent Rondeau
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Amélie Bonaud
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Maria Kalogeraki
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Julia P Lemos
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Valeria Bisio
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Joyce Koenen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Lea Hanna Doumit Sakr
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amandine Picart
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amélie Coudert
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Sylvain Provot
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Nicolas Dulphy
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Michel Aurrand-Lions
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane J C Mancini
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gwendal Lazennec
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- CNRS, SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France
| | - David H McDermott
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Fabien Guidez
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1131, Paris, France
| | | | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Martine Cohen-Solal
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Marion Espéli
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Karl Balabanian
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France.
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France.
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France.
| |
Collapse
|
16
|
Towards using 3D cellular cultures to model the activation and diverse functions of macrophages. Biochem Soc Trans 2023; 51:387-401. [PMID: 36744644 PMCID: PMC9987999 DOI: 10.1042/bst20221008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
The advent of 3D cell culture technology promises to enhance understanding of cell biology within tissue microenvironments. Whilst traditional cell culturing methods have been a reliable tool for decades, they inadequately portray the complex environments in which cells inhabit in vivo. The need for better disease models has pushed the development of effective 3D cell models, providing more accurate drug screening assays. There has been great progress in developing 3D tissue models in fields such as cancer research and regenerative medicine, driven by desires to recreate the tumour microenvironment for the discovery of new chemotherapies, or development of artificial tissues or scaffolds for transplantation. Immunology is one field that lacks optimised 3D models and the biology of tissue resident immune cells such as macrophages has yet to be fully explored. This review aims to highlight the benefits of 3D cell culturing for greater understanding of macrophage biology. We review current knowledge of macrophage interactions with their tissue microenvironment and highlight the potential of 3D macrophage models in the development of more effective treatments for disease.
Collapse
|
17
|
Meng B, Yang B, Qu Y, Liu Y, Wu D, Fu C, He Y, Chen X, Liu C, Kou X, Cao Y. Dual Role of Interleukin-20 in Different Stages of Osteoclast Differentiation and Its Osteoimmune Regulation during Alveolar Bone Remodeling. Int J Mol Sci 2023; 24:ijms24043810. [PMID: 36835229 PMCID: PMC9961846 DOI: 10.3390/ijms24043810] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
Osteoimmunology mediators are critical to balance osteoblastogenesis and osteoclastogenesis to maintain bone homeostasis. A lot of the osteoimmunology mediators are regulated by interleukin-20 (IL-20). However, little is known about the role of IL-20 in bone remodeling. Here, we showed that IL-20 expression was correlated with osteoclast (OC) activity in remodeled alveolar bone during orthodontic tooth movement (OTM). Ovariectomize (OVX) in rats promoted OC activity and enhanced IL-20 expression, while blocking OC inhibited IL-20 expression in osteoclasts. In vitro, IL-20 treatment promoted survival, inhibited apoptosis of the preosteoclast at the early stages of osteoclast differentiation, and boosted the formation of osteoclasts and their bone resorption function at the late stages. More importantly, anti-IL-20 antibody treatment blocked IL-20-induced osteoclastogenesis and the subsequent bone resorption function. Mechanistically, we showed that IL-20 synergistically acts with RANKL to activate the NF-κB signaling pathway to promote the expression of c-Fos and NFATc1 to promote osteoclastogenesis. Moreover, we found that local injection of IL-20 or anti-IL-20 antibody enhanced osteoclast activity and accelerated OTM in rats, while blocking IL-20 reversed this phenomenon. This study revealed a previously unknown role of IL-20 in regulating alveolar bone remodeling and implies the application of IL-20 to accelerated OTM.
Collapse
Affiliation(s)
- Bowen Meng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Benyi Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yan Qu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yuanbo Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Dongle Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Chaoran Fu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Yifan He
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Xi Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Chufeng Liu
- Department of Orthodontics, Stomatological Hospital, Southern Medical University, Guangzhou 510260, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Correspondence: (X.K.); (Y.C.)
| | - Yang Cao
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- South China Center of Craniofacial Stem Cell Research, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Correspondence: (X.K.); (Y.C.)
| |
Collapse
|
18
|
Remmers SJ, van der Heijden FC, de Wildt BW, Ito K, Hofmann S. Tuning the resorption-formation balance in an in vitro 3D osteoblast-osteoclast co-culture model of bone. Bone Rep 2022; 18:101646. [PMID: 36578830 PMCID: PMC9791323 DOI: 10.1016/j.bonr.2022.101646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to further improve an in vitro 3D osteoblast (OB) - osteoclast (OC) co-culture model of bone by tuning it towards states of formation, resorption, and equilibrium for their future applications in fundamental research, drug development and personalized medicine. This was achieved by varying culture medium composition and monocyte seeding density, the two external parameters that affect cell behavior the most. Monocytes were seeded at two seeding densities onto 3D silk-fibroin constructs pre-mineralized by MSC-derived OBs and were co-cultured in one of three different media (OC stimulating, Neutral and OB stimulating medium) for three weeks. Histology showed mineralized matrix after co-culture and OC markers in the OC medium group. Scanning Electron Microscopy showed large OC-like cells in the OC medium group. Micro-computed tomography showed increased formation in the OB medium group, equilibrium in the Neutral medium group and resorption in the OC medium group. Culture supernatant samples showed high early tartrate resistant acid phosphatase (TRAP) release in the OC medium group, a later and lower release in the Neutral medium group, and almost no release in the OB medium group. Increased monocyte seeding density showed a less-than-proportional increase in TRAP release and resorption in OC medium, while it proportionally increased TRAP release in Neutral medium without affecting net resorption. The 3D OB-OC co-culture model was effectively used to show an excess of mineral deposition using OB medium, resorption using OC medium, or an equilibrium using Neutral medium. All three media applied to the model may have their own distinct applications in fundamental research, drug development, and personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | - Sandra Hofmann
- Corresponding author at: Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, the Netherlands.
| |
Collapse
|
19
|
A New Method to Sort Differentiating Osteoclasts into Defined Homogeneous Subgroups. Cells 2022; 11:cells11243973. [PMID: 36552735 PMCID: PMC9777285 DOI: 10.3390/cells11243973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts regulate skeletal development but also drive pathological osteolysis, making them prime therapeutic targets. Osteoclast research is limited by the heterogeneity of osteoclast populations generated in vitro, where the mixture of undifferentiated monocytes, binuclear pre-osteoclasts and multinucleated osteoclasts has by necessity been considered a single osteoclast population. This study describes the differentiation of primary human CD14+ monocyte-derived osteoclasts in 3D collagen gels. These osteoclasts remained small (>95% with ≤5 nuclei) but were viable and active; when released from the gel with collagenase, they fused rapidly when reseeded onto solid substrates and resorbed dentine for 2-3 weeks. 3D-generated osteoclasts expressed cell surface markers of osteoclast differentiation (e.g., CD9, RANK, OSCAR, CD63, CD51/61) which, with their small size, enabled live cell sorting of highly enriched viable subpopulations of human osteoclasts that retained full functional resorption capacity. Low-yield osteoclast preparations were strongly enriched to remove undifferentiated cells (e.g., 13.3% CD51/61+ to 84.2% CD51/61+), and subpopulations of CD9+CD51/61- early osteoclasts and CD9+CD51/61+ mature cells were distinguished. This novel approach allows the study of selected populations of differentiating osteoclasts in vitro and opens the door to in-depth transcriptomic and proteomic analysis of these cells, increasing our ability to study human osteoclast molecular mechanisms relevant to development, aging and disease.
Collapse
|
20
|
Zou L, Jiang W, Wang Z, Zhu S, Chen J. Effect of Advanced Oxidation Protein Products (AOPPs) and aging on the osteoclast differentiation of Myeloid-Derived Suppressor Cells (MDSCs) and its preliminary mechanism. Biochem Biophys Res Commun 2022; 636:87-96. [DOI: 10.1016/j.bbrc.2022.10.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 11/28/2022]
|
21
|
Kohtala S, Nedal TMV, Kriesi C, Moen SH, Ma Q, Ødegaard KS, Standal T, Steinert M. Automated Quantification of Human Osteoclasts Using Object Detection. Front Cell Dev Biol 2022; 10:941542. [PMID: 35865628 PMCID: PMC9294346 DOI: 10.3389/fcell.2022.941542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022] Open
Abstract
A balanced skeletal remodeling process is paramount to staying healthy. The remodeling process can be studied by analyzing osteoclasts differentiated in vitro from mononuclear cells isolated from peripheral blood or from buffy coats. Osteoclasts are highly specialized, multinucleated cells that break down bone tissue. Identifying and correctly quantifying osteoclasts in culture are usually done by trained personnel using light microscopy, which is time-consuming and susceptible to operator biases. Using machine learning with 307 different well images from seven human PBMC donors containing a total of 94,974 marked osteoclasts, we present an efficient and reliable method to quantify human osteoclasts from microscopic images. An open-source, deep learning-based object detection framework called Darknet (YOLOv4) was used to train and test several models to analyze the applicability and generalizability of the proposed method. The trained model achieved a mean average precision of 85.26% with a correlation coefficient of 0.99 with human annotators on an independent test set and counted on average 2.1% more osteoclasts per culture than the humans. Additionally, the trained models agreed more than two independent human annotators, supporting a more reliable and less biased approach to quantifying osteoclasts while saving time and resources. We invite interested researchers to test their datasets on our models to further strengthen and validate the results.
