1
|
Kawano Y, Kawano H, Busch S, Li AJ, Zhang J, Salama NA, Quarato ER, Georger M, Vdovichenko N, Azadniv M, Byun DK, LaMere EA, LaMere MW, Liesveld JL, Becker MW, Calvi LM. Monocytes/macrophages contamination disrupts functional and transcriptional characteristics of murine bone marrow- and bone-derived stromal cells. JBMR Plus 2025; 9:ziaf047. [PMID: 40329992 PMCID: PMC12054994 DOI: 10.1093/jbmrpl/ziaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 05/08/2025] Open
Abstract
Stromal cells are critical regulators of hematopoietic stem/progenitor cells and skeletal homeostasis. Although precise systems for functional analysis are critical to investigate mechanistically bone and bone marrow (BM)-derived stromal cells, the establishment of reproducible, highly enriched ex vivo methods for stromal cell isolation, culture and evaluation have been challenging, leading to inconsistent data on stromal cell function. In this work, we carefully tested ex vivo culture of murine stromal cells from BM and bone and discovered abundant and persistent contamination of monocytes and macrophages. We succeeded in establishing highly enriched ex vivo culture system for stromal cells by eliminating persistent monocytes and macrophages using selection against the immunological markers F4/80, Ly6C, and CD45. Transcriptional and functional assays of enriched stromal cell culture revealed differential characteristics of stromal cells from different origins, a dormant signature for bone-derived cells and a highly proliferative progenitor-like signature for BM-derived cells. Monocyte and macrophage contamination reduced signatures of immature stromal cells such as expression levels of SOX9 and CD140a as well as the cells' ability to support hematopoietic stem and progenitor cells based on our growth factor-free co-culture system of hematopoietic cells and stromal cells followed by in vivo functional assays. The inhibitory effects of macrophages on stromal cells may be explained by their potent production of inflammatory cytokines such as CXCL2, CCL3, and complement factor (C1q) confirmed by protein immunoassay of culture supernatant, as well as the differential contribution of pre-osteoblasts to the stromal cell population. This study highlights the functional diversity of stromal cells depending on the microenvironment of origin while addressing a critical limitation of murine ex vivo systems. Our robust culture system enables the study of isolated stromal cells function as well as the impact of stromal cells-macrophage crosstalk.
Collapse
Affiliation(s)
- Yuko Kawano
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Hiroki Kawano
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Stephanie Busch
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Allison J Li
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Jane Zhang
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Noah A Salama
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Emily R Quarato
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Mary Georger
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Nataliia Vdovichenko
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Mitra Azadniv
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Daniel K Byun
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Elizabeth A LaMere
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Mark W LaMere
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Jane L Liesveld
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Michael W Becker
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Hematology/Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| | - Laura M Calvi
- James P. Wilmot Cancer Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, United States
| |
Collapse
|
2
|
Xie Y, Su J, Yang M, Liu Z, Chen T, Qian J, Yu B, Zhang X. Prenatal dexamethasone exposure reduces osteoprogenitor proliferation in mice via histone modifications at the Mkp-1 gene locus. Commun Biol 2024; 7:1589. [PMID: 39609620 PMCID: PMC11604782 DOI: 10.1038/s42003-024-07288-x] [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: 04/12/2023] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Prenatal dexamethasone exposure (PDE) has long-term consequences in bone development, though the underlying mechanisms remain unclear. Our results show that PDE offspring exhibit reduced bone mass, fewer osteoblasts and diminished osteoprogenitors proliferation. Further analyses show that PDE increases MKP-1 expression, while decreasing H3 lysine 9 dimethylation (H3K9me2) and H3 lysine 27 trimethylation (H3K27me3) at the Mkp-1 gene locus. Mechanistically, dexamethasone suppresses osteoprogenitors proliferation by upregulating MKP-1 expression, notably through the inhibition of H3K9me2 and H3K27me3 modifications, which promote demethylation and transcriptional activation of the Mkp-1 gene. Importantly, restoring histone methylation balance with PFI-90 or GSK-J4 treatment blocks the inhibitory effects of PDE on MAPK signaling in osteoprogenitors, and mitigates the detrimental impact of PDE on osteoprogenitor proliferation and bone development in the offspring. This study provides new insights into the epigenetic mechanism by which PDE disrupts long-term programming of fetal osteoprogenitor proliferation, ultimately impairing long bone growth in offspring.
