1
|
Ko PY, Hsu CC, Chen SY, Hsu CH, Li CL, Jou IM, Wu PT. Pulsed Nd:YAG laser therapy accelerates fracture healing in a rat femoral osteotomy model. Bone Joint Res 2025; 14:376-388. [PMID: 40312041 PMCID: PMC12045664 DOI: 10.1302/2046-3758.145.bjr-2024-0285.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
Aims This study aimed to evaluate the effects of Nd:YAG laser treatment on fracture healing in a rat model. We hypothesized that laser therapy would accelerate healing by stimulating early neovascularization and osteoblast recruitment. Methods A total of 54 male Sprague-Dawley rats received intramedullary Kirschner wire (K-wire) osteosynthesis following femoral osteotomy, and were randomly divided into two groups (n = 27 each): the control group, and the laser group that received daily pulsed Nd:YAG laser for ten days immediately after osteotomy. Fracture sites were assessed using micro-CT (μCT; n = 8 at each timepoint), histology (n = 4), and three-point bending tests (n = 4) at week 2, week 4, and week 6, respectively. At week 2, an additional three rats per group were selected for the western blot tests. Results Compared to controls, the laser group showed higher vascular endothelial growth factor (VEGF), CD31, and Runx2 protein expression, and significantly higher neovascular area density and osteoblast density (p = 0.025 and p = 0.008, respectively) at week 2. At week 4, the laser treatment led to higher histological fracture healing scale and flexural modulus, and less strain (p = 0.001, p = 0.020, and p = 0.004, respectively). Macroscopically, the laser group showed higher mature bone volume fraction and radiological union score at weeks 4 and 6 (volume fraction: p = 0.017 and p = 0.001; union score: p = 0.001 and p = 0.024, respectively). Conclusion Pulsed Nd:YAG laser therapy accelerates multiple quantitative indicators of fracture healing within six weeks in a rat femoral osteotomy model, which was associated with enhanced angiogenesis and osteogenesis during the early healing phase.
Collapse
Affiliation(s)
- Po-Yen Ko
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Che-Chia Hsu
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yao Chen
- Department of Nursing, College of Nursing, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Chieh-Hsiang Hsu
- Department of Occupational Therapy, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Lung Li
- Department of Orthopedics, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, E-Da Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- GEG Orthopedic Clinic, Tainan, Taiwan
| | - Po-Ting Wu
- Department of Orthopedics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Orthopedics, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
2
|
Yu X, Kawakami R, Yambe S, Yoshimoto Y, Sasaki T, Higuchi S, Watanabe H, Akiyama H, Miura S, Hu K, Kondoh G, Sagasaki R, Inui M, Adachi T, Docheva D, Imamura T, Shukunami C. Dynamic interactions between cartilaginous and tendinous/ligamentous primordia during musculoskeletal integration. Development 2025; 152:dev204512. [PMID: 40135875 DOI: 10.1242/dev.204512] [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: 06/12/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Proper connections between cartilaginous and muscular primordia through tendinous/ligamentous primordia are essential for musculoskeletal integration. Herein, we report a novel double-reporter mouse model for investigating this process via fluorescently visualising scleraxis (Scx) and SRY-box containing gene 9 (Sox9) expression. We generated ScxTomato transgenic mice and crossed them with Sox9EGFP knock-in mice to obtain ScxTomato;Sox9EGFP mice. Deep imaging of optically cleared double-reporter embryos at E13.5 and E16.5 revealed previously unknown differences in the dynamic interactions between cartilaginous and tendinous/ligamentous primordia in control and Scx-deficient mice. Tendon/ligament maturation was evaluated through simultaneous detection of fluorescence and visualisation of collagen fibre formation using second harmonic generation imaging. Lack of deltoid tuberosity in Scx-deficient mice caused misdirected muscle attachment with morphological changes. Loss of Scx also dysregulated progenitor cell fate determination in the chondrotendinous junction, resulting in the formation of a rounded enthesis rather than the protruding enthesis observed in the control. Hence, our double-reporter mouse system, in combination with loss- or gain-of-function approaches, is a unique and powerful tool that could be used to gain a comprehensive understanding of musculoskeletal integration.
Collapse
Affiliation(s)
- Xinyi Yu
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan
| | - Shinsei Yambe
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takako Sasaki
- Department of Biochemistry II, Faculty of Medicine, Oita University, Oita 879-5593, Japan
| | - Shinnosuke Higuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Haruhiko Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu 501-1193, Japan
| | - Shigenori Miura
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kadi Hu
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ramu Sagasaki
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue Regeneration, Orthopaedic Hospital König-Ludwig-Haus, University of Würzburg, 97074 Würzburg, Germany
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime 791-0295, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
3
|
Jiang C, Miao T, Xing X, Schilling KJ, Lenhard N, Wang L, McDowell S, Nilsson BL, Wang H, Zhang X. Masquelet Inspired in Vivo Engineered Extracellular Matrix as Functional Periosteum for Bone Defect Repair and Reconstruction. Adv Healthc Mater 2025; 14:e2404975. [PMID: 39840608 PMCID: PMC11913577 DOI: 10.1002/adhm.202404975] [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: 12/10/2024] [Revised: 01/09/2025] [Indexed: 01/23/2025]
Abstract
The Masquelet technique that combines a foreign body reaction (FBR)-induced vascularized tissue membrane with staged bone grafting for reconstruction of segmental bone defect has gained wide attention in Orthopedic surgery. The success of Masquelet hinges on its ability to promote formation of a "periosteum-like" FBR-induced membrane at the bone defect site. Inspired by Masquelet's technique, here a novel approach is devised to create periosteum mimetics from decellularized extracellular matrix (dECM), engineered in vivo through FBR, for reconstruction of segmental bone defects. The approach involved 3D printing of polylactic acid (PLA) template with desired pattern/architecture, followed by subcutaneous implantation of the template to form tissue, and depolymerization and decellularization to generate dECM with interconnected channels. The dECM matrices produces from the same mice (autologous) or from different mice (allogenic) are used as a functional periosteum for repair of structural bone allograft in a murine segmental bone defect model. This study shows that autologous dECM performed better than allogenic dECM, further permitting local delivery of low dose BMP-2 to enhance allograft incorporation. The success of this current approach can establish a new line of versatile, patient-specific, and periosteum-like autologous dECM for bone regeneration, offering personalized therapeutics to patients with impaired healing.
Collapse
Affiliation(s)
- Chen Jiang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Tianfeng Miao
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Kevin J Schilling
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Nicholas Lenhard
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Lichen Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Susan McDowell
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, NY, 14627, USA
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| |
Collapse
|
4
|
Fujii Y, Okabe I, Hatori A, Sah SK, Kanaujiya J, Fisher M, Norris R, Terasaki M, Reichenberger EJ, Chen IP. Skeletal abnormalities caused by a Connexin43 R239Q mutation in a mouse model for autosomal recessive craniometaphyseal dysplasia. Bone Res 2025; 13:14. [PMID: 39848944 PMCID: PMC11757998 DOI: 10.1038/s41413-024-00383-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/04/2024] [Accepted: 10/15/2024] [Indexed: 01/30/2025] Open
Abstract
Craniometaphyseal dysplasia (CMD), a rare craniotubular disorder, occurs in an autosomal dominant (AD) or autosomal recessive (AR) form. CMD is characterized by hyperostosis of craniofacial bones and metaphyseal flaring of long bones. Many patients with CMD suffer from neurological symptoms. The pathogenesis of CMD is not fully understood. Treatment is limited to craniofacial surgery. Here, we report a knock in (KI) mouse model for AR CMD carrying a Cx43R239Q mutation. Cx43KI/KI mice replicate typical features of AR CMD, including thickening of craniofacial bones, club-shaped femurs, and widened diaphyseal cortical bones. Female Cx43KI/KI mice display remarkably more bone overgrowth than male Cx43KI/KI mice as they age. In contrast to Cx43+/+ littermates, Cx43KI/KI mice exhibit periosteal bone deposition and increased osteoclast (OC) numbers in the endosteum of long bones. Although formation of resting OCs in Cx43+/+ and Cx43KI/KI mice is comparable, the actively resorbing Cx43KI/KI OCs have reduced resorption on bone chips. Cx43KI/KI mice display reduced osteocyte dendrites. RNA from Cx43KI/KI femoral cortical bones show reduced expression levels of Sost, Tnf-α, IL-1β, Esr1, Esr2, and a lower Rankl/Opg ratio. Moreover, the Cx43R239Q mutation results in altered spatial expression of Cx43 protein and mild reduction of gap junction and hemichannel activity. The distinct phenotype seen in Cx43KI/KI mice but not in Cx43 ablation models suggests that Cx43 loss-of-function is unlikely the main cause of AR CMD. Additional studies are required to investigate new roles of CMD-mutant Cx43.
Collapse
Affiliation(s)
- Yasuyuki Fujii
- Department of Endodontology, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| | - Iichiro Okabe
- Department of Endodontology, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| | - Ayano Hatori
- Department of Endodontology, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| | - Shyam Kishor Sah
- Department of Endodontology, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| | - Jitendra Kanaujiya
- Department of Cell Biology, University of Connecticut Health, Farmington, CT, USA
| | - Melanie Fisher
- Department of Cell Biology, University of Connecticut Health, Farmington, CT, USA
| | - Rachael Norris
- Department of Cell Biology, University of Connecticut Health, Farmington, CT, USA
| | - Mark Terasaki
- Department of Cell Biology, University of Connecticut Health, Farmington, CT, USA
| | - Ernst J Reichenberger
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA
| | - I-Ping Chen
- Department of Endodontology, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA.
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA.
| |
Collapse
|
5
|
Zhai Y, Zhou Z, Xing X, Nuzzle M, Zhang X. Differential bone and vessel type formation at superior and dura periosteum during cranial bone defect repair. Bone Res 2025; 13:8. [PMID: 39805832 PMCID: PMC11729862 DOI: 10.1038/s41413-024-00379-9] [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: 04/17/2024] [Revised: 08/14/2024] [Accepted: 10/09/2024] [Indexed: 01/16/2025] Open
Abstract
The cranial mesenchyme, originating from both neural crest and mesoderm, imparts remarkable regional specificity and complexity to postnatal calvarial tissue. While the distinct embryonic origins of the superior and dura periosteum of the cranial parietal bone have been described, the extent of their respective contributions to bone and vessel formation during adult bone defect repair remains superficially explored. Utilizing transgenic mouse models in conjunction with high-resolution multiphoton laser scanning microscopy (MPLSM), we have separately evaluated bone and vessel formation in the superior and dura periosteum before and after injury, as well as following intermittent treatment of recombinant peptide of human parathyroid hormone (rhPTH), Teriparatide. Our results show that new bone formation along the dura surface is three times greater than that along the superior periosteal surface following injury, regardless of Teriparatide treatment. Targeted deletion of PTH receptor PTH1R via SMA-CreER and Col 1a (2.3)-CreER results in selective reduction of bone formation, suggesting different progenitor cell pools in the adult superior and dura periosteum. Consistently, analyses of microvasculature show higher vessel density and better organized arterial-venous vessel network associated with a 10-fold more osteoblast clusters at dura periosteum as compared to superior periosteum. Intermittent rhPTH treatment further enhances the arterial vessel ratio at dura periosteum and type H vessel formation in cortical bone marrow space. Taken together, our study demonstrates a site-dependent coordinated osteogenic and angiogenic response, which is determined by regional osteogenic progenitor pool as well as the coupling blood vessel network at the site of cranial defect repair.
Collapse
Affiliation(s)
- Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xiaojie Xing
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Mark Nuzzle
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
6
|
Thakore P, Karki S, Hrdlicka HC, Garcia-Munoz J, Pereira RC, Delany AM. Decreasing miR-433-3p Activity in the Osteoblast Lineage Blunts Glucocorticoid-mediated Bone Loss. Endocrinology 2025; 166:bqaf008. [PMID: 39820728 PMCID: PMC11791524 DOI: 10.1210/endocr/bqaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
Glucocorticoid excess causes bone loss due to decreased bone formation and increased bone resorption; miR-433-3p is a microRNA (miRNA) that negatively regulates bone formation in male mice by targeting Runx2 as well as RNAs involved in Wnt, protein kinase A, and endogenous glucocorticoid signaling. To examine the impact of miR-433-3p on glucocorticoid-mediated bone loss, transgenic mice expressing a miR-433-3p tough decoy inhibitor in the osteoblast lineage were administered prednisolone via slow-release pellets. Bone loss was greater in control mice treated with prednisolone compared with miR-433-3p tough decoy mice due to higher osteoclast activity in the controls. In whole femurs, Rankl was significantly higher in prednisolone-treated controls compared with miR-433-3p tough decoy mice. Surprisingly, negative regulators of Wnt signaling Sost and Dkk1 were higher in miR-433-3p tough decoy mice and were unaffected by prednisolone. Luciferase- 3'-untranslated region reporter assays demonstrated that Sost is a novel miR-433-3p target, whereas Dkk1 is a previously validated miR-433-3p target. miR-433-3p levels are lower in matrix-synthesizing osteoblasts than in more osteocytic cells; thus the impact of miR-433-3p on the osteoblast lineage may be dependent on cell context: it is a negative regulator in matrix-depositing osteoblasts by targeting RNAs important for differentiation and function but a positive regulator in osteocytes, due to its ability to target prominently expressed negative regulators of Wnt signaling, Sost and Dkk1. The mechanisms by which miR-433-3p indirectly regulates glucocorticoid-mediated osteoclastogenesis remain unknown. However, we speculate that this regulation may be mediated by miR-433-3p activity in osteocytes, which play an important role in controlling osteoclastogenesis.