Collapse
Affiliation(s)
- Sampsa Kohtala
- TrollLABS, Department of Mechanical and Industrial Engineering, Faculty of Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- *Correspondence: Sampsa Kohtala,
| | - Tonje Marie Vikene Nedal
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Carlo Kriesi
- TrollLABS, Department of Mechanical and Industrial Engineering, Faculty of Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Vitroscope AS, Trondheim, Norway
| | - Siv Helen Moen
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Qianli Ma
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kristin Sirnes Ødegaard
- Department of Mechanical and Industrial Engineering, Faculty of Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Therese Standal
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Hematology, St. Olavs University Hospital, Trondheim, Norway
| | - Martin Steinert
- TrollLABS, Department of Mechanical and Industrial Engineering, Faculty of Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
22
|
Neto E, Monteiro AC, Leite Pereira C, Simões M, Conde JP, Chu V, Sarmento B, Lamghari M. Micropathological Chip Modeling the Neurovascular Unit Response to Inflammatory Bone Condition. Adv Healthc Mater 2022; 11:e2102305. [PMID: 35158409 PMCID: PMC11468530 DOI: 10.1002/adhm.202102305] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Organ-on-a-chip in vitro platforms accurately mimic complex microenvironments offering the ability to recapitulate and dissect mechanisms of physiological and pathological settings, revealing their major importance to develop new therapeutic targets. Bone diseases, such as osteoarthritis, are extremely complex, comprising of the action of inflammatory mediators leading to unbalanced bone homeostasis and de-regulation of sensory innervation and angiogenesis. Although there are models to mimic bone vascularization or innervation, in vitro platforms merging the complexity of bone, vasculature, innervation, and inflammation are missing. Therefore, in this study a microfluidic-based neuro-vascularized bone chip (NVB chip) is proposed to 1) model the mechanistic interactions between innervation and angiogenesis in the inflammatory bone niche, and 2) explore, as a screening tool, novel strategies targeting inflammatory diseases, using a nano-based drug delivery system. It is possible to set the design of the platform and achieve the optimized conditions to address the neurovascular network under inflammation. Moreover, this system is validated by delivering anti-inflammatory drug-loaded nanoparticles to counteract the neuronal growth associated with pain perception. This reliable in vitro tool will allow understanding the bone neurovascular system, enlightening novel mechanisms behind the inflammatory bone diseases, bone destruction, and pain opening new avenues for new therapies discovery.
Collapse
Affiliation(s)
- Estrela Neto
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Ana Carolina Monteiro
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Catarina Leite Pereira
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Miguel Simões
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Bruno Sarmento
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- CESPUInstituto de Investigação e Formação Avançada em Ciências e Tecnologias da SaúdeRua Central da Gandra, 137Gandra4585‐116Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| |
Collapse
|
23
|
Zhang Y, Polman M, Mohammad AF, Hermens I, Zhuang Z, Wang H, van den Beucken JJJP. Species-independent stimulation of osteogenic differentiation induced by osteoclasts. Biochem Biophys Res Commun 2022; 606:149-155. [DOI: 10.1016/j.bbrc.2022.03.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/26/2022]
|
24
|
Cui Y, Wang F, Zhang D, Huang J, Yang Y, Xu J, Gao Y, Ding H, Qu Y, Zhang W, Liu W, Pan L, Zhang L, Liu Z, Niu T, Liu T, Zheng Y. Estrogen-Responsive Gene MAST4 Regulates Myeloma Bone Disease. J Bone Miner Res 2022; 37:711-723. [PMID: 35064934 DOI: 10.1002/jbmr.4507] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 02/05/2023]
Abstract
Our previous data showed that young female multiple myeloma (MM) patients had a low frequency of osteolytic lesions. Based on this clinical observation, we found that estrogen cell signaling played a regulatory role in MM bone disease (MMBD), and the estrogen-responsive gene microtubule-associated serine/threonine kinase family member 4 (MAST4) was a critical factor. The presence of estrogen in cell cultures promoted MAST4 expression in MM cells, while knocking down estrogen receptor 1 (ESR1) inhibited MAST4 expression. Chromatin immunoprecipitation assay suggested a binding site of ESR1 on the MAST4 promoter. Bisphosphonates, such as zoledronic acid (ZOL), which was widely used in MMBD control, could stimulate MAST4 expression in MM cells by promoting ESR1 expression. MAST4 interacted with phosphatase and tensin homolog (PTEN), therefore regulating the PI3K-Akt-mTOR pathway and the expression of downstream cytokines, such as CCL2/3/4. MAST4 knockdown (MAST4-KD) or ESR1 knockdown (ESR1-KD) MM cells had repressed PTEN activity, elevated PI3K-Akt-mTOR activity, and increased CCL2/3/4 expressions. Coculture of MAST4-KD or ESR1-KD MM cells with pre-osteoclasts (pre-OCs) stimulated OC formation in vitro, whereas neutralizing antibodies of CCL2/3/4 attenuated such stimulation. In mouse models, mice inoculated with MAST4-KD or ESR1-KD MM cells had severer MMBD than control knockdown (CTR-KD). The correlations between MAST4 and ESR1 expressions in MMBD, as well as related cell signaling pathways, were confirmed in analyses using gene expression profiles (GEPs) of patients' MM cells. The negative correlation of MAST4 expression and occurrence of MMBD was further validated by patients' immunohistochemical tissue array. Overall, our data suggested that estrogen cell signaling negatively regulated MMBD through MAST4. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yushan Cui
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Danfeng Zhang
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
- Department of Hematology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Yan Yang
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Juan Xu
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Yuhan Gao
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Hong Ding
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Ying Qu
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Wenyan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Pan
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Zhigang Liu
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Ting Liu
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital/State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Liu X, Chen M, Liu Q, Li G, Yang P, Zhang G. LncRNA PTCSC3 is upregulated in osteoporosis and negatively regulates osteoblast apoptosis. BMC Med Genomics 2022; 15:57. [PMID: 35296322 PMCID: PMC8925152 DOI: 10.1186/s12920-022-01182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND It is known that long non-coding RNA (lncRNA) PTCSC3 is involved in thyroid cancer and glioma, but its function in osteoporosis is unknown. The aim of our study was to investigate the role of lncRNA PTCSC3 in osteoporosis. METHODS A total of 80 patients with osteoporosis (4 clinical stages) and four corresponding groups of healthy controls were enrolled. Plasma PTCSC3 levels in the 80 osteoporosis patients and 80 healthy volunteers were measured using RT-qPCR. The diagnostic potential of plasma PTCSC3 for osteoporosis was evaluated by ROC curve analysis with healthy volunteers as the true negative cases and corresponding osteoporosis patients as the true positive cases. RESULTS PTCSC3 was upregulated in osteoporosis patients compared with healthy controls. PTCSC3 levels increased with osteoporosis stages increasing, but not with healthy controls aging. PTCSC3 overexpression separated each stage of osteoporosis from corresponding controls. PTCSC3 overexpression promoted while PTCSC3 silencing inhibited osteoblast apoptosis. However, PTCSC3 overexpression and silencing showed no significant effect on osteoclast apoptosis. LncRNA PTCSC3 was upregulated in osteoporosis and negatively regulated osteoblast apoptosis. CONCLUSION LncRNA PTCSC3 may serve as a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Xingchao Liu
- Orthopedics Department, Hebei Yanda Hospital, Yanjiao Development Zone, Sanhe City, Langfang City, 065201, Hebei Province, People's Republic of China
| | - Mingliang Chen
- Orthopedics Department, Hebei Yanda Hospital, Yanjiao Development Zone, Sanhe City, Langfang City, 065201, Hebei Province, People's Republic of China
| | - Qinghe Liu
- Orthopedics Department, Beijing Chao-Yang Hospital, Capital Medical University, No 8 Gongtinan Road, Beijing, 100020, People's Republic of China.