Collapse
Affiliation(s)
- Yongheng Xie
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Jianwen Su
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mankai Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Te Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jikun Qian
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xianrong Zhang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, No.1838 North of Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China.
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
3
|
Abbasizadeh N, Burns CS, Verrinder R, Ghazali F, Seyedhassantehrani N, Spencer JA. Age and dose dependent changes to the bone and bone marrow microenvironment after cytotoxic conditioning with busulfan. Front Cell Dev Biol 2024; 12:1441381. [PMID: 39139448 PMCID: PMC11319712 DOI: 10.3389/fcell.2024.1441381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Preparative regimens before Hematopoietic Cell Transplantation (HCT) damage the bone marrow (BM) microenvironment, potentially leading to secondary morbidity and even mortality. The precise effects of cytotoxic preconditioning on bone and BM remodeling, regeneration, and subsequent hematopoietic recovery over time remain unclear. Moreover, the influence of recipient age and cytotoxic dose have not been fully described. In this study, we longitudinally investigated bone and BM remodeling after busulfan treatment with low intensity (LI) and high intensity (HI) regimens as a function of animal age. As expected, higher donor chimerism was observed in young mice in both LI and HI regimens compared to adult mice. Noticeably in adult mice, significant engraftment was only observed in the HI group. The integrity of the blood-bone marrow barrier in calvarial BM blood vessels was lost after busulfan treatment in the young mice and remained altered even 6 weeks after HCT. In adult mice, the severity of vascular leakage appeared to be dose-dependent, being more pronounced in HI compared to LI recipients. Interestingly, no noticeable change in blood flow velocity was observed following busulfan treatment. Ex vivo imaging of the long bones revealed a reduction in the frequency and an increase in the diameter and density of the blood vessels shortly after treatment, a phenomenon that largely recovered in young mice but persisted in older mice after 6 weeks. Furthermore, analysis of bone remodeling indicated a significant alteration in bone turnover at 6 weeks compared to earlier timepoints in both young and adult mice. Overall, our results reveal new aspects of bone and BM remodeling, as well as hematopoietic recovery, which is dependent on the cytotoxic dose and recipient age.
Collapse
Affiliation(s)
- Nastaran Abbasizadeh
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Christian S. Burns
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Ruth Verrinder
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Farhad Ghazali
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
| | - Negar Seyedhassantehrani
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
| | - Joel A. Spencer
- Department of Bioengineering, University of California, Merced, Merced, CA, United States
- Center for Cellular and Biomolecular Machines, University of California, Merced, Merced, CA, United States
- Health Sciences Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
4
|
Zhao YJ, Yin G, Liu B, Deng XQ, Cao HY, Liu Y. Variability of BMP-2 content in DBM products derived from different long bone. Cell Tissue Bank 2024; 25:697-703. [PMID: 38489016 DOI: 10.1007/s10561-024-10132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/30/2024] [Indexed: 03/17/2024]
Abstract
Demineralized bone matrix (DBM) has been regarded as an ideal bone substitute as a native carrier of bone morphogenetic proteins (BMPs) and other growth factors. However, the osteoinductive properties diverse in different DBM products. We speculate that the harvest origin further contributing to variability of BMPs contents in DBM products besides the process technology. In the study, the cortical bone of femur, tibia, humerus, and ulna from a signal donor were prepared and followed demineralizd into DBM products. Proteins in bone martix were extracted using guanidine-HCl and collagenase, respectively, and BMP-2 content was detected by sandwich enzyme-linked immunosorbent assay (ELISA). Variability of BMP-2 content was found in 4 different DBM products. By guanidine-HCl extraction, the average concentration in DBMs harvested from ulna, humerus, tibia, and femur were 0.613 ± 0.053, 0.848 ± 0.051, 3.293 ± 0.268, and 21.763 ± 0.344, respectively (p < 0.05), while using collagenase, the levels were 0.089 ± 0.004, 0.097 ± 0.004, 0.330 ± 0.012, and 1.562 ± 0.008, respectively (p < 0.05). In general, the content of BMP-2 in long bones of Lower limb was higher than that in long bones of upper limb, and GuHCl had remarkably superior extracted efficiency for BMP-2 compared to collagenase. The results suggest that the origin of cortical bones harvested to fabricate DBM products contribute to the variability of native BMP-2 content, while the protein extracted method only changes the measured values of BMP-2.