Collapse
Affiliation(s)
- Prachi Thakore
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Henry C Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - John Garcia-Munoz
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| | - Renata C Pereira
- Division of Pediatric Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
7
|
Minciacchi VR, Bravo J, Karantanou C, Pereira RS, Zanetti C, Kumar R, Thomasberger N, Llavona P, Krack T, Bankov K, Meister M, Hartmann S, Maguer-Satta V, Lefort S, Putyrski M, Ernst A, Huntly BJP, Meduri E, Ruf W, Krause DS. Exploitation of the fibrinolytic system by B-cell acute lymphoblastic leukemia and its therapeutic targeting. Nat Commun 2024; 15:10059. [PMID: 39567540 PMCID: PMC11579293 DOI: 10.1038/s41467-024-54361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Fibrinolysis influences the mobilization of hematopoietic stem cells from their bone marrow microenvironment (BMM). Here we show that activation of plasmin, a key fibrinolytic agent, by annexin A2 (ANXA2) distinctly impacts progression of BCR-ABL1+ B-cell acute lymphoblastic leukemia (B-ALL) via modulation of the extracellular matrix (ECM) in the BMM. The dense ECM in a BMM with decreased plasmin activity entraps insulin-like growth factor (IGF) 1 and reduces mTORC2-dependent signaling and proliferation of B-ALL cells. Conversely, B-ALL conditions the BMM to induce hepatic generation of plasminogen, the plasmin precursor. Treatment with ε-aminocaproic acid (EACA), which inhibits plasmin activation, reduces tumor burden and prolongs survival, including in xenogeneic models via increased fibronectin in the BMM. Human data confirm that IGF1 and fibronectin staining in trephine biopsies are correlated. Our studies suggest that fibrinolysis-mediated ECM remodeling and subsequent growth factor release influence B-ALL progression and inhibition of this process by EACA may be beneficial as adjunct therapy.
Collapse
Affiliation(s)
- Valentina R Minciacchi
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131, Mainz, Germany
| | - Jimena Bravo
- Institute of Transfusion Medicine - Transfusion Center, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany
| | - Christina Karantanou
- Department of Vascular Dysfunction - Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Raquel S Pereira
- Institute for Experimental Pediatric Hematology and Oncology, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Costanza Zanetti
- Division of mRNA Cancer Immunotherapy, Helmholtz Institute for Translational Oncology Mainz, Mainz, Germany
| | - Rahul Kumar
- Institute of Transfusion Medicine - Transfusion Center, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany
| | | | | | - Theresa Krack
- Institute of Transfusion Medicine - Transfusion Center, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany
| | - Katrin Bankov
- Department of Pediatrics (Hematology/Oncology), Charité-Universitätsmedizin, Berlin, Germany
| | | | - Sylvia Hartmann
- Department of Pathology, Goethe University, Frankfurt am Main, Germany
| | | | - Sylvain Lefort
- CRCL, Inserm U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Mateusz Putyrski
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, Frankfurt am Main, Germany
| | - Andreas Ernst
- Pharmazentrum/ZAFES Frankfurt, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Brian J P Huntly
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Eshwar Meduri
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA, USA
| | - Daniela S Krause
- Institute of Transfusion Medicine - Transfusion Center, Johannes Gutenberg University Medical Center, 55131, Mainz, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center, University of Mainz, Mainz, Germany.
| |
Collapse
|
8
|
Alshaweesh J, Dash R, Lee MSJ, Kahyaoglu P, Erci E, Xu M, Matsuo-Dapaah J, Del Rosario Zorrilla C, Aykac K, Ekemen S, Kobiyama K, Ishii KJ, Coban C. MyD88 in osteoclast and osteoblast lineages differentially controls bone remodeling in homeostasis and malaria. Int Immunol 2024; 36:451-464. [PMID: 38642134 PMCID: PMC11319481 DOI: 10.1093/intimm/dxae023] [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: 12/21/2023] [Accepted: 04/16/2024] [Indexed: 04/22/2024] Open
Abstract
Chronic bone loss is an under-recognized complication of malaria, the underlying mechanism of which remains incompletely understood. We have previously shown that persistent accumulation of Plasmodium products in the bone marrow leads to chronic inflammation in osteoblast (OB) and osteoclast (OC) precursors causing bone loss through MyD88, an adaptor molecule for diverse inflammatory signals. However, the specific contribution of MyD88 signaling in OB or OC precursors in malaria-induced bone loss remains elusive. To assess the direct cell-intrinsic role of MyD88 signaling in adult bone metabolism under physiological and infection conditions, we used the Lox-Cre system to specifically deplete MyD88 in the OB or OC lineages. Mice lacking MyD88 primarily in the maturing OBs showed a comparable decrease in trabecular bone density by microcomputed tomography to that of controls after Plasmodium yoelii non-lethal infection. In contrast, mice lacking MyD88 in OC precursors showed significantly less trabecular bone loss than controls, suggesting that malaria-mediated inflammatory mediators are primarily controlled by MyD88 in the OC lineage. Surprisingly, however, depletion of MyD88 in OB, but not in OC, precursors resulted in reduced bone mass with decreased bone formation rates in the trabecular areas of femurs under physiological conditions. Notably, insulin-like growth factor-1, a key molecule for OB differentiation, was significantly lower locally and systemically when MyD88 was depleted in OBs. Thus, our data demonstrate an indispensable intrinsic role for MyD88 signaling in OB differentiation and bone formation, while MyD88 signaling in OC lineages plays a partial role in controlling malaria-induced inflammatory mediators and following bone pathology. These findings may lead to the identification of novel targets for specific intervention of bone pathologies, particularly in malaria-endemic regions.
Collapse
Affiliation(s)
- Jalal Alshaweesh
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- International Vaccine Design Center, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
| | - Rashmi Dash
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- Department of Computational Biology and Medical Science (CBMS), Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
| | - Michelle S J Lee
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- International Vaccine Design Center, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
| | - Pinar Kahyaoglu
- Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
- Department of Paediatrics, Hacettepe University School of Medicine, Ankara 06100, Turkey
| | - Ece Erci
- Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
- Department of Paediatrics, Hacettepe University School of Medicine, Ankara 06100, Turkey
| | - Mengling Xu
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- Department of Computational Biology and Medical Science (CBMS), Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
| | - Julia Matsuo-Dapaah
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Camila Del Rosario Zorrilla
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- Department of Computational Biology and Medical Science (CBMS), Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
| | - Kubra Aykac
- Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
- Department of Paediatrics, Hacettepe University School of Medicine, Ankara 06100, Turkey
- Ministry of Health University, Ankara Education and Research Hospital, Paediatric Infectious Diseases Unit, Ankara 06230, Turkey
| | - Suheyla Ekemen
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
| | - Kouji Kobiyama
- International Vaccine Design Center, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Division of Vaccine Science, Department of Microbiology and Immunology, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ken J Ishii
- International Vaccine Design Center, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
- Division of Vaccine Science, Department of Microbiology and Immunology, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
| | - Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science (IMSUT), The University of Tokyo, Tokyo 108-8639, Japan
- International Vaccine Design Center, IMSUT, The University of Tokyo, Tokyo 108-8639, Japan
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Department of Computational Biology and Medical Science (CBMS), Graduate School of Frontier Sciences, University of Tokyo, Tokyo 108-8639, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
9
|
Janssen A, Buschang PH, Tadlock LP, Kesterke MJ, Jing Y. The effects of dietary loading on the transdifferentiation of condylar chondrocytes. Am J Orthod Dentofacial Orthop 2024; 165:697-710. [PMID: 38573296 DOI: 10.1016/j.ajodo.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Transdifferentiation of chondrocytes into bone cells explains most condylar growth during prenatal and early postnatal stages, but the mechanisms regulating chondrocyte transdifferentiation during late postnatal growth remain unknown. This study aimed to quantify the effects of dietary loading on chondrocyte-derived osteogenesis during late postnatal condylar growth. METHODS Two compound mouse lines were used to trace the fate of chondrocyte lineage in vivo. Twelve 3-week-old male Aggrecan-CreERT2 (AcanLineage); R26RTdTomato; 2.3 Col10a1-GFP and twelve 3-week-old male Col10a1-Cre (Col10a1Lineage); R26RTdTomato; 2.3Col1a1-GFP were randomly divided into experimental (soft-food diet, n = 6) and control (hard-food diet, n = 6) groups and kept for 6 weeks. One time, tamoxifen injections were given to AcanLineage mice at 3 weeks. Radiographic, microcomputed tomographic, and histomorphometric analyses were performed. RESULTS Radiologic analysis showed that mice with a soft-food diet had smaller mandible lengths as well as decreased bone volume and density for their condylar process. Histologically, mice with soft diets had reduced activity in chondrocyte proliferation and maturation compared with the controls. Cell lineage tracing results showed the number of AcanLineage-derived bone cells (293.8 ± 39.8 vs 207.1 ± 44.6; P = 0.005), as well as total bone cells (445.6 ± 31.7 vs 360.7 ± 46.9; P = 0.004), was significantly higher in the hard-diet group than in the soft-diet group, whereas the number of non-AcanLineage-derived bone cells was not significantly different among groups (P = 0.938). Col10a1Lineage mice showed the same trend. CONCLUSIONS Dietary loading directly affects condyle chondrogenesis and chondrocyte transdifferentiation, which alters the extent of condylar growth and remodeling.
Collapse
Affiliation(s)
- Abbey Janssen
- Department of Orthodontics, Texas A&M School of Dentistry, Dallas, Tex
| | - Peter H Buschang
- Department of Orthodontics, Texas A&M School of Dentistry, Dallas, Tex
| | - Larry P Tadlock
- Department of Orthodontics, Texas A&M School of Dentistry, Dallas, Tex
| | | | - Yan Jing
- Department of Orthodontics, Texas A&M School of Dentistry, Dallas, Tex.
| |
Collapse
|
10
|
Novak S, Tanigawa H, Singh V, Root SH, Schmidt TA, Hankenson KD, Kalajzic I. Endothelial to mesenchymal Notch signaling regulates skeletal repair. JCI Insight 2024; 9:e181073. [PMID: 38781018 PMCID: PMC11383173 DOI: 10.1172/jci.insight.181073] [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: 03/12/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
We present a transcriptomic analysis that provides a better understanding of regulatory mechanisms within the healthy and injured periosteum. The focus of this work is on characterizing early events controlling bone healing during formation of periosteal callus on day 3 after fracture. Building on our previous findings showing that induced Notch1 signaling in osteoprogenitors leads to better healing, we compared samples in which the Notch 1 intracellular domain is overexpressed by periosteal stem/progenitor cells, with control intact and fractured periosteum. Molecular mechanisms and changes in skeletal stem/progenitor cells (SSPCs) and other cell populations within the callus, including hematopoietic lineages, were determined. Notably, Notch ligands were differentially expressed in endothelial and mesenchymal populations, with Dll4 restricted to endothelial cells, whereas Jag1 was expressed by mesenchymal populations. Targeted deletion of Dll4 in endothelial cells using Cdh5CreER resulted in negative effects on early fracture healing, while deletion in SSPCs using α-smooth muscle actin-CreER did not impact bone healing. Translating these observations into a clinically relevant model of bone healing revealed the beneficial effects of delivering Notch ligands alongside the osteogenic inducer, BMP2. These findings provide insights into the regulatory mechanisms within the healthy and injured periosteum, paving the way for novel translational approaches to bone healing.