| | - Gang Li
- Orthopedics Department, Hebei Yanda Hospital, Yanjiao Development Zone, Sanhe City, Langfang City, 065201, Hebei Province, People's Republic of China
| | - Pei Yang
- Orthopedics Department, Hebei Yanda Hospital, Yanjiao Development Zone, Sanhe City, Langfang City, 065201, Hebei Province, People's Republic of China
| | - Guodong Zhang
- Orthopedics Department, Hebei Yanda Hospital, Yanjiao Development Zone, Sanhe City, Langfang City, 065201, Hebei Province, People's Republic of China
| |
Collapse
|
26
|
Tang CY, Wang H, Zhang Y, Wang Z, Zhu G, McVicar A, Li YP, Chen W. GPR125 positively regulates osteoclastogenesis potentially through AKT-NF-κB and MAPK signaling pathways. Int J Biol Sci 2022; 18:2392-2405. [PMID: 35414778 PMCID: PMC8990458 DOI: 10.7150/ijbs.70620] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/17/2022] [Indexed: 01/26/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) signaling is critical to cell differentiation and activation. However, the function of GPCRs in osteoclast differentiation and activation remains unclear. We found that the G-protein coupled receptor 125 (GPCR 125) gene (Gpr125) gene was highly expressed in osteoclasts through RNA-sequencing technology, qRT-PCR, and Western blot analysis. We characterized the role of GPCR125 in osteoclast differentiation and activation by loss-of-function and gain-of-function methods in osteoclasts. Osteoclasts with lentivirus-mediated GPR125 silencing demonstrated a dramatic reduction in differentiation and impaired bone resorption function. In contrast, overexpression of Gpr125 in osteoclasts increased NFATC1 expression and enhanced osteoclast differentiation and enhanced osteoclast-mediated bone resorption. These results indicated that GPCR125 positively regulates osteoclast formation and function. Following receptor activator of nuclear factor kappa-Β ligand (RANKL) stimulation, the expression levels of MAPK signaling pathway proteins phosphorylated-ERK (p-ERK) and phosphorylated-p38 (p-p38) were significantly decreased in the Gpr125 knockdown (sh-GPR125) group compared to its control group. We also found that phosphorylated AKT (p-AKT) expression was downregulated, as well as nuclear factor kappa-B (NF-κB) signaling pathway protein phosphorylated-IKB alpha (p-IKBα). Our results demonstrated that GPCR125 positively regulates osteoclasts via RANKL-stimulated MAPK and AKT-NF-κB signaling pathways, and GPCR125 could potentially be utilized as a novel therapeutic target in bone related diseases including osteoporosis.
Collapse
Affiliation(s)
- Chen-Yi Tang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - He Wang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhongliang Wang
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
27
|
Dang L, Li N, Wu X, Li D, Zhang Z, Zhang BT, Lyu A, Chen L, Zhang G, Liu J. A Rapid Protocol for Direct Isolation of Osteoclast Lineage Cells from Mouse Bone Marrow. Bio Protoc 2022; 12:e4338. [PMID: 35592608 PMCID: PMC8918216 DOI: 10.21769/bioprotoc.4338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/30/2021] [Accepted: 01/12/2022] [Indexed: 12/29/2022] Open
Abstract
Osteoclast lineage cells (OLCs), including osteoclast precursors (OCPs) and mature osteoclasts (MOCs), participate in bone remodeling and mediate pathologic bone loss. Thus, it is essential to obtain OLCs for exploring their molecular features in both physiological and pathological conditions in vivo. However, the conventional protocols for obtaining OLCs ex vivo are not only time-consuming, but also unable to capture the cellular status of OLCs in vivo. In addition, the current antibody-based isolation approaches, such as fluorescence-/ magnetic-activated cell sorting, are not able to obtain pure osteoclasts because no unique surface antigen for osteoclasts has been identified. Here, we develop a rapid protocol for directly isolating OLCs from mouse bone marrow through magnetic-activated cell sorting (MACS). This protocol can rapidly enrich OCPs and MOCs, respectively, depending on the expression of the distinctive surface markers at their differentiation stages. It is optimized to isolate OLCs from four mice concurrently, of which sorting procedure could be completed within ~5 h.
Collapse
Affiliation(s)
- Lei Dang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Nanxi Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiaohao Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Dijie Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bao-Ting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Aiping Lyu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jin Liu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
28
|
Vitale M, Ligorio C, McAvan B, Hodson NW, Allan C, Richardson SM, Hoyland JA, Bella J. Hydroxyapatite-decorated Fmoc-hydrogel as a bone-mimicking substrate for osteoclast differentiation and culture. Acta Biomater 2022; 138:144-154. [PMID: 34781025 PMCID: PMC8756142 DOI: 10.1016/j.actbio.2021.11.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/19/2021] [Accepted: 11/09/2021] [Indexed: 12/25/2022]
Abstract
Hydrogels are water-swollen networks with great potential for tissue engineering applications. However, their use in bone regeneration is often hampered due to a lack of materials' mineralization and poor mechanical properties. Moreover, most studies are focused on osteoblasts (OBs) for bone formation, while osteoclasts (OCs), cells involved in bone resorption, are often overlooked. Yet, the role of OCs is pivotal for bone homeostasis and aberrant OC activity has been reported in several pathological diseases, such as osteoporosis and bone cancer. For these reasons, the aim of this work is to develop customised, reinforced hydrogels to be used as material platform to study cell function, cell-material interactions and ultimately to provide a substrate for OC differentiation and culture. Here, Fmoc-based RGD-functionalised peptide hydrogels have been modified with hydroxyapatite nanopowder (Hap) as nanofiller, to create nanocomposite hydrogels. Atomic force microscopy showed that Hap nanoparticles decorate the peptide nanofibres with a repeating pattern, resulting in stiffer hydrogels with improved mechanical properties compared to Hap- and RGD-free controls. Furthermore, these nanocomposites supported adhesion of Raw 264.7 macrophages and their differentiation in 2D to mature OCs, as defined by the adoption of a typical OC morphology (presence of an actin ring, multinucleation, and ruffled plasma membrane). Finally, after 7 days of culture OCs showed an increased expression of TRAP, a typical OC differentiation marker. Collectively, the results suggest that the Hap/Fmoc-RGD hydrogel has a potential for bone tissue engineering, as a 2D model to study impairment or upregulation of OC differentiation. STATEMENT OF SIGNIFICANCE: Altered osteoclasts (OC) function is one of the major cause of bone fracture in the most commonly skeletal disorders (e.g. osteoporosis). Peptide hydrogels can be used as a platform to mimic the bone microenvironment and provide a tool to assess OC differentiation and function. Moreover, hydrogels can incorporate different nanofillers to yield hybrid biomaterials with enhanced mechanical properties and improved cytocompatibility. Herein, Fmoc-based RGD-functionalised peptide hydrogels were decorated with hydroxyapatite (Hap) nanoparticles to generate a hydrogel with improved rheological properties. Furthermore, they are able to support osteoclastogenesis of Raw264.7 cells in vitro as confirmed by morphology changes and expression of OC-markers. Therefore, this Hap-decorated hydrogel can be used as a template to successfully differentiate OC and potentially study OC dysfunction.
Collapse
Affiliation(s)
- Mattia Vitale
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Cosimo Ligorio
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Bethan McAvan
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom
| | - Nigel W Hodson
- BioAFM Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Chris Allan
- Biogelx Ltd-BioCity Scotland, Bo'Ness Rd, Newhouse, Chapelhall, Motherwell ML1 5UH, United Kingdom
| | - Stephen M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom.
| | - Jordi Bella
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, United Kingdom.
| |
Collapse
|
29
|
The Non-Erythropoietic EPO Analogue Cibinetide Inhibits Osteoclastogenesis In Vitro and Increases Bone Mineral Density in Mice. Int J Mol Sci 2021; 23:ijms23010055. [PMID: 35008482 PMCID: PMC8744753 DOI: 10.3390/ijms23010055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 01/21/2023] Open
Abstract
The two erythropoietin (EPO) receptor forms mediate different cellular responses to erythropoietin. While hematopoiesis is mediated via the homodimeric EPO receptor (EPOR), tissue protection is conferred via a heteromer composed of EPOR and CD131. In the skeletal system, EPO stimulates osteoclast precursors and induces bone loss. However, the underlying molecular mechanisms are still elusive. Here, we evaluated the role of the heteromeric complex in bone metabolism in vivo and in vitro by using Cibinetide (CIB), a non-erythropoietic EPO analogue that exclusively binds the heteromeric receptor. CIB is administered either alone or in combination with EPO. One month of CIB treatment significantly increased the cortical (~5.8%) and trabecular (~5.2%) bone mineral density in C57BL/6J WT female mice. Similarly, administration of CIB for five consecutive days to female mice that concurrently received EPO on days one and four, reduced the number of osteoclast progenitors, defined by flow cytometry as Lin−CD11b−Ly6Chi CD115+, by 42.8% compared to treatment with EPO alone. In addition, CIB alone or in combination with EPO inhibited osteoclastogenesis in vitro. Our findings introduce CIB either as a stand-alone treatment, or in combination with EPO, as an appealing candidate for the treatment of the bone loss that accompanies EPO treatment.