Collapse
Affiliation(s)
- Yong-Jie Zhao
- Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Gang Yin
- Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Bin Liu
- Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Xiao-Qiang Deng
- Xing'an League People's Hospital, Ulanhot, Inner Mongolia, China
| | - Hai-Yan Cao
- Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Ying Liu
- Binzhou Medical University Hospital, Binzhou, Shandong Province, China.
| |
Collapse
|
5
|
Li X, Liang T, Dai B, Chang L, Zhang Y, Hu S, Guo J, Xu S, Zheng L, Yao H, Lian H, Nie Y, Li Y, He X, Yao Z, Tong W, Wang X, Chow DHK, Xu J, Qin L. Excess glucocorticoids inhibit murine bone turnover via modulating the immunometabolism of the skeletal microenvironment. J Clin Invest 2024; 134:e166795. [PMID: 38512413 DOI: 10.1172/jci166795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Elevated bone resorption and diminished bone formation have been recognized as the primary features of glucocorticoid-associated skeletal disorders. However, the direct effects of excess glucocorticoids on bone turnover remain unclear. Here, we explored the outcomes of exogenous glucocorticoid treatment on bone loss and delayed fracture healing in mice and found that reduced bone turnover was a dominant feature, resulting in a net loss of bone mass. The primary effect of glucocorticoids on osteogenic differentiation was not inhibitory; instead, they cooperated with macrophages to facilitate osteogenesis. Impaired local nutrient status - notably, obstructed fatty acid transportation - was a key factor contributing to glucocorticoid-induced impairment of bone turnover in vivo. Furthermore, fatty acid oxidation in macrophages fueled the ability of glucocorticoid-liganded receptors to enter the nucleus and then promoted the expression of BMP2, a key cytokine that facilitates osteogenesis. Metabolic reprogramming by localized fatty acid delivery partly rescued glucocorticoid-induced pathology by restoring a healthier immune-metabolic milieu. These data provide insights into the multifactorial metabolic mechanisms by which glucocorticoids generate skeletal disorders, thus suggesting possible therapeutic avenues.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Tongzhou Liang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Yuan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shiwen Hu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Hong Lian
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, and
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Zhi Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Xinluan Wang
- Centre for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, Faculty of Medicine
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, and
| |
Collapse
|
6
|
Matsushita Y, Liu J, Chu AKY, Ono W, Welch JD, Ono N. Endosteal stem cells at the bone-blood interface: A double-edged sword for rapid bone formation: Bone marrow endosteal stem cells provide a robust source of bone-making osteoblasts both in normal and abnormal bone formation. Bioessays 2024; 46:e2300173. [PMID: 38161246 PMCID: PMC11729589 DOI: 10.1002/bies.202300173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Endosteal stem cells are a subclass of bone marrow skeletal stem cell populations that are particularly important for rapid bone formation occurring in growth and regeneration. These stem cells are strategically located near the bone surface in a specialized microenvironment of the endosteal niche. These stem cells are abundant in young stages but eventually depleted and replaced by other stem cell types residing in a non-endosteal perisinusoidal niche. Single-cell molecular profiling and in vivo cell lineage analyses play key roles in discovering endosteal stem cells. Importantly, endosteal stem cells can transform into bone tumor-making cells when deleterious mutations occur in tumor suppressor genes. The emerging hypothesis is that osteoblast-chondrocyte transitional identities confer a special subset of endosteal stromal cells with stem cell-like properties, which may make them susceptible for tumorigenic transformation. Endosteal stem cells are likely to represent an important therapeutic target of bone diseases caused by aberrant bone formation.