Collapse
Affiliation(s)
- Sanja Novak
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Hitoshi Tanigawa
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Vijender Singh
- Institute for Systems Genomics, Computational Biology Core, UConn, Storrs, Connecticut, USA
| | - Sierra H Root
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Tannin A Schmidt
- Biomedical Engineering Department, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ivo Kalajzic
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
11
|
Liu G, Wei J, Xiao W, Xie W, Ru Q, Chen L, Wu Y, Mobasheri A, Li Y. Insights into the Notch signaling pathway in degenerative musculoskeletal disorders: Mechanisms and perspectives. Biomed Pharmacother 2023; 169:115884. [PMID: 37981460 DOI: 10.1016/j.biopha.2023.115884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Degenerative musculoskeletal disorders are a group of age-related diseases of the locomotive system that severely affects the patient's ability to work and cause adverse sequalae such as fractures and even death. The incidence and prevalence of degenerative musculoskeletal disorders is rising owing to the aging of the world's population. The Notch signaling pathway, which is expressed in almost all organ systems, extensively regulates cell proliferation and differentiation as well as cellular fate. Notch signaling shows increased activity in degenerative musculoskeletal disorders and retards the progression of degeneration to some extent. The review focuses on four major degenerative musculoskeletal disorders (osteoarthritis, intervertebral disc degeneration, osteoporosis, and sarcopenia) and summarizes the pathophysiological functions of Notch signaling in these disorders, especially its role in stem/progenitor cells in each disorder. Finally, a conclusion will be presented to explore the research and application of the perspectives on Notch signaling in degenerative musculoskeletal disorders.
Collapse
Affiliation(s)
- Gaoming Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jun Wei
- Department of Clinical Medical School, Xinjiang Medical University, Urumqi 830054, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China; World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410011, China; Department of Clinical Medical School, Xinjiang Medical University, Urumqi 830054, China.
| |
Collapse
|
12
|
Bhattacharyya ND, Kyaw W, McDonald MM, Dhenni R, Grootveld AK, Xiao Y, Chai R, Khoo WH, Danserau LC, Sergio CM, Timpson P, Lee WM, Croucher PI, Phan TG. Minimally invasive longitudinal intravital imaging of cellular dynamics in intact long bone. Nat Protoc 2023; 18:3856-3880. [PMID: 37857852 DOI: 10.1038/s41596-023-00894-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/28/2023] [Indexed: 10/21/2023]
Abstract
Intravital two-photon microscopy enables deep-tissue imaging at high temporospatial resolution in live animals. However, the endosteal bone compartment and underlying bone marrow pose unique challenges to optical imaging as light is absorbed, scattered and dispersed by thick mineralized bone matrix and the adipose-rich bone marrow. Early bone intravital imaging methods exploited gaps in the cranial sutures to bypass the need to penetrate through cortical bone. More recently, investigators have developed invasive methods to thin the cortical bone or implant imaging windows to image cellular dynamics in weight-bearing long bones. Here, we provide a step-by-step procedure for the preparation of animals for minimally invasive, nondestructive, longitudinal intravital imaging of the murine tibia. This method involves the use of mixed bone marrow radiation chimeras to unambiguously double-label osteoclasts and osteomorphs. The tibia is exposed by a simple skin incision and an imaging chamber constructed using thermoconductive T-putty. Imaging sessions up to 12 h long can be repeated over multiple timepoints to provide a longitudinal time window into the endosteal and marrow niches. The approach can be used to investigate cellular dynamics in bone remodeling, cancer cell life cycle and hematopoiesis, as well as long-lived humoral and cellular immunity. The procedure requires an hour to complete and is suitable for users with minimal prior expertise in small animal surgery.
Collapse
Affiliation(s)
- Nayan Deger Bhattacharyya
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Wunna Kyaw
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle M McDonald
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Rama Dhenni
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Abigail K Grootveld
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ya Xiao
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ryan Chai
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Weng Hua Khoo
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Linda C Danserau
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - C Marcelo Sergio
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Woei Ming Lee
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, New South Wales, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia.
| |
Collapse
|
13
|
Huang AH, Galloway JL. Current and emerging technologies for defining and validating tendon cell fate. J Orthop Res 2023; 41:2082-2092. [PMID: 37211925 DOI: 10.1002/jor.25632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
The tendon field has been flourishing in recent years with the advent of new tools and model systems. The recent ORS 2022 Tendon Section Conference brought together researchers from diverse disciplines and backgrounds, showcasing studies in biomechanics and tissue engineering to cell and developmental biology and using models from zebrafish and mouse to humans. This perspective aims to summarize progress in tendon research as it pertains to understanding and studying tendon cell fate. The successful integration of new technologies and approaches have the potential to further propel tendon research into a new renaissance of discovery. However, there are also limitations with the current methodologies that are important to consider when tackling research questions. Altogether, we will highlight recent advances and technologies and propose new avenues to explore tendon biology.
Collapse
Affiliation(s)
- Alice H Huang
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - Jenna L Galloway
- Department of Orthopaedic Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Zhu H, Su Y, Wang J, Wu JY. The role of vesicle trafficking genes in osteoblast differentiation and function. Sci Rep 2023; 13:16079. [PMID: 37752218 PMCID: PMC10522589 DOI: 10.1038/s41598-023-43116-8] [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: 05/05/2023] [Accepted: 09/20/2023] [Indexed: 09/28/2023] Open
Abstract
Using Col2.3GFP transgenic mice expressing GFP in maturing osteoblasts, we isolated Col2.3GFP+ enriched osteoblasts from 3 sources. We performed RNA-sequencing, identified 593 overlapping genes and confirmed these genes are highly enriched in osteoblast differentiation and bone mineralization annotation categories. The top 3 annotations are all associated with endoplasmic reticulum and Golgi vesicle transport. We selected 22 trafficking genes that have not been well characterized in bone for functional validation in MC3T3-E1 pre-osteoblasts. Transient siRNA knockdown of trafficking genes including Sec24d, Gosr2, Rab2a, Stx5a, Bet1, Preb, Arf4, Ramp1, Cog6 and Pacs1 significantly increased mineralized nodule formation and expression of osteoblast markers. Increased mineralized nodule formation was suppressed by concurrent knockdown of P4ha1 and/or P4ha2, encoding collagen prolyl 4-hydroxylase isoenzymes. MC3T3-E1 pre-osteoblasts with knockdown of Cog6, Gosr2, Pacs1 or Arf4 formed more and larger ectopic mineralized bone nodules in vivo, which was attenuated by concurrent knockdown P4ha2. Permanent knockdown of Cog6 and Pacs1 by CRISPR/Cas9 gene editing in MC3T3-E1 pre-osteoblasts recapitulated increased mineralized nodule formation and osteoblast differentiation. In summary, we have identified several vesicle trafficking genes with roles in osteoblast function. Our findings provide potential targets for regulating bone formation.
Collapse
Affiliation(s)
- Hui Zhu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yingying Su
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jamie Wang
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joy Y Wu
- Division of Endocrinology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
15
|
Gross A, Buschang PH, Shakya A, Jing Y. Short-term effects of mechanical loading on the transdifferentiation of condylar chondrocytes. Am J Orthod Dentofacial Orthop 2023; 164:201-214. [PMID: 36922241 PMCID: PMC10659147 DOI: 10.1016/j.ajodo.2022.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 03/14/2023]
Abstract
INTRODUCTION Transdifferentiation of chondrocytes into bone cells explains most of the prenatal and early postnatal condylar growth, but its role during later postnatal growth and the mechanisms regulating transdifferentiation remain unknown. This study aimed to quantify the effects of mechanical loading on chondrocyte-derived osteogenesis during late postnatal condylar growth using a short-term mandibular laterotrusion model. METHODS Thirty 4-week-old Aggrecan-CreERT2, R26RtdTomato, and 2.3Col1a1-GFP compound mice received tamoxifen injections and were divided into control and experimental groups. Appliances were bonded to shift the mandibles of the experimental mice for 5 days, causing protrusion and retrusion of the right and left condyles, respectively. Radiographic, microcomputed tomographic, and histomorphometric analyses were performed. RESULTS The experimental and control groups showed substantial transdifferentiation of chondrocytes into bone cells. The experimental mice developed asymmetric mandibles, with the protrusive side significantly longer than the retrusive side. The protrusive condyles showed significantly increased chondrogenesis and greater numbers of chondrocyte-derived osteogenic cells, especially in the posterior third. The opposite effects were seen on the retrusive side. CONCLUSIONS Transdifferentiation of chondrocytes into bone cells occurs during late postnatal condylar growth. Laterotrusion regulates condylar chondrogenesis and chondrocyte transdifferentiation, which alters the amount and direction of condylar growth. Our study demonstrated that chondrocytes are key players in condylar bone formation and should be the focus of studies to control and further understand condylar growth.
Collapse
Affiliation(s)
- Amanda Gross
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex
| | - Peter H Buschang
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex
| | - Ajay Shakya
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Tex
| | - Yan Jing
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex.
| |
Collapse
|
16
|
Wu EY, Singh NP, Choi K, Zakeri M, Vincent M, Churchill GA, Ackert-Bicknell CL, Patro R, Love MI. SEESAW: detecting isoform-level allelic imbalance accounting for inferential uncertainty. Genome Biol 2023; 24:165. [PMID: 37438847 PMCID: PMC10337143 DOI: 10.1186/s13059-023-03003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Detecting allelic imbalance at the isoform level requires accounting for inferential uncertainty, caused by multi-mapping of RNA-seq reads. Our proposed method, SEESAW, uses Salmon and Swish to offer analysis at various levels of resolution, including gene, isoform, and aggregating isoforms to groups by transcription start site. The aggregation strategies strengthen the signal for transcripts with high uncertainty. The SEESAW suite of methods is shown to have higher power than other allelic imbalance methods when there is isoform-level allelic imbalance. We also introduce a new test for detecting imbalance that varies across a covariate, such as time.
Collapse
Affiliation(s)
- Euphy Y Wu
- Department of Biostatistics, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Noor P Singh
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | | | - Mohsen Zakeri
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | | | | | - Cheryl L Ackert-Bicknell
- Department of Orthopedics, School of Medicine, University of Colorado, Anschutz Campus, Aurora, CO, USA
| | - Rob Patro
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Michael I Love
- Department of Biostatistics, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
17
|
Kalinina A, Semenova M, Bruter A, Varlamova E, Kubekina M, Pavlenko N, Silaeva Y, Deikin A, Antoshina E, Gorkova T, Trukhanova L, Salmina A, Novikova S, Voronkov D, Kazansky D, Khromykh L. Cyclophilin A as a Pro-Inflammatory Factor Exhibits Embryotoxic and Teratogenic Effects during Fetal Organogenesis. Int J Mol Sci 2023; 24:11279. [PMID: 37511039 PMCID: PMC10380070 DOI: 10.3390/ijms241411279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The precise balance of Th1, Th2, and Th17 cytokines is a key factor in successful pregnancy and normal embryonic development. However, to date, not all humoral factors that regulate and influence physiological pregnancy have been completely studied. Our data here pointed out cyclophilin A (CypA) as the adverse pro-inflammatory factor negatively affecting fetal development and associated with pregnancy complications. In different mouse models in vivo, we demonstrated dramatic embryotoxicity and teratogenicity of increased CypA levels during pregnancy. Using generated transgenic models, we showed that CypA overexpression in fetal tissues induced the death of all transgenic fetuses and complete miscarriage. Administration of recombinant human CypA in a high dose to pregnant females during fetal organogenesis (6.5-11.5 dpc) exhibited teratogenic effects, causing severe defects in the brain and bone development that could lead to malformations and postnatal behavioral and cognitive disorders in the offspring. Embryotoxic and teratogenic effects could be mediated by CypA-induced up-regulation of M1 macrophage polarization via activation of the STAT1/3 signaling pathways. Here, we propose secreted CypA as a novel marker of complicated pregnancy and a therapeutic target for the correction of pregnancy complications.
Collapse
Affiliation(s)
- Anastasiia Kalinina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Maria Semenova
- Department of Embryology, Faculty of Biology, Moscow State University, 1/12 Leninskie Gory, Moscow 119992, Russia
| | - Alexandra Bruter
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Ekaterina Varlamova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Marina Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Natalia Pavlenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, Moscow 119334, Russia
| | - Yulia Silaeva
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, Moscow 119334, Russia
| | - Alexey Deikin
- United Center for Genetic Technologies, Belgorod State National Research University, 85 Pobedi Street, Belgorod 308001, Russia
| | - Elena Antoshina
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Tatyana Gorkova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Lubov Trukhanova
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Alla Salmina
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Svetlana Novikova
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Dmitry Voronkov
- Research Center of Neurology, 80 Volokolamskoye Shosse, Moscow 125367, Russia
| | - Dmitry Kazansky
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Ludmila Khromykh
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 24 Kashirskoe Shosse, Moscow 115478, Russia
| |
Collapse
|
18
|
Abstract
Although osteoblasts and osteocytes are descended from the same lineage, they each have unique and essential roles in bone. Targeting gene deletion to osteoblasts and osteocytes using the Cre/loxP system has greatly increased our current understanding of how these cells function. Additionally, the use of the Cre/loxP system in conjunction with cell-specific reporters has enabled lineage tracing of these bone cells both in vivo and ex vivo. However, concerns have been raised regarding the specificity of the promoters used and the resulting off-target effects on cells within and outside of the bone. In this review, we have summarized the main mouse models that have been used to determine the functions of specific genes in osteoblasts and osteocytes. We discuss the expression patterns and specificity of the different promoter fragments during osteoblast to osteocyte differentiation in vivo. We also highlight how their expression in non-skeletal tissues may complicate the interpretation of study results. A thorough understanding of when and where these promoters are activated will enable improved study design and greater confidence in data interpretation.