Collapse
|
30
|
Non-polar lipid from greenshell mussel (Perna canaliculus) inhibits osteoclast differentiation. Bone Rep 2021; 15:101132. [PMID: 34632003 PMCID: PMC8493498 DOI: 10.1016/j.bonr.2021.101132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 11/23/2022] Open
Abstract
The osteoclast-dependent bone resorption process is a crucial part of the bone regulatory system. The excessive function of osteoclasts can cause diseases of bone, joint, and other tissues such as osteoporosis and osteoarthritis. Greenshell mussel oil (GSM), a good source of long chain omega-3 polyunsaturated fatty acids (LCn-3PUFAs), was fractionated into total lipid, polar lipid, and non-polar lipid components and their anti-osteoclastogenic activity tested in RAW 264.7 cell cultures. Osteoclast differentiation process was achieved after 5 days of incubation with RANKL in 24-well culture plates. Introducing the non-polar lipid fraction into the culture caused a lack of cell differentiation, and a reduction in tartrate-resistant acid phosphatase (TRAP) activity and TRAP cell numbers in a dose-dependent manner (50% reduction at the concentration of 20 μg/mL, p < 0.001). Moreover, actin ring formation was significantly diminished by non-polar lipids at 10-20 μg/mL. The bone digestive enzymes released by osteoclasts into the pit formation were also compromised by downregulating gene expression of cathepsin K, carbonic anhydrase II (CA II), matrix metalloproteinase 9 (MMP-9), and nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1). This study revealed that the non-polar lipid fraction of GSM oil contains bioactive substances which possess potent anti-osteoclastogenic activity.
Collapse
Key Words
- AA, Arachidonic acid
- ALA, Alpha linolenic acid
- CAII, Carbonic anhydrase II
- DHA, Docosahexaenoic acid
- DMSO, dimethyl sulfoxide
- DPA, Docosapentaenoic acid
- EPA, Eicosapentaenoic acid
- FFAR, Free fatty acid receptor
- GSM, Greenshell mussel
- Greenshell mussel
- LA, Linoleic acid
- LPS, Lipopolysaccharide
- MMP-9, Matrix metalloproteinase 9
- MUFA, Monounsaturated fatty acid
- NF-κB, Nuclear factor κB
- NFATc1, Nuclear factor of activated T-cells, cytoplasmic 1
- OA, Osteoarthritis
- Omega 3 fatty acid
- Osteoarthritis
- Osteoclasts
- Osteoporosis
- PA, Palmitic acid
- PPAR, Peroxisome proliferator activated receptor
- PUFA, Polyunsaturated fatty acid
- RANKL, Receptor activator of nuclear factor κB ligand
- SFA, Saturated fatty acid
- TRAP, Tartrate-resistant acid phosphatase
Collapse
|
31
|
Intracellular Signaling Responses Induced by Radiation within an In Vitro Bone Metastasis Model after Pre-Treatment with an Estrone Analogue. Cells 2021; 10:cells10082105. [PMID: 34440874 PMCID: PMC8394480 DOI: 10.3390/cells10082105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 12/27/2022] Open
Abstract
2-Ethyl-3-O-sulfamoyl-estra-1,3,5(10)16-tetraene (ESE-16) is an in silico-designed estradiol analogue which has improved the parent compound’s efficacy in anti-cancer studies. In this proof-of-concept study, the potential radiosensitizing effects of ESE-16 were investigated in an in vitro deconstructed bone metastasis model. Prostate (DU 145) and breast (MDA-MB-231) tumor cells, osteoblastic (MC3T3-E1) and osteoclastic (RAW 264.7) bone cells and human umbilical vein endothelial cells (HUVECs) were representative components of such a lesion. Cells were exposed to a low-dose ESE-16 for 24 hours prior to radiation at non-lethal doses to determine early signaling and molecular responses of this combination treatment. Tartrate-resistant acid phosphatase activity and actin ring formation were investigated in osteoclasts, while cell cycle progression, reactive oxygen species generation and angiogenic protein expression were investigated in HUVECs. Increased cytotoxicity was evident in tumor and endothelial cells while bone cells appeared to be spared. Increased mitotic indices were calculated, and evidence of increased deoxyribonucleic acid damage with retarded repair, together with reduced metastatic signaling was observed in tumor cells. RAW 264.7 macrophages retained their ability to differentiate into osteoclasts. Anti-angiogenic effects were observed in HUVECs, and expression of hypoxia-inducible factor 1-α was decreased. Through preferentially inducing tumor cell death and potentially inhibiting neovascularization whilst preserving bone physiology, this low-dose combination regimen warrants further investigation for its promising therapeutic application in bone metastases management, with the additional potential of limited treatment side effects.
Collapse
|
32
|
ELMO1 signaling is a promoter of osteoclast function and bone loss. Nat Commun 2021; 12:4974. [PMID: 34404802 PMCID: PMC8371122 DOI: 10.1038/s41467-021-25239-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 07/28/2021] [Indexed: 01/02/2023] Open
Abstract
Osteoporosis affects millions worldwide and is often caused by osteoclast induced bone loss. Here, we identify the cytoplasmic protein ELMO1 as an important ‘signaling node’ in osteoclasts. We note that ELMO1 SNPs associate with bone abnormalities in humans, and that ELMO1 deletion in mice reduces bone loss in four in vivo models: osteoprotegerin deficiency, ovariectomy, and two types of inflammatory arthritis. Our transcriptomic analyses coupled with CRISPR/Cas9 genetic deletion identify Elmo1 associated regulators of osteoclast function, including cathepsin G and myeloperoxidase. Further, we define the ‘ELMO1 interactome’ in osteoclasts via proteomics and reveal proteins required for bone degradation. ELMO1 also contributes to osteoclast sealing zone on bone-like surfaces and distribution of osteoclast-specific proteases. Finally, a 3D structure-based ELMO1 inhibitory peptide reduces bone resorption in wild type osteoclasts. Collectively, we identify ELMO1 as a signaling hub that regulates osteoclast function and bone loss, with relevance to osteoporosis and arthritis. Osteoporosis and bone fractures affect millions of patients worldwide and are often due to increased bone resorption. Here the authors identify the cytoplasmic protein ELMO1 as an important ‘signaling node’ promoting the bone resorption function of osteoclasts.
Collapse
|
33
|
Halper J, Madel MB, Blin-Wakkach C. Differentiation and Phenotyping of Murine Osteoclasts from Bone Marrow Progenitors, Monocytes, and Dendritic Cells. Methods Mol Biol 2021; 2308:21-34. [PMID: 34057711 DOI: 10.1007/978-1-0716-1425-9_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Bone physiology is dictated by various players, including osteoclasts (OCLs) as bone resorbing cells, osteoblasts (capable of bone formation), osteocytes, or mesenchymal stem cells, to mention the most important players. All these cells are in tight communication with each other and influence the constantly occurring process of bone remodeling to meet changing requirements on the skeletal system. In order to understand these interplays, one must investigate isolated functions of the various cell types. However, OCL research displays a special drawback: due to their giant size, low abundance, and tight attachment on the bone surface, ex vivo isolation of sufficient amounts of mature OCLs is limited or not conceivable in most species including mice. Moreover, OCLs can be obtained from different progenitors in vivo as well as in vitro. Thus, in vitro differentiation of OCLs from various progenitor cells remains essential in the analysis of OCL biology, underlining the importance of reliable gold standard protocols to be applied throughout OCL research. This chapter will deal with in vitro differentiation of OCLs from murine bone marrow cells, as well as isolated monocytes and dendritic cells that have already been validated in numerous studies.