Collapse
Affiliation(s)
- Y Kitase
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America
| | - M Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
19
|
Caligiuri G, Tuveson DA. Activated fibroblasts in cancer: Perspectives and challenges. Cancer Cell 2023; 41:434-449. [PMID: 36917949 PMCID: PMC11022589 DOI: 10.1016/j.ccell.2023.02.015] [Citation(s) in RCA: 147] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/13/2023] [Accepted: 02/13/2023] [Indexed: 03/16/2023]
Abstract
Activated fibroblasts in tumors, or cancer-associated fibroblasts (CAFs), have become a popular research area over the past decade. As important players in many aspects of tumor biology, with functions ranging from collagen deposition to immunosuppression, CAFs have been the target of clinical and pre-clinical studies that have revealed their potential pro- and anti-tumorigenic dichotomy. In this review, we describe the important role of CAFs in the tumor microenvironment and the technological advances that made these discoveries possible, and we detail the models that are currently available for CAF investigation. Additionally, we present evidence to support the value of encompassing CAF investigation as a future therapeutic avenue alongside immune and cancer cells while highlighting the challenges that must be addressed for successful clinical translation of new findings.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
20
|
Bautista CA, Srikumar A, Tichy ED, Qian G, Jiang X, Qin L, Mourkioti F, Dyment NA. CD206+ tendon resident macrophages and their potential crosstalk with fibroblasts and the ECM during tendon growth and maturation. Front Physiol 2023; 14:1122348. [PMID: 36909235 PMCID: PMC9992419 DOI: 10.3389/fphys.2023.1122348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023] Open
Abstract
Resident macrophages exist in a variety of tissues, including tendon, and play context-specific roles in their tissue of residence. In this study, we define the spatiotemporal distribution and phenotypic profile of tendon resident macrophages and their crosstalk with neighboring tendon fibroblasts and the extracellular matrix (ECM) during murine tendon development, growth, and homeostasis. Fluorescent imaging of cryosections revealed that F4/80+ tendon resident macrophages reside adjacent to Col1a1-CFP+ Scx-GFP+ fibroblasts within the tendon fascicle from embryonic development (E15.5) into adulthood (P56). Through flow cytometry and qPCR, we found that these tendon resident macrophages express several well-known macrophage markers, including Adgre1 (F4/80), Mrc1 (CD206), Lyve1, and Folr2, but not Ly-6C, and express the Csf1r-EGFP ("MacGreen") reporter. The proportion of Csf1r-EGFP+ resident macrophages in relation to the total cell number increases markedly during early postnatal growth, while the density of macrophages per mm2 remains constant during this same time frame. Interestingly, proliferation of resident macrophages is higher than adjacent fibroblasts, which likely contributes to this increase in macrophage proportion. The expression profile of tendon resident macrophages also changes with age, with increased pro-inflammatory and anti-inflammatory cytokine expression in P56 compared to P14 macrophages. In addition, the expression profile of limb tendon resident macrophages diverges from that of tail tendon resident macrophages, suggesting differential phenotypes across anatomically and functionally different tendons. As macrophages are known to communicate with adjacent fibroblasts in other tissues, we conducted ligand-receptor analysis and found potential two-way signaling between tendon fibroblasts and resident macrophages. Tendon fibroblasts express high levels of Csf1, which encodes macrophage colony stimulating factor (M-CSF) that acts on the CSF1 receptor (CSF1R) on macrophages. Importantly, Csf1r-expressing resident macrophages preferentially localize to Csf1-expressing fibroblasts, supporting the "nurturing scaffold" model for tendon macrophage patterning. Lastly, we found that tendon resident macrophages express high levels of ECM-related genes, including Mrc1 (mannose receptor), Lyve1 (hyaluronan receptor), Lair1 (type I collagen receptor), Ctss (elastase), and Mmp13 (collagenase), and internalize DQ Collagen in explant cultures. Overall, our study provides insights into the potential roles of tendon resident macrophages in regulating fibroblast phenotype and the ECM during tendon growth.
Collapse
Affiliation(s)
- Catherine A. Bautista
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| | - Anjana Srikumar
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Elisia D. Tichy
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Grace Qian
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| | - Xi Jiang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Ling Qin
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
| | - Foteini Mourkioti
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PA, Philadelphia, PA, United States
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of PA, Philadelphia, PA, United States
| | - Nathaniel A. Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, University of PA, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of PA, Philadelphia, PA, United States
| |
Collapse
|
21
|
García-Recio E, Costela-Ruiz VJ, Illescas-Montes R, Melguizo-Rodríguez L, García-Martínez O, Ruiz C, De Luna-Bertos E. Modulation of Osteogenic Gene Expression by Human Osteoblasts Cultured in the Presence of Bisphenols BPF, BPS, or BPAF. Int J Mol Sci 2023; 24:ijms24054256. [PMID: 36901687 PMCID: PMC10002049 DOI: 10.3390/ijms24054256] [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: 12/17/2022] [Revised: 01/26/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Bone effects attributed to bisphenols (BPs) include the inhibition of growth and differentiation. This study analyzes the effect of BPA analogs (BPS, BPF, and BPAF) on the gene expression of the osteogenic markers RUNX2, osterix (OSX), bone morphogenetic protein-2 (BMP-2), BMP-7, alkaline phosphatase (ALP), collagen-1 (COL-1), and osteocalcin (OSC). Human osteoblasts were obtained by primary culture from bone chips harvested during routine dental work in healthy volunteers and were treated with BPF, BPS, or BPAF for 24 h at doses of 10-5, 10-6, and 10-7 M. Untreated cells were used as controls. Real-time PCR was used to determine the expression of the osteogenic marker genes RUNX2, OSX, BMP-2, BMP-7, ALP, COL-1, and OSC. The expression of all studied markers was inhibited in the presence of each analog; some markers (COL-1; OSC, BMP2) were inhibited at all three doses and others only at the highest doses (10-5 and 10-6 M). Results obtained for the gene expression of osteogenic markers reveal an adverse effect of BPA analogs (BPF, BPS, and BPAF) on the physiology of human osteoblasts. The impact on ALP, COL-1, and OSC synthesis and therefore on bone matrix formation and mineralization is similar to that observed after exposure to BPA. Further research is warranted to determine the possible contribution of BP exposure to the development of bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Enrique García-Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| | - Víctor J. Costela-Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| | - Rebeca Illescas-Montes
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| | - Lucía Melguizo-Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| | - Olga García-Martínez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
- Institute of Neuroscience, University of Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958-243-497
| | - Elvira De Luna-Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain
- Institute of Biosanitary Research, ibs.GRANADA, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2a Planta, 18012 Granada, Spain
| |
Collapse
|
22
|
Novak S, Madunic J, Shum L, Vucetic M, Wang X, Tanigawa H, Ghosh M, Sanjay A, Kalajzic I. PDGF inhibits BMP2-induced bone healing. NPJ Regen Med 2023; 8:3. [PMID: 36631491 PMCID: PMC9834334 DOI: 10.1038/s41536-023-00276-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Bone regeneration depends on a pool of bone/cartilage stem/progenitor cells and signaling mechanisms regulating their differentiation. Using in vitro approach, we have shown that PDGF signaling through PDGFRβ inhibits BMP2-induced osteogenesis, and significantly attenuates expression of BMP2 target genes. We evaluated outcomes of treatment with two anabolic agents, PDGF and BMP2 using different bone healing models. Targeted deletion of PDGFRβ in αSMA osteoprogenitors, led to increased callus bone mass, resulting in improved biomechanical properties of fractures. In critical size bone defects BMP2 treatment increased proportion of osteoprogenitors, while the combined treatment of PDGF BB with BMP2 decreased progenitor number at the injury site. BMP2 treatment induced significant bone formation and increased number of osteoblasts, while in contrast combined treatment with PDGF BB decreased osteoblast numbers. This is in vivo study showing that PDGF inhibits BMP2-induced osteogenesis, but inhibiting PDGF signaling early in healing process does not improve BMP2-induced bone healing.
Collapse
Affiliation(s)
- Sanja Novak
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Josip Madunic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA ,grid.414681.e0000 0004 0452 3941Biochemistry and Organic Analytical Chemistry Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Laura Shum
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Milan Vucetic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Xi Wang
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Hitoshi Tanigawa
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Mallika Ghosh
- grid.208078.50000000419370394Center for Vascular Biology, UConn Health, Farmington, CT USA
| | - Archana Sanjay
- grid.208078.50000000419370394Department of Orthopeadic Surgery, UConn Health, Farmington, CT USA
| | - Ivo Kalajzic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| |
Collapse
|
23
|
Kalyanaraman H, China SP, Cabriales JA, Moininazeri J, Casteel DE, Garcia JJ, Wong VW, Chen A, Sah RL, Boss GR, Pilz RB. Protein Kinase G2 Is Essential for Skeletal Homeostasis and Adaptation to Mechanical Loading in Male but Not Female Mice. J Bone Miner Res 2023; 38:171-185. [PMID: 36371651 PMCID: PMC9825661 DOI: 10.1002/jbmr.4746] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/14/2022]
Abstract
We previously showed that the NO/cGMP/protein kinase G (PKG) signaling pathway positively regulates osteoblast proliferation, differentiation, and survival in vitro, and that cGMP-elevating agents have bone-anabolic effects in mice. Here, we generated mice with an osteoblast-specific (OB) knockout (KO) of type 2 PKG (gene name Prkg2) using a Col1a1(2.3 kb)-Cre driver. Compared to wild type (WT) littermates, 8-week-old male OB Prkg2-KO mice had fewer osteoblasts, reduced bone formation rates, and lower trabecular and cortical bone volumes. Female OB Prkg2-KO littermates showed no bone abnormalities, despite the same degree of PKG2 deficiency in bone. Expression of osteoblast differentiation- and Wnt/β-catenin-related genes was lower in primary osteoblasts and bones of male KO but not female KO mice compared to WT littermates. Osteoclast parameters were unaffected in both sexes. Since PKG2 is part of a mechano-sensitive complex in osteoblast membranes, we examined its role during mechanical loading. Cyclical compression of the tibia increased cortical thickness and induced mechanosensitive and Wnt/β-catenin-related genes to a similar extent in male and female WT mice and female OB Prkg2-KO mice, but loading had a minimal effect in male KO mice. We conclude that PKG2 drives bone acquisition and adaptation to mechanical loading via the Wnt/β-catenin pathway in male mice. The striking sexual dimorphism of OB Prkg2-KO mice suggests that current U.S. Food and Drug Administration-approved cGMP-elevating agents may represent novel effective treatment options for male osteoporosis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These two authors contributed equally to the work
| | - Shyamsundar Pal China
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- These two authors contributed equally to the work
| | - Justin A. Cabriales
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jafar Moininazeri
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Darren E. Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Julian J. Garcia
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Van W. Wong
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Albert Chen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert L. Sah
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gerry R. Boss
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Renate B. Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
24
|
Jeffery EC, Mann TLA, Pool JA, Zhao Z, Morrison SJ. Bone marrow and periosteal skeletal stem/progenitor cells make distinct contributions to bone maintenance and repair. Cell Stem Cell 2022; 29:1547-1561.e6. [PMID: 36272401 DOI: 10.1016/j.stem.2022.10.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 09/15/2022] [Accepted: 10/06/2022] [Indexed: 01/09/2023]
Abstract
A fundamental question in bone biology concerns the contributions of skeletal stem/progenitor cells (SSCs) in the bone marrow versus the periosteum to bone repair. We found that SSCs in adult bone marrow can be identified based on Leprcre and Adiponectin-cre/creER expression while SSCs in adult periosteum can be identified based on Gli1creERT2 expression. Under steady-state conditions, new bone arose primarily from bone marrow SSCs. After bone injuries, both SSC populations began proliferating but made very different contributions to bone repair. Drill injuries were primarily repaired by LepR+/Adiponectin+ bone marrow SSCs. Conversely, bicortical fractures were primarily repaired by Gli1+ periosteal SSCs, though LepR+/Adiponectin+ bone marrow cells transiently formed trabecular bone at the fracture site. Gli1+ periosteal cells also regenerated LepR+ bone marrow stromal cells that expressed hematopoietic niche factors at fracture sites. Different bone injuries are thus repaired by different SSCs, with periosteal cells regenerating bone and marrow stroma after non-stabilized fractures.