Collapse
Affiliation(s)
- Julia Halper
- Université Côte d'Azur, CNRS, UMR7370, LP2M, Nice, France
| | - Maria-Bernadette Madel
- Université Côte d'Azur, CNRS, UMR7370, LP2M, Nice, France
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
34
|
Pouraghaei Sevari S, Kim JK, Chen C, Nasajpour A, Wang CY, Krebsbach PH, Khademhosseini A, Ansari S, Weiss PS, Moshaverinia A. Whitlockite-Enabled Hydrogel for Craniofacial Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35342-35355. [PMID: 34297530 DOI: 10.1021/acsami.1c07453] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Growth-factor-free bone regeneration remains a challenge in craniofacial engineering. Here, we engineered an osteogenic niche composed of a commercially modified alginate hydrogel and whitlockite microparticles (WHMPs), which impart tunable physicochemical properties that can direct osteogenesis of human gingival mesenchymal stem cells (GMSCs). Our in vitro studies demonstrate that WHMPs induce osteogenesis of GMSCs more effectively than previously demonstrated hydroxyapatite microparticles (HApMPs). Alginate-WHMP hydrogels showed higher elasticity without any adverse effects on the viability of the encapsulated GMSCs. Moreover, the alginate-WHMP hydrogels upregulate the mitogen-activated protein kinase (MAPK) pathway, which in turn orchestrates several osteogenic markers, such as RUNX2 and OCN, in the encapsulated GMSCs. Concurrent coculture studies with human osteoclasts demonstrate that GMSCs encapsulated in alginate-WHMP hydrogels downregulate osteoclastic activity, potentially due to release of Mg2+ ions from the WHMPs along with secretion of osteoprotegerin from the GMSCs. In vivo studies demonstrated that the GMSCs encapsulated in our osteogenic niche were able to promote bone repair in calvarial defects in murine models. Altogether, our results confirmed the development of a promising treatment modality for craniofacial bone regeneration based on an injectable growth-factor-free hydrogel delivery system.
Collapse
Affiliation(s)
- Sevda Pouraghaei Sevari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Jin Koo Kim
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Amir Nasajpour
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Cun-Yu Wang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Paul H Krebsbach
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90049, United States
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Paul S Weiss
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
35
|
Husch JFA, Stessuk T, den Breejen C, van den Boom M, Leeuwenburgh SCG, van den Beucken JJJP. A Practical Procedure for the In Vitro Generation of Human Osteoclasts and Their Characterization. Tissue Eng Part C Methods 2021; 27:421-432. [PMID: 34162266 DOI: 10.1089/ten.tec.2021.0122] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Osteoclasts are multinucleated cells derived from the hematopoietic monocyte/macrophage lineage that possess the unique capacity to resorb bone. Due to the crucial role of osteoclasts in maintaining bone homeostasis and pathologies, this cell type is pivotal in multiple research areas dedicated to bone physiology in health and disease. Although numerous methods for generation of human osteoclasts are already available, those rely either on cell labeling-based purification or an intermediate adhesion step after which cells are directly differentiated toward osteoclasts. While the former requires additional reagents and equipment, the latter harbors the risk of variable osteoclast formation due to varying numbers of osteoclast precursors available for different donors. In this study, we report a facile and reliable three-step method for the generation of human osteoclasts from blood-derived precursor cells. Monocytes were obtained after adhering peripheral blood-derived mononuclear cells to plastic substrates followed by macrophage induction and proliferation resulting in a homogeneous population of osteoclast precursors. Finally, macrophages were seeded into suitable culture vessels and differentiated toward osteoclasts. Osteoclastogenesis was monitored longitudinally using nondestructive techniques, while the functionality of mature osteoclasts was confirmed after 14 days of culture by analysis of functional (e.g., elevated tartrate-resistant acid phosphatase [TRAP]-activity, resorption) and morphological (e.g., presence of TRAP, actin ring, and integrin β3) characteristics. Furthermore, we propose to use combinatory staining of three morphological osteoclast markers, rather than previously reported staining of a single or maximal two markers, to clearly distinguish osteoclasts from undifferentiated mononuclear cells. Impact statement Research related to bone biology requires a standardized and reliable method for in vitro generation of human osteoclasts. We here describe such a procedure which avoids shortcomings of previously published protocols. Further, we report on nondestructive methods to qualitatively and quantitatively monitor osteoclastogenesis longitudinally, and on analysis of osteoclast generation and functionality after 14 days. Specifically, we recommend assessment of morphological human osteoclast characteristics using combinatory staining of three markers to confirm successful osteoclast generation.
Collapse
Affiliation(s)
- Johanna F A Husch
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Talita Stessuk
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Cèzanne den Breejen
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Manouk van den Boom
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Sander C G Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | - Jeroen J J P van den Beucken
- Department of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
36
|
Chaichit S, Sato T, Yu H, Tanaka YK, Ogra Y, Mizoguchi T, Itoh M. Evaluation of Dexamethasone-Induced Osteoporosis In Vivo Using Zebrafish Scales. Pharmaceuticals (Basel) 2021; 14:ph14060536. [PMID: 34205111 PMCID: PMC8228068 DOI: 10.3390/ph14060536] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 02/03/2023] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) is a major cause of secondary osteoporosis, and the pathogenic mechanisms of GIOP remain to be elucidated. Here, we show a rapid dexamethasone-induced osteoporosis animal model using zebrafish scales. Intraperitoneal injection of dexamethasone over a 5-day period suppressed the regeneration of scales. Furthermore, the circularity of the newly formed regenerated scales was also slightly reduced compared to that of the control group on day 5. The changes in bone-related enzymes, such as cathepsin K, tartrate-resistant acid phosphatase (TRAP) for bone resorption, and alkaline phosphatase (ALP) for bone formation, provide insight into the progression of bone diseases; therefore, we further developed a method to measure the activities of cathepsin K, TRAP, and ALP using zebrafish scales. We found that a lysis buffer with detergent at neutral pH under sonication efficiently helped extract these three enzymes with high activity levels. Interestingly, treatment with a dexamethasone injection produced considerably higher levels of cathepsin K activity and a lower Ca/P ratio than those in the control group, suggesting that dexamethasone increased osteoclast activity, with no significant changes in the activities of TRAP and ALP. Our GIOP model and enzyme assay method could help to design better treatments for GIOP.
Collapse
Affiliation(s)
- Siripat Chaichit
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Takuto Sato
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
| | - Huiqing Yu
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
| | - Yu-ki Tanaka
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
| | - Yasumitsu Ogra
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
| | - Takamasa Mizoguchi
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
| | - Motoyuki Itoh
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (S.C.); (T.S.); (H.Y.); (Y.-k.T.); (Y.O.); (T.M.)
- Correspondence: ; Tel.: +81-43-226-2890
| |
Collapse
|
37
|
Cohen-Karlik E, Awida Z, Bergman A, Eshed S, Nestor O, Kadashev M, Yosef SB, Saed H, Mansour Y, Globerson A, Neumann D, Gabet Y. Quantification of Osteoclasts in Culture, Powered by Machine Learning. Front Cell Dev Biol 2021; 9:674710. [PMID: 34113621 PMCID: PMC8186397 DOI: 10.3389/fcell.2021.674710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/26/2021] [Indexed: 12/02/2022] Open
Abstract
In vitro osteoclastogenesis is a central assay in bone biology to study the effect of genetic and pharmacologic cues on the differentiation of bone resorbing osteoclasts. To date, identification of TRAP+ multinucleated cells and measurements of osteoclast number and surface rely on a manual tracing requiring specially trained lab personnel. This task is tedious, time-consuming, and prone to operator bias. Here, we propose to replace this laborious manual task with a completely automatic process using algorithms developed for computer vision. To this end, we manually annotated full cultures by contouring each cell, and trained a machine learning algorithm to detect and classify cells into preosteoclast (TRAP+ cells with 1-2 nuclei), osteoclast type I (cells with more than 3 nuclei and less than 15 nuclei), and osteoclast type II (cells with more than 15 nuclei). The training usually requires thousands of annotated samples and we developed an approach to minimize this requirement. Our novel strategy was to train the algorithm by working at "patch-level" instead of on the full culture, thus amplifying by >20-fold the number of patches to train on. To assess the accuracy of our algorithm, we asked whether our model measures osteoclast number and area at least as well as any two trained human annotators. The results indicated that for osteoclast type I cells, our new model achieves a Pearson correlation (r) of 0.916 to 0.951 with human annotators in the estimation of osteoclast number, and 0.773 to 0.879 for estimating the osteoclast area. Because the correlation between 3 different trained annotators ranged between 0.948 and 0.958 for the cell count and between 0.915 and 0.936 for the area, we can conclude that our trained model is in good agreement with trained lab personnel, with a correlation that is similar to inter-annotator correlation. Automation of osteoclast culture quantification is a useful labor-saving and unbiased technique, and we suggest that a similar machine-learning approach may prove beneficial for other morphometrical analyses.