Collapse
Affiliation(s)
- Elise C Jeffery
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Terry L A Mann
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jade A Pool
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Children's Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
25
|
Muruganandan S, Pierce R, Teguh DA, Perez RF, Bell N, Nguyen B, Hohl K, Snyder BD, Grinstaff MW, Alberico H, Woods D, Kong Y, Sima C, Bhagat S, Ho K, Rosen V, Gamer L, Ionescu AM. A FoxA2+ long-term stem cell population is necessary for growth plate cartilage regeneration after injury. Nat Commun 2022; 13:2515. [PMID: 35523895 PMCID: PMC9076650 DOI: 10.1038/s41467-022-30247-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/14/2022] [Indexed: 01/14/2023] Open
Abstract
Longitudinal bone growth, achieved through endochondral ossification, is accomplished by a cartilaginous structure, the physis or growth plate, comprised of morphologically distinct zones related to chondrocyte function: resting, proliferating and hypertrophic zones. The resting zone is a stem cell-rich region that gives rise to the growth plate, and exhibits regenerative capabilities in response to injury. We discovered a FoxA2+group of long-term skeletal stem cells, situated at the top of resting zone, adjacent the secondary ossification center, distinct from the previously characterized PTHrP+ stem cells. Compared to PTHrP+ cells, FoxA2+ cells exhibit higher clonogenicity and longevity. FoxA2+ cells exhibit dual osteo-chondro-progenitor activity during early postnatal development (P0-P28) and chondrogenic potential beyond P28. When the growth plate is injured, FoxA2+ cells expand in response to trauma, and produce physeal cartilage for growth plate tissue regeneration.
Collapse
Affiliation(s)
- Shanmugam Muruganandan
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Rachel Pierce
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Dian Astari Teguh
- Centre for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215, USA
| | | | - Nicole Bell
- New York University College of Dentistry, 345 E.24th St, New York, NY, 10010, USA
| | - Brandon Nguyen
- Moderna Therapeutics, One Upland Rd, Norwood, Ohio, MA, 02062, USA
| | - Katherine Hohl
- Centre for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215, USA.,Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, 590 Commonwealth Ave, SCI 518, Boston, MA, 02215, USA
| | - Brian D Snyder
- Department of Orthopedic Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, 590 Commonwealth Ave, SCI 518, Boston, MA, 02215, USA
| | - Hannah Alberico
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Dori Woods
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Yiwei Kong
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Corneliu Sima
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Sanket Bhagat
- Ultragenyx Pharmaceutical, 840 Memorial Drive, Cambridge, MA, 02139, USA
| | - Kailing Ho
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, MA, 02115, USA
| | - Andreia M Ionescu
- Department of Biology, 134 Mugar Life Sciences Building, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Chai RC, McDonald MM. Visualisation of tumour cells in bone in vivo at single-cell resolution. Bone 2022; 158:116113. [PMID: 34273634 DOI: 10.1016/j.bone.2021.116113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/28/2021] [Accepted: 07/09/2021] [Indexed: 12/26/2022]
Abstract
The skeleton is a common site for the establishment of distant metastases. Once cancers occupy bone, the prognosis is poor as disease recurrence and visceral spread is imminent. Understanding the pathways and cellular interactions, which regulate tumour cell seeding, dormancy and growth in bone, is pertinent to improving outcomes for patients with advanced cancers. Advances in imaging techniques have facilitated the development of the concept that the behavior of bone marrow resident cells dictates the fate of tumour cells upon arrival in bone. This review summarises recent findings achieved through intravital imaging. It highlights the importance of developing both longitudinal static and acute dynamic data to develop our understanding of tumour cell engraftment, dormancy, activation and the subsequent establishment of metastases. We also describe how imaging techniques have developed our knowledge of the elements that make up the complex bone microenvironment which tumour cells interact with to survive and grow. We also discuss how through combining these imaging insights with single cell RNA sequencing data, we are entering a new era of research which has the power to define the cell-cell interactions which control tumour cell growth in bone.
Collapse
Affiliation(s)
- Ryan C Chai
- Bone Biology Program, Healthy Ageing Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | - Michelle M McDonald
- Bone Biology Program, Healthy Ageing Theme, Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia.
| |
Collapse
|
27
|
Daneshmandi L, Holt BD, Arnold AM, Laurencin CT, Sydlik SA. Ultra-low binder content 3D printed calcium phosphate graphene scaffolds as resorbable, osteoinductive matrices that support bone formation in vivo. Sci Rep 2022; 12:6960. [PMID: 35484292 PMCID: PMC9050648 DOI: 10.1038/s41598-022-10603-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/02/2022] [Indexed: 12/18/2022] Open
Abstract
Bone regenerative engineering could replace autografts; however, no synthetic material fulfills all design criteria. Nanocarbons incorporated into three-dimensional printed (3DP) matrices can improve properties, but incorporation is constrained to low wt%. Further, unmodified nanocarbons have limited osteogenic potential. Functionalization to calcium phosphate graphene (CaPG) imparts osteoinductivity and osteoconductivity, but loading into matrices remained limited. This work presents ultra-high content (90%), 3DP-CaPG matrices. 3DP-CaPG matrices are highly porous (95%), moderately stiff (3 MPa), and mechanically robust. In vitro, they are cytocompatible and induce osteogenic differentiation of human mesenchymal stem cells (hMSCs), indicated by alkaline phosphatase, mineralization, and COL1α1 expression. In vivo, bone regeneration was studied using a transgenic fluorescent-reporter mouse non-union calvarial defect model. 3DP-CaPG stimulates cellular ingrowth, retains donor cells, and induces osteogenic differentiation. Histology shows TRAP staining around struts, suggesting potential osteoclast activity. Apparent resorption of 3DP-CaPG was observed and presented no toxicity. 3DP-CaPG represents an advancement towards a synthetic bone regeneration matrix.
Collapse
Affiliation(s)
- Leila Daneshmandi
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA
| | - Brian D Holt
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Anne M Arnold
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA
- National Security Directorate, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Cato T Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, 06030, USA.
- Raymond and Beverly Sackler Center for Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT, 06030, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, 06030, USA.
- Department of Material Science and Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Stefanie A Sydlik
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, 5000 Forbes Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
28
|
Florio F, Accordini S, Libergoli M, Biressi S. Targeting Muscle-Resident Single Cells Through in vivo Electro-Enhanced Plasmid Transfer in Healthy and Compromised Skeletal Muscle. Front Physiol 2022; 13:834705. [PMID: 35431987 PMCID: PMC9010744 DOI: 10.3389/fphys.2022.834705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal muscle is composed of syncytial muscle fibers, and by various mononucleated cellular types, such as muscle stem cells, immune cells, interstitial and stromal progenitors. These cell populations play a crucial role during muscle regeneration, and alterations of their phenotypic properties have been associated with defective repair and fibrosis in aging and dystrophic muscle. Studies involving in vivo gene modulation are valuable to investigate the mechanisms underlining cell function and dysfunction in complex pathophysiological settings. Electro-enhanced transfer of plasmids using square-wave generating devices represents a cost-effective approach that is widely used to transport DNA to muscle fibers efficiently. Still, it is not clear if this method can also be applied to mononuclear cells present in muscle. We demonstrate here that it is possible to efficiently deliver DNA into different muscle–resident cell populations in vivo. We evaluated the efficiency of this approach not only in healthy muscle but also in muscles of aging and dystrophic animal models. As an exemplificative application of this method, we used a strategy relying on a reporter gene-based plasmid containing regulatory sequences from the collagen 1 locus, and we determined collagen expression in various cell types reportedly involved in the production of fibrotic tissue in the dystrophic settings. The results enclosed in this manuscript reveal the suitability in applying electro-enhanced transfer of plasmid DNA to mononucleated muscle-resident cells to get insights into the molecular events governing diseased muscle physiology.
Collapse
Affiliation(s)
- Francesca Florio
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Dulbecco Telethon Institute at University of Trento, Trento, Italy
| | - Silvia Accordini
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michela Libergoli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Dulbecco Telethon Institute at University of Trento, Trento, Italy
| | - Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Dulbecco Telethon Institute at University of Trento, Trento, Italy
- *Correspondence: Stefano Biressi,
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Periosteal apposition and endosteal remodeling regulate cortical bone expansion and thickness, both critical determinants of bone strength. Yet, the cellular characteristics and local or paracrine factors that regulate the periosteum and endosteum remain largely elusive. Here we discuss novel insights in cortical bone growth, expansion, and homeostasis, provided by the study of Secreted Frizzled Receptor Protein 4 (Sfrp4), a decoy receptor for Wnt ligands. RECENT FINDINGS SFRP4 loss-of function mutations cause Pyle disease, a rare skeletal disorder characterized by cortical bone thinning and increased fragility fractures despite increased trabecular bone density. On the endosteal surface, Sfrp4-mediated repression of non-canonical Wnt signaling regulates endosteal resorption. On the periosteum, Sfrp4 identifies as a critical functional mediator of periosteal stem cell/progenitor expansion and differentiation. Analysis of signaling pathways regulating skeletal stem cells/progenitors provides an opportunity to advance our understanding of the mechanisms involved in cortical bone biology.
Collapse
Affiliation(s)
- Ruiying Chen
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Roland Baron
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
- Endocrine Unit, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Francesca Gori
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA.
| |
Collapse
|
30
|
Mo C, Guo J, Qin J, Zhang X, Sun Y, Wei H, Cao D, Zhang Y, Zhao C, Xiong Y, Zhang Y, Sun Y, Shen L, Yue R. Single-cell transcriptomics of LepR-positive skeletal cells reveals heterogeneous stress-dependent stem and progenitor pools. EMBO J 2022; 41:e108415. [PMID: 34957577 PMCID: PMC8844986 DOI: 10.15252/embj.2021108415] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
Leptin receptor (LepR)-positive cells are key components of the bone marrow hematopoietic microenvironment, and highly enrich skeletal stem and progenitor cells that maintain homeostasis of the adult skeleton. However, the heterogeneity and lineage hierarchy within this population has been elusive. Using genetic lineage tracing and single-cell RNA sequencing, we found that Lepr-Cre labels most bone marrow stromal cells and osteogenic lineage cells in adult long bones. Integrated analysis of Lepr-Cre-traced cells under homeostatic and stress conditions revealed dynamic changes of the adipogenic, osteogenic, and periosteal lineages. Importantly, we discovered a Notch3+ bone marrow sub-population that is slow-cycling and closely associated with the vasculatures, as well as key transcriptional networks promoting osteo-chondrogenic differentiation. We also identified a Sca-1+ periosteal sub-population with high clonogenic activity but limited osteo-chondrogenic potential. Together, we mapped the transcriptomic landscape of adult LepR+ stem and progenitor cells and uncovered cellular and molecular mechanisms underlying their maintenance and lineage specification.
Collapse
Affiliation(s)
- Chunyang Mo
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Jingxin Guo
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery2nd Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
| | - Jiachen Qin
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Xiaoying Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yuxi Sun
- Department of CardiologyShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Hanjing Wei
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Dandan Cao
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yiying Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Chengchen Zhao
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yanhong Xiong
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yong Zhang
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Yao Sun
- Department of ImplantologySchool & Hospital of StomatologyShanghai Engineering Research Center of Tooth Restoration and RegenerationTongji UniversityShanghaiChina
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences InstituteZhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery2nd Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouChina
- Hangzhou Innovation CenterZhejiang UniversityHangzhouChina
| | - Rui Yue
- Institute for Regenerative MedicineShanghai East HospitalFrontier Science Center for Stem Cell ResearchShanghai Key Laboratory of Signaling and Disease ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Shanghai Institute of Stem Cell Research and Clinical TranslationShanghaiChina
| |
Collapse
|
31
|
Scheiber AL, Wilkinson KJ, Suzuki A, Enomoto-Iwamoto M, Kaito T, Cheah KS, Iwamoto M, Leikin S, Otsuru S. 4PBA reduces growth deficiency in osteogenesis imperfecta by enhancing transition of hypertrophic chondrocytes to osteoblasts. JCI Insight 2022; 7:149636. [PMID: 34990412 PMCID: PMC8855815 DOI: 10.1172/jci.insight.149636] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022] Open
Abstract
Short stature is a major skeletal phenotype in osteogenesis imperfecta (OI), a genetic disorder mainly caused by mutations in genes encoding type I collagen. However, the underlying mechanism is poorly understood, and no effective treatment is available. In OI mice that carry a G610C mutation in COL1A2, we previously found that mature hypertrophic chondrocytes (HCs) are exposed to cell stress due to accumulation of misfolded mutant type I procollagen in the endoplasmic reticulum (ER). By fate mapping analysis of HCs in G610C OI mice, we found that HCs stagnate in the growth plate, inhibiting translocation of HC descendants to the trabecular area and their differentiation to osteoblasts. Treatment with 4-phenylbutyric acid (4PBA), a chemical chaperone, restored HC ER structure and rescued this inhibition, resulting in enhanced longitudinal bone growth in G610C OI mice. Interestingly, the effects of 4PBA on ER dilation were limited in osteoblasts, and the bone fragility was not ameliorated. These results highlight the importance of targeting HCs to treat growth deficiency in OI. Our findings demonstrate that HC dysfunction induced by ER disruption plays a critical role in the pathogenesis of OI growth deficiency, which lays the foundation for developing new therapies for OI.