Collapse
Affiliation(s)
- Edo Cohen-Karlik
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Zamzam Awida
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ayelet Bergman
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Shahar Eshed
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Omer Nestor
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Michelle Kadashev
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sapir Ben Yosef
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hussam Saed
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yishay Mansour
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Amir Globerson
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
38
|
Morimoto A, Kikuta J, Nishikawa K, Sudo T, Uenaka M, Furuya M, Hasegawa T, Hashimoto K, Tsukazaki H, Seno S, Nakamura A, Okuzaki D, Sugihara F, Ninomiya A, Yoshimura T, Takao-Kawabata R, Matsuda H, Ishii M. SLPI is a critical mediator that controls PTH-induced bone formation. Nat Commun 2021; 12:2136. [PMID: 33837198 PMCID: PMC8035405 DOI: 10.1038/s41467-021-22402-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoclastic bone resorption and osteoblastic bone formation/replenishment are closely coupled in bone metabolism. Anabolic parathyroid hormone (PTH), which is commonly used for treating osteoporosis, shifts the balance from osteoclastic to osteoblastic, although it is unclear how these cells are coordinately regulated by PTH. Here, we identify a serine protease inhibitor, secretory leukocyte protease inhibitor (SLPI), as a critical mediator that is involved in the PTH-mediated shift to the osteoblastic phase. Slpi is highly upregulated in osteoblasts by PTH, while genetic ablation of Slpi severely impairs PTH-induced bone formation. Slpi induction in osteoblasts enhances its differentiation, and increases osteoblast-osteoclast contact, thereby suppressing osteoclastic function. Intravital bone imaging reveals that the PTH-mediated association between osteoblasts and osteoclasts is disrupted in the absence of SLPI. Collectively, these results demonstrate that SLPI regulates the communication between osteoblasts and osteoclasts to promote PTH-induced bone anabolism.
Collapse
Affiliation(s)
- Akito Morimoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| | - Keizo Nishikawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takao Sudo
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Maki Uenaka
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masayuki Furuya
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kunihiko Hashimoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hiroyuki Tsukazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Akira Nakamura
- Division of Immunology, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Daisuke Okuzaki
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Akinori Ninomiya
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takeshi Yoshimura
- Medical Affairs Department, Asahi Kasei Pharma Corporation, Tokyo, Japan
| | - Ryoko Takao-Kawabata
- Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, Tokyo, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan.
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan.
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.
| |
Collapse
|
39
|
Knapinska AM, Singh C, Drotleff G, Blanco D, Chai C, Schwab J, Herd A, Fields GB. Matrix Metalloproteinase 13 Inhibitors for Modulation of Osteoclastogenesis: Enhancement of Solubility and Stability. ChemMedChem 2021; 16:1133-1142. [PMID: 33331147 PMCID: PMC8035250 DOI: 10.1002/cmdc.202000911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/14/2020] [Indexed: 11/08/2022]
Abstract
Matrix metalloproteinase 13 (MMP-13) activity has been correlated to breast cancer bone metastasis. It has been proposed that MMP-13 contributes to bone metastasis through the promotion of osteoclastogenesis. To explore the mechanisms of MMP-13 action, we previously described a highly efficacious and selective MMP-13 inhibitor, RF036. Unfortunately, further pursuit of RF036 as a probe of MMP-13 in vitro and in vivo activities was not practical due to the limited solubility and stability of the inhibitor. Our new study has explored replacing the RF036 backbone sulfur atom and terminal methyl group to create inhibitors with more favorable pharmacokinetic properties. One compound, designated inhibitor 3, in which the backbone sulfur and terminal methyl group of RF036 were replaced by nitrogen and oxetane, respectively, had comparable activity, selectivity, and membrane permeability to RF036, while exhibiting greatly enhanced solubility and stability. Inhibitor 3 effectively inhibited MMP-13-mediated osteoclastogenesis but spared collagenolysis, and thus represents a next-generation MMP-13 probe applicable for in vivo studies of breast cancer metastasis.
Collapse
Affiliation(s)
- Anna M Knapinska
- Department of Chemistry & Biochemistry, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Chandani Singh
- Department of Chemistry & Biochemistry, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Gary Drotleff
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | - Daniela Blanco
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Cedric Chai
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA
| | - Jason Schwab
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Anu Herd
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
| | - Gregg B Fields
- Department of Chemistry & Biochemistry, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
- Institute for Human Health & Disease Intervention (I-HEALTH), Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL 33458, USA
- Department of Chemistry, The Scripps Research Institute/Scripps Florida, 120 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
40
|
Biological Evaluation and Transcriptomic Analysis of Corylin as an Inhibitor of Osteoclast Differentiation. Int J Mol Sci 2021; 22:ijms22073540. [PMID: 33805517 PMCID: PMC8036378 DOI: 10.3390/ijms22073540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/27/2022] Open
Abstract
Corylin, a flavonoid isolated from the fruit of Psoralea corylifolia, has an osteogenic effect on osteoblasts in vitro and bone micromass ex vivo. However, the effect and mechanism of corylin in regulating osteoclastogenesis remain unknown. By using murine bone marrow macrophages as the osteoclast precursor, corylin was found to inhibit the receptor activator of nuclear factor (NF) κB ligand (RANKL)-induced osteoclast differentiation via down-regulating osteoclastic marker genes. In parallel, F-actin formation and osteoclast migration were diminished in corylin-treated cultured osteoclasts, and subsequently the expressions of osteoclastic proteins were suppressed: the suppression of protein expression was further illustrated by transcriptomic analysis. Furthermore, corylin inhibited the nuclear translocation of p65, giving rise to a restraint in osteoclastic differentiation through the attenuation of transcription factors nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and nuclear factor of activated T cells c1 (NFATc1). There was no obvious change in apoptosis when the RANKL-induce osteoclasts were cultured in the presence of corylin. The finding supports the potential development of corylin as an osteoclast inhibitor against osteoporosis.
Collapse
|
41
|
Tavares SJS, Lima V. Bone anti-resorptive effects of coumarins on RANKL downstream cellular signaling: a systematic review of the literature. Fitoterapia 2021; 150:104842. [PMID: 33556550 DOI: 10.1016/j.fitote.2021.104842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/27/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Members of the botanical families Apiaceae/Umbelliferae, Asteraceae, Fabaceae/Leguminosae, and Thymelaeaceae are rich in coumarins and have traditionally been used as ethnomedicines in many regions including Europe, Asia, and South America. Coumarins are a class of secondary metabolites that are widely present in plants, fungi, and bacteria and exhibit several pharmacological, biochemical, and therapeutic effects. Recently, many plants rich in coumarins and their derivatives were found to affect bone metabolism. OBJECTIVE To review scientific literature describing the mechanisms of action of coumarins in osteoclastogenesis and bone resorption. MATERIALS AND METHODS For this systematic review, the PubMed, Scopus, and Periodical Capes databases and portals were searched. We included in vitro research articles published between 2010 and 2020 that evaluated coumarins using osteoclastogenic markers. RESULTS Coumarins have been reported to downregulate RANKL-RANK signaling and various downstream signaling pathways required for osteoclast development, such as NF-κB, MAPK, Akt, and Ca2+ signaling, as well as pathways downstream of the nuclear factor of activated T-cells (NFATc1), including tartrate-resistant acid phosphatase (TRAP), cathepsin K (CTSK), and matrix metalloproteinase 9 (MMP-9). CONCLUSIONS Coumarins primarily inhibit osteoclast differentiation and activation by modulating different intracellular signaling pathways; therefore, they could serve as potential candidates for controlled randomized clinical trials aimed at improving human bone health.
Collapse
Affiliation(s)
- Samia Jessica Silva Tavares
- School of Pharmacy, Nursing, and Dentistry, Federal University of Ceará, Fortaleza, Ceará 60430-355, Brazil.
| | - Vilma Lima
- School of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará 60.430-275, Brazil.
| |
Collapse
|
42
|
Zhou L, Poon CCW, Wong KY, Cao S, Dong X, Zhang Y, Wong MS. Icariin ameliorates estrogen-deficiency induced bone loss by enhancing IGF-I signaling via its crosstalk with non-genomic ERα signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153413. [PMID: 33339654 DOI: 10.1016/j.phymed.2020.153413] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 10/20/2020] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Rapid, non-genomic estrogen receptor (ER) signaling plays an integral role in mediating the tissue selective properties of ER modulators. Icariin, a bone bioactive flavonoid, has been reported to selectively activate non-genomic ERα signaling in in vitro and in vivo studies. PURPOSE The mechanisms underlying the estrogen-like bone protective effects of icariin are not fully understood, especially those that are related to insulin-like growth factor I (IGF-1) signaling. The bone protective effects of icariin were investigated in female mature ovariectomized (OVX) rats and the signaling of IGF-IR- ERα cross-talk was determined in osteoblastic cells. STUDY DESIGN AND METHODS Icariin at 3 different dosages (50, 500 and 3000 ppm) were orally administrated to rats for 3 months through daily intake of phytoestrogen-free animal diets containing icariin. Bone marrow stromal cells (BMSCs) and osteoclast precursors from femurs were harvested for experiments and RNA-sequencing. The interactions between IGF-IR and non-genomic ERα signaling were examined in pre-osteoblastic MC3T3-E1 cells and mature osteoblasts differentiated from BMSCs. RESULTS Our results show that chronic administration of icariin to OVX rats significantly protected them against bone loss at the long bone and lumbar spine without inducing any uterotrophic effects. Ex vivo studies using BMSCs and osteoclast precursors confirmed the stimulatory effects of icariin on osteoblastogenesis and its inhibitory effects on osteoclastogenesis, respectively. RNA-sequencing analysis of mRNA from BMSCs revealed that icariin at 500 ppm significantly altered IGF-1 signaling as well as PI3K-Akt pathways. Our results demonstrated for the first time the rapid induction of interactions between IGF-IR and ERα as well as IGF-IR signaling and the downstream Akt phosphorylation by icariin in MC3T3-E1 cells. The activation of ERα and Akt phosphorylation by icariin in MC3T3-E1 cells and the osteogenic effects of icariin on ALP activity in mature osteoblasts were shown to be IGF-IR-dependent. CONCLUSION Our findings reveal that icariin activates both ERα and Akt via enhancing rapid induction of IGF-1 signaling in osteoblastic cells for osteogenesis and might be regarded as a novel pathway-selective phytoestrogen for management of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Christina Chui-Wa Poon
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Ka-Ying Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Sisi Cao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Xiaoli Dong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR
| | - Yan Zhang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| | - Man-Sau Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR; State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, PR China.