Collapse
Affiliation(s)
- Amanda L Scheiber
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| | - Kevin J Wilkinson
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| | - Akiko Suzuki
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kathryn Se Cheah
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| | - Sergey Leikin
- Section on Physical Biochemistry, Eunice Kennedy Shriver National Institute of Child Health & Human Developme, Bethesda, United States of America
| | - Satoru Otsuru
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, United States of America
| |
Collapse
|
32
|
Schilling K, Brown E, Zhang X. NAD(P)H autofluorescence lifetime imaging enables single cell analyses of cellular metabolism of osteoblasts in vitro and in vivo via two-photon microscopy. Bone 2022; 154:116257. [PMID: 34781049 PMCID: PMC8671374 DOI: 10.1016/j.bone.2021.116257] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
Two-photon fluorescence lifetime microscopy (2P-FLIM) is a non-invasive optical technique that can obtain cellular metabolism information based on the intrinsic autofluorescence lifetimes of free and enzyme-bound NAD(P)H, which reflect the metabolic state of single cells within the native microenvironment of the living tissue. NAD(P)H 2P-FLIM was initially performed in bone marrow stromal cell (BMSC) cultures established from Col (I) 2.3GFP or OSX-mCherry mouse models, in which osteoblastic lineage cells were labelled with green or red fluorescence protein, respectively. Measurement of the mean NAD(P)H lifetime, τM, demonstrated that osteoblasts in osteogenic media had a progressively increased τM compared to cells in regular media, suggesting that osteoblasts undergoing mineralization had higher NAD+/NAD(P)H ratio and may utilize more oxidative phosphorylation (OxPhos). In vivo NAD(P)H 2P-FLIM was conducted in conjunction with two-photon phosphorescence lifetime microscopy (2P-PLIM) to evaluate cellular metabolism of GFP+ osteoblasts as well as bone tissue oxygen at different locations of the native cranial bone in Col (I) 2.3GFP mice. Our data showed that osteocytes dwelling within lacunae had higher τM than osteoblasts at the bone edge of suture and marrow space. Measurement of pO2 showed poor correlation of pO2 and τM in native bone. However, when NAD(P)H 2P-FLIM was used to examine osteoblast cellular metabolism at the leading edge of the cranial defects during repair in Col (I) 2.3GFP mouse model, a significantly lower τM was recorded, which was associated with lower pO2 at an early stage of healing, indicating an impact of hypoxia on energy metabolism during bone tissue repair. Taken together, our current study demonstrates the feasibility of using non-invasive optical NAD(P)H 2P-FLIM technique to examine cellular energy metabolism at single cell resolution in living animals. Our data further support that both glycolysis and OxPhos are being used in the osteoblasts, with more mature osteoblasts exhibiting higher ratio of NAD+/NAD(P)H, indicating a potential change of energy mode during differentiation. Further experiments utilizing animals with genetic modification of cellular metabolism could enhance our understanding of energy metabolism in various cell types in living bone microenvironment.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Edward Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
33
|
Siadat SM, Silverman AA, Susilo ME, Paten JA, DiMarzio CA, Ruberti JW. Development of Fluorescently Labeled, Functional Type I Collagen Molecules. Macromol Biosci 2021; 22:e2100144. [PMID: 34856056 DOI: 10.1002/mabi.202100144] [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: 04/12/2021] [Revised: 10/22/2021] [Indexed: 11/11/2022]
Abstract
While de novo collagen fibril formation is well-studied, there are few investigations into the growth and remodeling of extant fibrils, where molecular collagen incorporation into and erosion from the fibril surface must delicately balance during fibril growth and remodeling. Observing molecule/fibril interactions is difficult, requiring the tracking of molecular dynamics while, at the same time, minimizing the effect of the observation on fibril structure and assembly. To address the observation-interference problem, exogenous collagen molecules are tagged with small fluorophores and the fibrillogenesis kinetics of labeled collagen molecules as well as the structure and network morphology of assembled fibrils are examined. While excessive labeling significantly disturbs fibrillogenesis kinetics and network morphology of assembled fibrils, adding less than ≈1.2 labels per collagen molecule preserves these characteristics. Applications of the functional, labeled collagen probe are demonstrated in both cellular and acellular systems. The functional, labeled collagen associates strongly with native fibrils and when added to an in vitro model of corneal stromal development at low concentration, the labeled collagen is incorporated into a fine extracellular matrix (ECM) network associated with the cells within 24 h.
Collapse
Affiliation(s)
| | | | - Monica E Susilo
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| | - Jeffrey A Paten
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02134, USA
| | - Charles A DiMarzio
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Jeffrey W Ruberti
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
34
|
Hixon KR, McKenzie JA, Sykes DAW, Yoneda S, Hensley A, Buettmann EG, Zheng H, Skouteris D, McAlinden A, Miller AN, Silva MJ. Ablation of Proliferating Osteoblast Lineage Cells After Fracture Leads to Atrophic Nonunion in a Mouse Model. J Bone Miner Res 2021; 36:2243-2257. [PMID: 34405443 PMCID: PMC8719642 DOI: 10.1002/jbmr.4424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 07/15/2021] [Accepted: 08/10/2021] [Indexed: 01/19/2023]
Abstract
Nonunion is defined as the permanent failure of a fractured bone to heal, often necessitating surgical intervention. Atrophic nonunions are a subtype that are particularly difficult to treat. Animal models of atrophic nonunion are available; however, these require surgical or radiation-induced trauma to disrupt periosteal healing. These methods are invasive and not representative of many clinical nonunions where osseous regeneration has been arrested by a "failure of biology". We hypothesized that arresting osteoblast cell proliferation after fracture would lead to atrophic nonunion in mice. Using mice that express a thymidine kinase (tk) "suicide gene" driven by the 3.6Col1a1 promoter (Col1-tk), proliferating osteoblast lineage cells can be ablated upon exposure to the nucleoside analog ganciclovir (GCV). Wild-type (WT; control) and Col1-tk littermates were subjected to a full femur fracture and intramedullary fixation at 12 weeks age. We confirmed abundant tk+ cells in fracture callus of Col-tk mice dosed with water or GCV, specifically many osteoblasts, osteocytes, and chondrocytes at the cartilage-bone interface. Histologically, we observed altered callus composition in Col1-tk mice at 2 and 3 weeks postfracture, with significantly less bone and more fibrous tissue. Col1-tk mice, monitored for 12 weeks with in vivo radiographs and micro-computed tomography (μCT) scans, had delayed bone bridging and reduced callus size. After euthanasia, ex vivo μCT and histology showed failed union with residual bone fragments and fibrous tissue in Col1-tk mice. Biomechanical testing showed a failure to recover torsional strength in Col1-tk mice, in contrast to WT. Our data indicates that suppression of proliferating osteoblast-lineage cells for at least 2 weeks after fracture blunts the formation and remodeling of a mineralized callus leading to a functional nonunion. We propose this as a new murine model of atrophic nonunion. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Katherine R Hixon
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - David A W Sykes
- Department of Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Susumu Yoneda
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Austin Hensley
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Dimitrios Skouteris
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Cell Biology & Physiology, Washington University in St. Louis, St. Louis, MO, USA.,St. Louis Shriners Hospital Research Center, Shriners Hospital for Children, St. Louis, MO, USA
| | - Anna N Miller
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO, USA.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
35
|
Kang IH, Baliga UK, Wu Y, Mehrotra S, Yao H, LaRue AC, Mehrotra M. Hematopoietic stem cell-derived functional osteoblasts exhibit therapeutic efficacy in a murine model of osteogenesis imperfecta. Stem Cells 2021; 39:1457-1477. [PMID: 34224636 DOI: 10.1002/stem.3432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/03/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022]
Abstract
Currently, there is no cure for osteogenesis imperfecta (OI)-a debilitating pediatric skeletal dysplasia. Herein we show that hematopoietic stem cell (HSC) therapy holds promise in treating OI. Using single-cell HSC transplantation in lethally irradiated oim/oim mice, we demonstrate significant improvements in bone morphometric, mechanics, and turnover parameters. Importantly, we highlight that HSCs cause these improvements due to their unique property of differentiating into osteoblasts/osteocytes, depositing normal collagen-an attribute thus far assigned only to mesenchymal stem/stromal cells. To confirm HSC plasticity, lineage tracing was done by transplanting oim/oim with HSCs from two specific transgenic mice-VavR, in which all hematopoietic cells are GFP+ and pOBCol2.3GFP, where GFP is expressed only in osteoblasts/osteocytes. In both models, transplanted oim/oim mice demonstrated GFP+ HSC-derived osteoblasts/osteocytes in bones. These studies unequivocally establish that HSCs differentiate into osteoblasts/osteocytes, and HSC transplantation can provide a new translational approach for OI.
Collapse
Affiliation(s)
- In-Hong Kang
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Uday K Baliga
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Yongren Wu
- Department of Orthopedics, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
- Clemson-MUSC Joint Bioengineering Program, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Hai Yao
- Department of Orthopedics, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
- Clemson-MUSC Joint Bioengineering Program, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Amanda C LaRue
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Meenal Mehrotra
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
- Center for Oral Health Research, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
36
|
Garcia J, Smith SS, Karki S, Drissi H, Hrdlicka HH, Youngstrom DW, Delany AM. miR-433-3p suppresses bone formation and mRNAs critical for osteoblast function in mice. J Bone Miner Res 2021; 36:1808-1822. [PMID: 34004029 DOI: 10.1002/jbmr.4339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) are key posttranscriptional regulators of osteoblastic commitment and differentiation. miR-433-3p was previously shown to target Runt-related transcription factor 2 (Runx2) and to be repressed by bone morphogenetic protein (BMP) signaling. Here, we show that miR-433-3p is progressively decreased during osteoblastic differentiation of primary mouse bone marrow stromal cells in vitro, and we confirm its negative regulation of this process. Although repressors of osteoblastic differentiation often promote adipogenesis, inhibition of miR-433-3p did not affect adipocyte differentiation in vitro. Multiple pathways regulate osteogenesis. Using luciferase-3' untranslated region (UTR) reporter assays, five novel miR-433-3p targets involved in parathyroid hormone (PTH), mitogen-activated protein kinase (MAPK), Wnt, and glucocorticoid signaling pathways were validated. We show that Creb1 is a miR-433-3p target, and this transcription factor mediates key signaling downstream of PTH receptor activation. We also show that miR-433-3p targets hydroxysteroid 11-β dehydrogenase 1 (Hsd11b1), the enzyme that locally converts inactive glucocorticoids to their active form. miR-433-3p dampens glucocorticoid signaling, and targeting of Hsd11b1 could contribute to this phenomenon. Moreover, miR-433-3p targets R-spondin 3 (Rspo3), a leucine-rich repeat-containing G-protein coupled receptor (LGR) ligand that enhances Wnt signaling. Notably, Wnt canonical signaling is also blunted by miR-433-3p activity. In vivo, expression of a miR-433-3p inhibitor or tough decoy in the osteoblastic lineage increased trabecular bone volume. Mice expressing the miR-433-3p tough decoy displayed increased bone formation without alterations in osteoblast or osteoclast numbers or surface, indicating that miR-433-3p decreases osteoblast activity. Overall, we showed that miR-433-3p is a negative regulator of bone formation in vivo, targeting key bone-anabolic pathways including those involved in PTH signaling, Wnt, and endogenous glucocorticoids. Local delivery of miR-433-3p inhibitor could present a strategy for the management of bone loss disorders and bone defect repair. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- John Garcia
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Spenser S Smith
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Sangita Karki
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University and Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Henry H Hrdlicka
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| | - Daniel W Youngstrom
- Department of Orthopedic Surgery, UConn Health, Farmington, Connecticut, USA
| | - Anne M Delany
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
37
|
Jeon HH, Yang CY, Shin MK, Wang J, Patel JH, Chung CH, Graves DT. Osteoblast lineage cells and periodontal ligament fibroblasts regulate orthodontic tooth movement that is dependent on Nuclear Factor-kappa B (NF-kB) activation. Angle Orthod 2021; 91:664-671. [PMID: 33852725 PMCID: PMC8376154 DOI: 10.2319/031520-182.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 02/01/2021] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES To investigate the role of NF-κB in osteoblast lineage cells and periodontal ligament (PDL) fibroblasts during orthodontic tooth movement (OTM). MATERIALS AND METHODS Transgenic mice that expressed a dominant negative mutant of the inhibitor of kB kinase (IKK-DN) with lineage specific expression in osteoblastic cells and PDL fibroblasts driven by a response element in the collagen1α1 promoter and matched wild-type (WT) mice were examined. A 10-12 g force was applied by a NiTi coil and maintained for 5 or 12 days. OTM distance, PDL width, and bone volume fraction were measured using micro computed tomography. Osteoclast numbers were counted in tartrate-resistant acid phosphatase-stained sections. Activation of nuclear factor kappa B (NF-kB) was assessed by nuclear localization of p65, and the receptor activator of nuclear factor-κB ligand (RANKL) was measured by immunofluorescence and compared to control specimens with no orthodontic force. RESULTS OTM-induced NF-kB activation (p65 nuclear localization) in WT mice was largely blocked in transgenic (TG) mice. OTM was significantly reduced in the TG mice compared to WT mice along with reduced osteoclastogenesis, narrower PDL width, higher bone volume fraction, and reduced RANKL expression. CONCLUSIONS Osteoblast lineage cells and PDL fibroblasts are key contributors to alveolar bone remodeling in OTM through IKKβ dependent NF-κB activation.
Collapse
|
38
|
Shi Y, Liao X, Long JY, Yao L, Chen J, Yin B, Lou F, He G, Ye L, Qin L, Long F. Gli1 + progenitors mediate bone anabolic function of teriparatide via Hh and Igf signaling. Cell Rep 2021; 36:109542. [PMID: 34407400 PMCID: PMC8432334 DOI: 10.1016/j.celrep.2021.109542] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/27/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023] Open
Abstract
Teriparatide is the most widely prescribed bone anabolic drug in the world, but its cellular targets remain incompletely defined. The Gli1+ metaphyseal mesenchymal progenitors (MMPs) are a main source for osteoblasts in postnatal growing mice, but their potential response to teriparatide is unknown. Here, by lineage tracing, we show that teriparatide stimulates both proliferation and osteoblast differentiation of MMPs. Single-cell RNA sequencing reveals heterogeneity among MMPs, including an unexpected chondrocyte-like osteoprogenitor (COP). COP expresses the highest level of Hedgehog (Hh) target genes and the insulin-like growth factor 1 receptor (Igf1r) among all cell clusters. COP also expresses Pth1r and further upregulates Igf1r upon teriparatide treatment. Inhibition of Hh signaling or deletion of Igf1r from MMPs diminishes the proliferative and osteogenic effects of teriparatide. The study therefore identifies COP as a teriparatide target wherein Hh and insulin-like growth factor (Igf) signaling are critical for the osteoanabolic response in growing mice.
Collapse
Affiliation(s)
- Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xueyang Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - James Y Long
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
| | - Lutian Yao
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jianquan Chen
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China
| | - Bei Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Feng Lou
- West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Qin
- Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Fanxin Long
- Translational Research Program of Pediatric Orthopedics, Department of Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Orthopedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Tsinman TK, Jiang X, Han L, Koyama E, Mauck RL, Dyment NA. Intrinsic and growth-mediated cell and matrix specialization during murine meniscus tissue assembly. FASEB J 2021; 35:e21779. [PMID: 34314047 DOI: 10.1096/fj.202100499r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/10/2021] [Accepted: 06/21/2021] [Indexed: 11/11/2022]
Abstract
The incredible mechanical strength and durability of mature fibrous tissues and their extremely limited turnover and regenerative capacity underscores the importance of proper matrix assembly during early postnatal growth. In tissues with composite extracellular matrix (ECM) structures, such as the adult knee meniscus, fibrous (Collagen-I rich), and cartilaginous (Collagen-II, proteoglycan-rich) matrix components are regionally segregated to the outer and inner portions of the tissue, respectively. While this spatial variation in composition is appreciated to be functionally important for resisting complex mechanical loads associated with gait, the establishment of these specialized zones is poorly understood. To address this issue, the following study tracked the growth of the murine meniscus from its embryonic formation through its first month of growth, encompassing the critical time-window during which animals begin to ambulate and weight bear. Using histological analysis, region specific high-throughput qPCR, and Col-1, and Col-2 fluorescent reporter mice, we found that matrix and cellular features defining specific tissue zones were already present at birth, before continuous weight-bearing had occurred. These differences in meniscus zones were further refined with postnatal growth and maturation, resulting in specialization of mature tissue regions. Taken together, this work establishes a detailed timeline of the concurrent spatiotemporal changes that occur at both the cellular and matrix level throughout meniscus maturation. The findings of this study provide a framework for investigating the reciprocal feedback between cells and their evolving microenvironments during assembly of a mechanically robust fibrocartilage tissue, thus providing insight into mechanisms of tissue degeneration and effective regenerative strategies.
Collapse
Affiliation(s)
- Tonia K Tsinman
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.,Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Xi Jiang
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Eiki Koyama
- Division of Orthopaedic Surgery, Department of Surgery, Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.,Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Cong Q, Liu Y, Zhou T, Zhou Y, Xu R, Cheng C, Chung HS, Yan M, Zhou H, Liao Z, Gao B, Bocobo GA, Covington TA, Song HJ, Su P, Yu PB, Yang Y. A self-amplifying loop of YAP and SHH drives formation and expansion of heterotopic ossification. Sci Transl Med 2021; 13:13/599/eabb2233. [PMID: 34162750 PMCID: PMC8638088 DOI: 10.1126/scitranslmed.abb2233] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 12/03/2020] [Accepted: 05/30/2021] [Indexed: 12/15/2022]
Abstract
Heterotopic ossification (HO) occurs as a common complication after injury or in genetic disorders. The mechanisms underlying HO remain incompletely understood, and there are no approved prophylactic or secondary treatments available. Here, we identify a self-amplifying, self-propagating loop of Yes-associated protein (YAP)-Sonic hedgehog (SHH) as a core molecular mechanism underlying diverse forms of HO. In mouse models of progressive osseous heteroplasia (POH), a disease caused by null mutations in GNAS, we found that Gnas-/- mesenchymal cells secreted SHH, which induced osteoblast differentiation of the surrounding wild-type cells. We further showed that loss of Gnas led to activation of YAP transcription activity, which directly drove Shh expression. Secreted SHH further induced YAP activation, Shh expression, and osteoblast differentiation in surrounding wild-type cells. This self-propagating positive feedback loop was both necessary and sufficient for HO expansion and could act independently of Gnas in fibrodysplasia ossificans progressiva (FOP), another genetic HO, and nonhereditary HO mouse models. Genetic or pharmacological inhibition of YAP or SHH abolished HO in POH and FOP and acquired HO mouse models without affecting normal bone homeostasis, providing a previously unrecognized therapeutic rationale to prevent, reduce, and shrink HO.
Collapse
Affiliation(s)
- Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Taifeng Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Yaxing Zhou
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Ruoshi Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Caiqi Cheng
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Hye Soo Chung
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Meijun Yan
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Hang Zhou
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Zhiheng Liao
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Bo Gao
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA
| | - Geoffrey A Bocobo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Taylor A Covington
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Hyeon Ju Song
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Peiqiang Su
- Department of Orthopaedic Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, 188 Longwood Ave., Boston, MA 02115, USA.
| |
Collapse
|
41
|
Theret M, Low M, Rempel L, Li FF, Tung LW, Contreras O, Chang CK, Wu A, Soliman H, Rossi FMV. In vitro assessment of anti-fibrotic drug activity does not predict in vivo efficacy in murine models of Duchenne muscular dystrophy. Life Sci 2021; 279:119482. [PMID: 33891939 DOI: 10.1016/j.lfs.2021.119482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 02/09/2023]
Abstract
AIM Fibrosis is the most common complication from chronic diseases, and yet no therapy capable of mitigating its effects is available. Our goal is to unveil specific signaling regulating the fibrogenic process and to identify potential small molecule candidates that block fibrogenic differentiation of fibro/adipogenic progenitors. METHOD We performed a large-scale drug screen using muscle-resident fibro/adipogenic progenitors from a mouse model expressing EGFP under the Collagen1a1 promotor. We first confirmed that the EGFP was expressed in response to TGFβ1 stimulation in vitro. Then we treated cells with TGFβ1 alone or with drugs from two libraries of known compounds. The drugs ability to block the fibrogenic differentiation was quantified by imaging and flow cytometry. From a two-rounds screening, positive hits were tested in vivo in the mice model for the Duchenne Muscular Dystrophy (mdx mice). The histopathology of the muscles was assessed with picrosirius red (fibrosis) and laminin staining (myofiber size). KEY FINDINGS From the in vitro drug screening, we identified 21 drugs and tested 3 in vivo on the mdx mice. None of the three drugs significantly improved muscle histopathology. SIGNIFICANCE The in vitro drug screen identified various efficient compounds, none of them strongly inhibited fibrosis in skeletal muscle of mdx mice. To explain these observations, we hypothesize that in Duchenne Muscular Dystrophy, in which fibrosis is a secondary event due to chronic degeneration and inflammation, the drugs tested could have adverse effect on regeneration or inflammation, balancing off any positive effects and leading to the absence of significant results.
Collapse
Affiliation(s)
- Marine Theret
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Marcela Low
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lucas Rempel
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Fang Fang Li
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Chih-Kai Chang
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Andrew Wu
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Hesham Soliman
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, Minia University, Minia, Egypt
| | - Fabio M V Rossi
- School of Biomedical Engineering and the Biomedical Research Centre, Department of Medical Genetics, 2222 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
42
|
Cosgrove BD, Loebel C, Driscoll TP, Tsinman TK, Dai EN, Heo SJ, Dyment NA, Burdick JA, Mauck RL. Nuclear envelope wrinkling predicts mesenchymal progenitor cell mechano-response in 2D and 3D microenvironments. Biomaterials 2021; 270:120662. [PMID: 33540172 PMCID: PMC7936657 DOI: 10.1016/j.biomaterials.2021.120662] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/24/2020] [Accepted: 01/03/2021] [Indexed: 12/21/2022]
Abstract
Exogenous mechanical cues are transmitted from the extracellular matrix to the nuclear envelope (NE), where mechanical stress on the NE mediates shuttling of transcription factors and other signaling cascades that dictate downstream cellular behavior and fate decisions. To systematically study how nuclear morphology can change across various physiologic microenvironmental contexts, we cultured mesenchymal progenitor cells (MSCs) in engineered 2D and 3D hyaluronic acid hydrogel systems. Across multiple contexts we observed highly 'wrinkled' nuclear envelopes, and subsequently developed a quantitative single-cell imaging metric to better evaluate how wrinkles in the nuclear envelope relate to progenitor cell mechanotransduction. We determined that in soft 2D environments the NE is predominately wrinkled, and that increases in cellular mechanosensing (indicated by cellular spreading, adhesion complex growth, and nuclear localization of YAP/TAZ) occurred only in absence of nuclear envelope wrinkling. Conversely, in 3D hydrogel and tissue contexts, we found NE wrinkling occurred along with increased YAP/TAZ nuclear localization. We further determined that these NE wrinkles in 3D were largely generated by actin impingement, and compared to other nuclear morphometrics, the degree of nuclear wrinkling showed the greatest correlation with nuclear YAP/TAZ localization. These findings suggest that the degree of nuclear envelope wrinkling can predict mechanotransduction state in mesenchymal progenitor cells and highlights the differential mechanisms of NE stress generation operative in 2D and 3D microenvironmental contexts.
Collapse
Affiliation(s)
- Brian D Cosgrove
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Tristan P Driscoll
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Tonia K Tsinman
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Eric N Dai
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania Philadelphia, PA, 19104, USA; Translational Musculoskeletal Research Center, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, 19104, USA.
| |
Collapse
|
43
|
Matthews BG, Novak S, Sbrana FV, Funnell JL, Cao Y, Buckels EJ, Grcevic D, Kalajzic I. Heterogeneity of murine periosteum progenitors involved in fracture healing. eLife 2021; 10:e58534. [PMID: 33560227 PMCID: PMC7906599 DOI: 10.7554/elife.58534] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
The periosteum is the major source of cells involved in fracture healing. We sought to characterize progenitor cells and their contribution to bone fracture healing. The periosteum is highly enriched with progenitor cells, including Sca1+ cells, fibroblast colony-forming units, and label-retaining cells compared to the endosteum and bone marrow. Using lineage tracing, we demonstrate that alpha smooth muscle actin (αSMA) identifies long-term, slow-cycling, self-renewing osteochondroprogenitors in the adult periosteum that are functionally important for bone formation during fracture healing. In addition, Col2.3CreER-labeled osteoblast cells contribute around 10% of osteoblasts but no chondrocytes in fracture calluses. Most periosteal osteochondroprogenitors following fracture can be targeted by αSMACreER. Previously identified skeletal stem cell populations were common in periosteum but contained high proportions of mature osteoblasts. We have demonstrated that the periosteum is highly enriched with skeletal progenitor cells, and there is heterogeneity in the populations of cells that contribute to mature lineages during periosteal fracture healing.