| |
Collapse
|
43
|
Place DE, Malireddi RKS, Kim J, Vogel P, Yamamoto M, Kanneganti TD. Osteoclast fusion and bone loss are restricted by interferon inducible guanylate binding proteins. Nat Commun 2021; 12:496. [PMID: 33479228 PMCID: PMC7820603 DOI: 10.1038/s41467-020-20807-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation during many diseases is associated with bone loss. While interferons (IFNs) are often inhibitory to osteoclast formation, the complex role that IFN and interferon-stimulated genes (ISGs) play in osteoimmunology during inflammatory diseases is still poorly understood. We show that mice deficient in IFN signaling components including IFN alpha and beta receptor 1 (IFNAR1), interferon regulatory factor 1 (IRF1), IRF9, and STAT1 each have reduced bone density and increased osteoclastogenesis compared to wild type mice. The IFN-inducible guanylate-binding proteins (GBPs) on mouse chromosome 3 (GBP1, GBP2, GBP3, GBP5, GBP7) are required to negatively regulate age-associated bone loss and osteoclastogenesis. Mechanistically, GBP2 and GBP5 both negatively regulate in vitro osteoclast differentiation, and loss of GBP5, but not GBP2, results in greater age-associated bone loss in mice. Moreover, mice deficient in GBP5 or chromosome 3 GBPs have greater LPS-mediated inflammatory bone loss compared to wild type mice. Overall, we find that GBP5 contributes to restricting age-associated and inflammation-induced bone loss by negatively regulating osteoclastogenesis.
Collapse
Affiliation(s)
- David E Place
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jieun Kim
- Center for In Vivo Imaging and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | |
Collapse
|
44
|
The muscle to bone axis (and viceversa): An encrypted language affecting tissues and organs and yet to be codified? Pharmacol Res 2021; 165:105427. [PMID: 33453372 DOI: 10.1016/j.phrs.2021.105427] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/20/2020] [Accepted: 01/10/2021] [Indexed: 12/15/2022]
Abstract
Skeletal muscles and bone tissue form the musculoskeletal apparatus, a complex system essential for the voluntary movement. The loss of muscle mass and muscle strength is often associated with a loss of bone mass, in a "hazardous duet" which implies the co-existence of sarcopenia-osteoporosis and exposes patients to a deterioration in quality of life and increased mortality. From the mechanostat theory to the recent definition of the osteosarcopenia syndrome, many aspects of muscle-bone interaction have been investigated in recent decades. The mechanical interaction is now accepted, considering the close anatomical relationship between the two tissues, however, much remains to be discovered regarding the biochemical muscle-bone interaction. Skeletal muscle has been defined as an endocrine organ capable of exerting an action on other tissues. Myokines, bioactive polypeptides released by the muscle, could represent the encrypted message in the communication between muscle and bone. These two tissues have a reciprocal influence on their metabolisms and respond in a similar way to the multiple external factors. The aim of this review is to stimulate the understanding of the encrypted language between muscle and bone, highlighting the role of catabolic pathways and oxidative stress in the musculoskeletal apparatus to elucidate the shared mechanisms and the similarity of response to the same stimuli by different tissues. Our understanding of muscle-bone interactions it could be useful to identify and develop new strategies to treat musculoskeletal diseases, together with pharmacological, nutritional and exercise-based approaches, which are already in use for the treatment of these pathologies.
Collapse
|
45
|
Establishment and validation of an in vitro co-culture model for oral cell lines using human PBMC-derived osteoclasts, osteoblasts, fibroblasts and keratinocytes. Sci Rep 2020; 10:16861. [PMID: 33033302 PMCID: PMC7544897 DOI: 10.1038/s41598-020-73941-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/27/2020] [Indexed: 12/30/2022] Open
Abstract
Indirect co-culture models with osteoclasts including oral cell lines may be influenced by M-CSF and RANKL in the common cell medium. Therefore, we investigated the viability and proliferation of osteoblasts (OB), fibroblasts (FB) and oral keratinocytes (OK) under stratified medium modification and assessed the differentiation of osteoclasts in each co-culture. The impact of M-CSF and RANKL in the common OC co-culture was assessed for OB, FB and OK via MTT assay via DAPI control. The multinuclearity and function of OC were evaluated by light microscopy, DAPI staining, resorption assay and FACS analysis. The PBMC showed the highest differentiation into OC after an incubation period of 7 days. Furthermore, co-culture with OB enhanced the number of differentiated multinucleated OC in comparison with monoculture, whereas co-culture with OK decreased PBMC multinuclearity and OC differentiation. FB did not influence the number of differentiated OC in a co-culture. RANKL and M-CSF reduction had no impact on OC differentiation in co-culture with FB or OB, whereas this medium modification for OK attenuated PBMC multinuclearity and OC differentiation in all approaches. Supplementation of RANKL and M-CSF can be modified for a co-culture of PBMC with FB or OB without disturbing OC differentiation. Thus, pathogenic processes of bone remodelling involving OB, OC, FB and OK in the oral cavity can be investigated thoroughly.
Collapse
|
46
|
Estell EG, Le PT, Vegting Y, Kim H, Wrann C, Bouxsein ML, Nagano K, Baron R, Spiegelman BM, Rosen CJ. Irisin directly stimulates osteoclastogenesis and bone resorption in vitro and in vivo. eLife 2020; 9:e58172. [PMID: 32780016 PMCID: PMC7444909 DOI: 10.7554/elife.58172] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022] Open
Abstract
Irisin, a skeletal-muscle secreted myokine, facilitates muscle-bone crosstalk and skeletal remodeling in part by its action on osteoblasts and osteocytes. In this study, we investigated whether irisin directly regulates osteoclasts. In vitro, irisin (2-10 ng/mL) increased osteoclast differentiation in C57BL/6J mouse bone marrow progenitors; however, this increase was blocked by a neutralizing antibody to integrin αVβ5. Irisin also increased bone resorption on several substrates in situ. RNAseq revealed differential gene expression induced by irisin including upregulation of markers for osteoclast differentiation and resorption, as well as osteoblast-stimulating 'clastokines'. Forced expression of the irisin precursor Fndc5 in transgenic C57BL/6J mice resulted in lower bone mass at three ages and greater in vitro osteoclastogenesis from Fndc5-transgenic bone marrow progenitors. This study demonstrates that irisin acts directly on osteoclast progenitors to increase differentiation and promote bone resorption, supporting the tenet that irisin not only stimulates bone remodeling but may also be an important counter-regulatory hormone.