Collapse
Affiliation(s)
- Brya G Matthews
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Sanja Novak
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Francesca V Sbrana
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Jessica L Funnell
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| | - Ye Cao
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Emma J Buckels
- Department of Molecular Medicine and Pathology, University of AucklandAucklandNew Zealand
| | - Danka Grcevic
- Department of Physiology and Immunology, University of ZagrebZagrebCroatia
- Croatian Intitute for Brain Research, University of ZagrebZagrebCroatia
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn HealthFarmingtonUnited States
| |
Collapse
|
44
|
Doherty L, Wan M, Kalajzic I, Sanjay A. Diabetes impairs periosteal progenitor regenerative potential. Bone 2021; 143:115764. [PMID: 33221502 PMCID: PMC7770068 DOI: 10.1016/j.bone.2020.115764] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Diabetics are at increased risk for fracture, and experience severely impaired skeletal healing characterized by delayed union or nonunion of the bone. The periosteum harbors osteochondral progenitors that can differentiate into chondrocytes and osteoblasts, and this connective tissue layer is required for efficient fracture healing. While bone marrow-derived stromal cells have been studied extensively in the context of diabetic skeletal repair and osteogenesis, the effect of diabetes on the periosteum and its ability to contribute to bone regeneration has not yet been explicitly evaluated. Within this study, we utilized an established murine model of type I diabetes to evaluate periosteal cell differentiation capacity, proliferation, and availability under the effect of a diabetic environment. Periosteal cells from diabetic mice were deficient in osteogenic differentiation ability in vitro, and diabetic mice had reduced periosteal populations of mesenchymal progenitors with a corresponding reduction in proliferation capacity following injury. Additionally, fracture callus mineralization and mature osteoblast activity during periosteum-mediated healing was impaired in diabetic mice compared to controls. We propose that the effect of diabetes on periosteal progenitors and their ability to aid in skeletal repair directly impairs fracture healing.
Collapse
Affiliation(s)
- Laura Doherty
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Matthew Wan
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn School of Dental Medicine, Farmington, CT, USA
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| |
Collapse
|
45
|
Yu W, Zhong L, Yao L, Wei Y, Gui T, Li Z, Kim H, Holdreith N, Jiang X, Tong W, Dyment N, Liu XS, Yang S, Choi Y, Ahn J, Qin L. Bone marrow adipogenic lineage precursors promote osteoclastogenesis in bone remodeling and pathologic bone loss. J Clin Invest 2021; 131:140214. [PMID: 33206630 DOI: 10.1172/jci140214] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is maintained by coupled activities of bone-forming osteoblasts/osteocytes and bone-resorbing osteoclasts. Alterations in this relationship can lead to pathologic bone loss such as osteoporosis. It is well known that osteogenic cells support osteoclastogenesis via production of RANKL. Interestingly, our recently identified bone marrow mesenchymal cell population-marrow adipogenic lineage precursors (MALPs) that form a multidimensional cell network in bone-was computationally demonstrated to be the most interactive with monocyte-macrophage lineage cells through high and specific expression of several osteoclast regulatory factors, including RANKL. Using an adipocyte-specific Adipoq-Cre to label MALPs, we demonstrated that mice with RANKL deficiency in MALPs have a drastic increase in trabecular bone mass in long bones and vertebrae starting from 1 month of age, while their cortical bone appears normal. This phenotype was accompanied by diminished osteoclast number and attenuated bone formation at the trabecular bone surface. Reduced RANKL signaling in calvarial MALPs abolished osteolytic lesions after LPS injections. Furthermore, in ovariectomized mice, elevated bone resorption was partially attenuated by RANKL deficiency in MALPs. In summary, our studies identified MALPs as a critical player in controlling bone remodeling during normal bone metabolism and pathological bone loss in a RANKL-dependent fashion.
Collapse
Affiliation(s)
- Wei Yu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Gui
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Bone and Joint Surgery, Institute of Orthopedic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Ziqing Li
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hyunsoo Kim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicholas Holdreith
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xi Jiang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nathaniel Dyment
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - X Sherry Liu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
46
|
Martin CS, Cooper MS, Hardy RS. Endogenous Glucocorticoid Metabolism in Bone: Friend or Foe. Front Endocrinol (Lausanne) 2021; 12:733611. [PMID: 34512556 PMCID: PMC8429897 DOI: 10.3389/fendo.2021.733611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023] Open
Abstract
The role of tissue specific metabolism of endogenous glucocorticoids (GCs) in the pathogenesis of human disease has been a field of intense interest over the last 20 years, fuelling clinical trials of metabolism inhibitors in the treatment of an array of metabolic diseases. Localised pre-receptor metabolism of endogenous and therapeutic GCs by the 11β-hydroxysteroid dehydrogenase (11β-HSD) enzymes (which interconvert endogenous GCs between their inactive and active forms) are increasingly recognised as being critical in mediating both their positive and negative actions on bone homeostasis. In this review we explore the roles of endogenous and therapeutic GC metabolism by the 11β-HSD enzymes in the context of bone metabolism and bone cell function, and consider future strategies aimed at modulating this system in order to manage and treat various bone diseases.
Collapse
Affiliation(s)
- Claire S. Martin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Mark S. Cooper
- Australian and New Zealand Army Corps (ANZAC) Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Rowan S. Hardy
- Arthritis Research United Kingdom (UK) Career Development Fellow, University of Birmingham, Birmingham, United Kingdom
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Rowan S. Hardy,
| |
Collapse
|
47
|
Ma C, Jing Y, Li H, Wang K, Wang Z, Xu C, Sun X, Kaji D, Han X, Huang A, Feng J. Scx Lin cells directly form a subset of chondrocytes in temporomandibular joint that are sharply increased in Dmp1-null mice. Bone 2021; 142:115687. [PMID: 33059101 PMCID: PMC7749445 DOI: 10.1016/j.bone.2020.115687] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
It has been assumed that the secondary cartilage in the temporomandibular joint (TMJ), which is the most complex and mystery joint and expands rapidly after birth, is formed by periochondrium-derived chondrocytes. The TMJ condyle has rich attachment sites of tendon, which is thought to be solely responsible for joint movement with a distinct cell lineage. Here, we used a Scx-Cre ERT2 mouse line (the tracing line for progenitor and mature tendon cells) to track the fate of tendon cells during TMJ postnatal growth. Our data showed a progressive differentiation of Scx lineage cells started at tendon and the fibrous layer, to cells at the prechondroblasts (Sox9 -/Col I +), and then to cells at the chondrocytic layer (Sox9 +/Col I -). Importantly, the Scx + chondrocytes remained as "permanent" chondrocytes to maintain cartilage mass with no further cell trandifferentiation to bone cells. This notion was substantiated in an assessment of these cells in Dmp1 -null mice (a hypophosphatemic rickets model), where there was a significant increase in the number of Scx lineage cells in response to hypophosphatemia. In addition, we showed the origin of disc, which is derived from Scx + cells. Thus, we propose Scx lineage cells play an important role in TMJ postnatal growth by forming the disc and a new subset of Scx + chondrocytes that do not undergo osteogenesis as the Scx - chondrocytes and are sensitive to the level of phosphorous.
Collapse
Affiliation(s)
- Chi Ma
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yan Jing
- Department of Orthodontics, Texas A&M College of Dentistry, Dallas, TX, USA
- Corresponding authors Yan Jing, Assistant professor, Department of Orthodontics, Texas A&M College of Dentistry, 3302 Gaston Ave, Dallas, Tx, USA, , 2143707237, Jian Feng, Professor, Department of Biomedical sciences, Texas A&M College of Dentistry, Texas A&M College of Dentistry, 3302 Gaston Ave, Dallas, Tx, USA, , 2143707235
| | - Hui Li
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Ke Wang
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Zheng Wang
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Chunmei Xu
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| | - Xiaolin Sun
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA; Zhongshan Affiliated Hospital of Dalian University, Dalian, China
| | - Deepak Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Xianglong Han
- Department of Orthodontics & Pediatric Dentistry, West China School of Stomatology, State Key Laboratory of Oral Diseases, Sichuan University, Chengdu, China
| | - Alice Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jian Feng
- Department of Biomedical Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
- Corresponding authors Yan Jing, Assistant professor, Department of Orthodontics, Texas A&M College of Dentistry, 3302 Gaston Ave, Dallas, Tx, USA, , 2143707237, Jian Feng, Professor, Department of Biomedical sciences, Texas A&M College of Dentistry, Texas A&M College of Dentistry, 3302 Gaston Ave, Dallas, Tx, USA, , 2143707235
| |
Collapse
|
48
|
Haltalli MLR, Watcham S, Wilson NK, Eilers K, Lipien A, Ang H, Birch F, Anton SG, Pirillo C, Ruivo N, Vainieri ML, Pospori C, Sinden RE, Luis TC, Langhorne J, Duffy KR, Göttgens B, Blagborough AM, Lo Celso C. Manipulating niche composition limits damage to haematopoietic stem cells during Plasmodium infection. Nat Cell Biol 2020; 22:1399-1410. [PMID: 33230302 PMCID: PMC7611033 DOI: 10.1038/s41556-020-00601-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/06/2020] [Indexed: 12/17/2022]
Abstract
Severe infections are a major stress on haematopoiesis, where the consequences for haematopoietic stem cells (HSCs) have only recently started to emerge. HSC function critically depends on the integrity of complex bone marrow (BM) niches; however, what role the BM microenvironment plays in mediating the effects of infection on HSCs remains an open question. Here, using a murine model of malaria and combining single-cell RNA sequencing, mathematical modelling, transplantation assays and intravital microscopy, we show that haematopoiesis is reprogrammed upon infection, whereby the HSC compartment turns over substantially faster than at steady-state and HSC function is drastically affected. Interferon is found to affect both haematopoietic and mesenchymal BM cells and we specifically identify a dramatic loss of osteoblasts and alterations in endothelial cell function. Osteo-active parathyroid hormone treatment abolishes infection-triggered HSC proliferation and-coupled with reactive oxygen species quenching-enables partial rescuing of HSC function.
Collapse
Affiliation(s)
- Myriam L R Haltalli
- Department of Life Sciences, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, UK
| | - Samuel Watcham
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, UK
| | - Nicola K Wilson
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, UK
| | - Kira Eilers
- Department of Life Sciences, Imperial College London, London, UK
| | - Alexander Lipien
- Department of Life Sciences, Imperial College London, London, UK
| | - Heather Ang
- Department of Life Sciences, Imperial College London, London, UK
| | - Flora Birch
- Department of Life Sciences, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Sara Gonzalez Anton
- Department of Life Sciences, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Chiara Pirillo
- Department of Life Sciences, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Nicola Ruivo
- Department of Life Sciences, Imperial College London, London, UK
| | - Maria L Vainieri
- Department of Life Sciences, Imperial College London, London, UK
- AO Research Institute, Davos Platz, Switzerland
| | - Constandina Pospori
- Department of Life Sciences, Imperial College London, London, UK
- The Francis Crick Institute, London, UK
| | - Robert E Sinden
- Department of Life Sciences, Imperial College London, London, UK
| | - Tiago C Luis
- Department of Life Sciences, Imperial College London, London, UK
| | | | - Ken R Duffy
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Berthold Göttgens
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Haematology, Cambridge Institute for Medical Research, Cambridge, UK
| | | | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
49
|
Abstract
Osteocytes are an ancient cell, appearing in fossilized skeletal remains of early fish and dinosaurs. Despite its relative high abundance, even in the context of nonskeletal cells, the osteocyte is perhaps among the least studied cells in all of vertebrate biology. Osteocytes are cells embedded in bone, able to modify their surrounding extracellular matrix via specialized molecular remodeling mechanisms that are independent of the bone forming osteoblasts and bone-resorbing osteoclasts. Osteocytes communicate with osteoclasts and osteoblasts via distinct signaling molecules that include the RankL/OPG axis and the Sost/Dkk1/Wnt axis, among others. Osteocytes also extend their influence beyond the local bone environment by functioning as an endocrine cell that controls phosphate reabsorption in the kidney, insulin secretion in the pancreas, and skeletal muscle function. These cells are also finely tuned sensors of mechanical stimulation to coordinate with effector cells to adjust bone mass, size, and shape to conform to mechanical demands.
Collapse
Affiliation(s)
- Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| |
Collapse
|
50
|
Hashimoto K, Kaito T, Kikuta J, Ishii M. Intravital imaging of orthotopic and ectopic bone. Inflamm Regen 2020; 40:26. [PMID: 33292699 PMCID: PMC7604953 DOI: 10.1186/s41232-020-00135-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/31/2020] [Indexed: 12/02/2022] Open
Abstract
Bone homeostasis is dynamically regulated by a balance between bone resorption by osteoclasts and bone formation by osteoblasts. Visualizing and evaluating the dynamics of bone cells in vivo remain difficult using conventional technologies, including histomorphometry and imaging analysis. Over the past two decades, multiphoton microscopy, which can penetrate thick specimens, has been utilized in the field of biological imaging. Using this innovative technique, the in vivo dynamic motion of bone metabolism-related cells and their interactions has been revealed. In this review, we summarize previous approaches used for bone imaging and provide an overview of current bone tissue imaging methods using multiphoton excitation microscopy.
Collapse
Affiliation(s)
- Kunihiko Hashimoto
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan.,Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine & Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|