Collapse
Affiliation(s)
- Eben G Estell
- Maine Medical Center Research InstituteScarboroughUnited States
| | - Phuong T Le
- Maine Medical Center Research InstituteScarboroughUnited States
| | - Yosta Vegting
- Maine Medical Center Research InstituteScarboroughUnited States
| | | | - Christiane Wrann
- Dana Farber Cancer InstituteBostonUnited States
- Cardiovascular Research Center, Massachusetts General HospitalBostonUnited States
- Department of Medicine, Harvard Medical SchoolBostonUnited States
- Department of Cell Biology, Harvard University Medical SchoolBostonUnited States
| | - Mary L Bouxsein
- Beth Israel Deaconess Department of Orthopedic Surgery, Harvard Medical SchoolBostonUnited States
| | | | - Roland Baron
- Harvard School of Dental MedicineBostonUnited States
| | | | | |
Collapse
|
47
|
Dennis EP, Edwards SM, Jackson RM, Hartley CL, Tsompani D, Capulli M, Teti A, Boot-Handford RP, Young DA, Piróg KA, Briggs MD. CRELD2 Is a Novel LRP1 Chaperone That Regulates Noncanonical WNT Signaling in Skeletal Development. J Bone Miner Res 2020; 35:1452-1469. [PMID: 32181934 DOI: 10.1002/jbmr.4010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
Cysteine-rich with epidermal growth factor (EGF)-like domains 2 (CRELD2) is an endoplasmic reticulum (ER)-resident chaperone highly activated under ER stress in conditions such as chondrodysplasias; however, its role in healthy skeletal development is unknown. We show for the first time that cartilage-specific deletion of Creld2 results in disrupted endochondral ossification and short limbed dwarfism, whereas deletion of Creld2 in bone results in osteopenia, with a low bone density and altered trabecular architecture. Our study provides the first evidence that CRELD2 promotes the differentiation and maturation of skeletal cells by modulating noncanonical WNT4 signaling regulated by p38 MAPK. Furthermore, we show that CRELD2 is a novel chaperone for the receptor low-density lipoprotein receptor-related protein 1 (LRP1), promoting its transport to the cell surface, and that LRP1 directly regulates WNT4 expression in chondrocytes through TGF-β1 signaling. Therefore, our data provide a novel link between an ER-resident chaperone and the essential WNT signaling pathways active during skeletal differentiation that could be applicable in other WNT-responsive tissues. © 2020 American Society for Bone and Mineral Research. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research..
Collapse
Affiliation(s)
- Ella P Dennis
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | - Sarah M Edwards
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Robert M Jackson
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | - Claire L Hartley
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Dimitra Tsompani
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | - Mattia Capulli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | | | - David A Young
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | - Katarzyna A Piróg
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| | - Michael D Briggs
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne, UK
| |
Collapse
|
48
|
Zheng ZG, Cheng HM, Zhou YP, Zhu ST, Thu PM, Li HJ, Li P, Xu X. Dual targeting of SREBP2 and ERRα by carnosic acid suppresses RANKL-mediated osteoclastogenesis and prevents ovariectomy-induced bone loss. Cell Death Differ 2020; 27:2048-2065. [PMID: 31907393 PMCID: PMC7308277 DOI: 10.1038/s41418-019-0484-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022] Open
Abstract
Osteoporosis develops because of impaired bone formation and/or excessive bone resorption. Several pharmacological treatment of osteoporosis has been developed; however, new treatments are still necessary. Cholesterol and estrogen receptor-related receptor alpha (ERRα) promote osteoclasts formation, survival, and cellular fusion and thus become high risk factors of osteoporosis. In this study, we identified that carnosic acid (CA) suppressed bone loss by dual-targeting of sterol regulatory element-binding protein 2 (SREBP2, a major regulator that regulates cholesterol synthesis) and ERRα. Mechanistically, CA reduced nuclear localization of mature SREBP2 and suppressed de novo biogenesis of cholesterol. CA subsequently decreased the interaction between ERRα and peroxisome proliferator-activated receptor gamma coactivator 1-beta (PGC1β), resulting in decreased the transcription activity of ERRα and its target genes expression. Meanwhile, CA directly bound to the ligand-binding domain of ERRα and significantly promoted its ubiquitination and proteasomal degradation. Subsequently, STUB1 was identified as the E3 ligase of ERRα. The lysine residues (K51 and K68) are essential for ubiquitination and proteasomal degradation of ERRα by CA. In conclusion, CA dually targets SREBP2 and ERRα, thus inhibits the RANKL-induced osteoclast formation and improves OVX-induced bone loss. CA may serve as a lead compound for pharmacological control of osteoporosis.
Collapse
Affiliation(s)
- Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Hui-Min Cheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Ya-Ping Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Si-Tong Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Pyone Myat Thu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China.
| | - Xiaojun Xu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China.
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China.
| |
Collapse
|
49
|
Møller AMJ, Delaissé JM, Olesen JB, Madsen JS, Canto LM, Bechmann T, Rogatto SR, Søe K. Aging and menopause reprogram osteoclast precursors for aggressive bone resorption. Bone Res 2020; 8:27. [PMID: 32637185 PMCID: PMC7329827 DOI: 10.1038/s41413-020-0102-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Women gradually lose bone from the age of ~35 years, but around menopause, the rate of bone loss escalates due to increasing bone resorption and decreasing bone formation levels, rendering these individuals more prone to developing osteoporosis. The increased osteoclast activity has been linked to a reduced estrogen level and other hormonal changes. However, it is unclear whether intrinsic changes in osteoclast precursors around menopause can also explain the increased osteoclast activity. Therefore, we set up a protocol in which CD14+ blood monocytes were isolated from 49 female donors (40-66 years old). Cells were differentiated into osteoclasts, and data on differentiation and resorption activity were collected. Using multiple linear regression analyses combining in vitro and in vivo data, we found the following: (1) age and menopausal status correlate with aggressive osteoclastic bone resorption in vitro; (2) the type I procollagen N-terminal propeptide level in vivo inversely correlates with osteoclast resorption activity in vitro; (3) the protein level of mature cathepsin K in osteoclasts in vitro increases with age and menopause; and (4) the promoter of the gene encoding the dendritic cell-specific transmembrane protein is less methylated with age. We conclude that monocytes are "reprogrammed" in vivo, allowing them to "remember" age, the menopausal status, and the bone formation status in vitro, resulting in more aggressive osteoclasts. Our discovery suggests that this may be mediated through DNA methylation. We suggest that this may have clinical implications and could contribute to understanding individual differences in age- and menopause-induced bone loss.
Collapse
Affiliation(s)
- Anaïs Marie Julie Møller
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Jean-Marie Delaissé
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jacob Bastholm Olesen
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Luisa Matos Canto
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Troels Bechmann
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Silvia Regina Rogatto
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Kent Søe
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
- OPEN, Odense Patient data Explorative Network, Odense University Hospital, 5000 Odense C, Denmark
| |
Collapse
|
50
|
Krieger NS, Chen L, Becker J, DeBoyace S, Wang H, Favus MJ, Bushinsky DA. Increased Osteoclast and Decreased Osteoblast Activity Causes Reduced Bone Mineral Density and Quality in Genetic Hypercalciuric Stone-Forming Rats. JBMR Plus 2020; 4:e10350. [PMID: 32258968 PMCID: PMC7117851 DOI: 10.1002/jbm4.10350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/09/2020] [Indexed: 11/16/2022] Open
Abstract
To study human idiopathic hypercalciuria (IH), we developed an animal model, genetic hypercalciuric stone-forming (GHS) rats, whose pathophysiology parallels that in IH. All GHS rats form kidney stones and have decreased BMD and bone quality compared with the founder Sprague-Dawley (SD) rats. To understand the bone defect, we characterized osteoclast and osteoblast activity in the GHS compared with SD rats. Bone marrow cells were isolated from femurs of GHS and SD rats and cultured to optimize differentiation into osteoclasts or osteoblasts. Osteoclasts were stained for TRAcP (tartrate resistant acid phosphatase), cultured to assess resorptive activity, and analyzed for specific gene expression. Marrow stromal cells or primary neonatal calvarial cells were differentiated to osteoblasts, and osteoblastic gene expression as well as mineralization was analyzed. There was increased osteoclastogenesis and increased resorption pit formation in GHS compared with SD cultures. Osteoclasts had increased expression of cathepsin K, Tracp, and MMP9 in cells from GHS compared with SD rats. Osteoblastic gene expression and mineralization was significantly decreased. Thus, alterations in baseline activity of both osteoclasts and osteoblasts in GHS rats, led to decreased BMD and bone quality, perhaps because of their known increase in vitamin D receptors. Better understanding of the role of GHS bone cells in decreased BMD and quality may provide new strategies to mitigate the low BMD and increased fracture risk found in patients with IH. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nancy S Krieger
- Division of NephrologyUniversity of Rochester School of MedicineRochesterNYUSA
| | - Luojing Chen
- Division of NephrologyUniversity of Rochester School of MedicineRochesterNYUSA
| | - Jennifer Becker
- Division of NephrologyUniversity of Rochester School of MedicineRochesterNYUSA
| | - Sean DeBoyace
- Division of NephrologyUniversity of Rochester School of MedicineRochesterNYUSA
| | - Hongwei Wang
- Section of EndocrinologyUniversity of Chicago Pritzker School of MedicineChicagoILUSA
| | - Murray J Favus
- Section of EndocrinologyUniversity of Chicago Pritzker School of MedicineChicagoILUSA
| | - David A Bushinsky
- Division of NephrologyUniversity of Rochester School of MedicineRochesterNYUSA
| |
Collapse
|