Collapse
Affiliation(s)
- Yuki Matsushita
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jialin Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Angel Ka Yan Chu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Joshua D. Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
7
|
Cao Y, Bolam SM, Boss AL, Murray HC, Munro JT, Poulsen RC, Dalbeth N, Brooks AES, Matthews BG. Characterization of adult human skeletal cells in different tissues reveals a CD90 +CD34 + periosteal stem/progenitor population. Bone 2024; 178:116926. [PMID: 37793499 DOI: 10.1016/j.bone.2023.116926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/06/2023]
Abstract
The periosteum plays a crucial role in bone healing and is an important source of skeletal stem and progenitor cells. Recent studies in mice indicate that diverse populations of skeletal progenitors contribute to growth, homeostasis and healing. Information about the in vivo identity and diversity of skeletal stem and progenitor cells in different compartments of the adult human skeleton is limited. In this study, we compared non-hematopoietic populations in matched tissues from the femoral head and neck of 21 human participants using spectral flow cytometry of freshly isolated cells. High-dimensional clustering analysis indicated significant differences in marker distribution between periosteum, articular cartilage, endosteum and bone marrow populations, and identified populations that were highly enriched or unique to specific tissues. Periosteum-enriched markers included CD90 and CD34. Articular cartilage, which has very poor regenerative potential, showed enrichment of multiple markers, including the PDPN+CD73+CD164+CD146- population previously reported to represent human skeletal stem cells. We further characterized periosteal populations by combining CD90 with other strongly expressed markers. CD90+CD34+ cells sorted directly from periosteum showed significant colony-forming unit fibroblasts (CFU-F) enrichment, rapid expansion, and consistent multi-lineage differentiation of clonal populations in vitro. In situ, CD90+CD34+ cells include a perivascular population in the outer layer of the periosteum and non-perivascular cells closer to the bone surface. CD90+ cells are also highly enriched for CFU-F in bone marrow and endosteum, but not articular cartilage. In conclusion, our study indicates considerable diversity in the non-hematopoietic cell populations in different tissue compartments within the adult human skeleton, and suggests that periosteal progenitor cells reside within the CD90+CD34+ population.
Collapse
Affiliation(s)
- Ye Cao
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Scott M Bolam
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Anna L Boss
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Helen C Murray
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Jacob T Munro
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Raewyn C Poulsen
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Brya G Matthews
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
8
|
Akerman AW, Alexander KC, Caranasos TG, Ikonomidis JS. Therapeutic potential of mesenchymal stem cells and their secreted extracellular vesicles in thoracic aortic aneurysm disease. J Thorac Cardiovasc Surg 2024; 167:89-93.e1. [PMID: 37084818 PMCID: PMC10882625 DOI: 10.1016/j.jtcvs.2023.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/23/2023]
Affiliation(s)
- Adam W Akerman
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Kyle C Alexander
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - Thomas G Caranasos
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC
| | - John S Ikonomidis
- Division of Cardiothoracic Surgery, Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, NC.
| |
Collapse
|
9
|
Atria PJ, Castillo AB. Skeletal adaptation to mechanical cues during homeostasis and repair: the niche, cells, and molecular signaling. Front Physiol 2023; 14:1233920. [PMID: 37916223 PMCID: PMC10616261 DOI: 10.3389/fphys.2023.1233920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Bones constantly change and adapt to physical stress throughout a person's life. Mechanical signals are important regulators of bone remodeling and repair by activating skeletal stem and progenitor cells (SSPCs) to proliferate and differentiate into bone-forming osteoblasts using molecular signaling mechanisms not yet fully understood. SSPCs reside in a dynamic specialized microenvironment called the niche, where external signals integrate to influence cell maintenance, behavior and fate determination. The nature of the niche in bone, including its cellular and extracellular makeup and regulatory molecular signals, is not completely understood. The mechanisms by which the niche, with all of its components and complexity, is modulated by mechanical signals during homeostasis and repair are virtually unknown. This review summarizes the current view of the cells and signals involved in mechanical adaptation of bone during homeostasis and repair, with an emphasis on identifying novel targets for the prevention and treatment of age-related bone loss and hard-to-heal fractures.
Collapse
Affiliation(s)
- Pablo J. Atria
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY, United States
| | - Alesha B. Castillo
- Department of Orthopedic Surgery, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, NY, United States
| |
Collapse
|
10
|
Sørensen NN, Andreasen CM, Jensen PR, Hauge EM, Bollerslev J, Delaissé JM, Kassem M, Jafari A, Diaz-delCastillo M, Andersen TL. Disturbed bone marrow adiposity in patients with Cushing's syndrome and glucocorticoid- and postmenopausal- induced osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1232574. [PMID: 37881495 PMCID: PMC10597666 DOI: 10.3389/fendo.2023.1232574] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Background Skeletal stem/progenitor cells (SSPCs) in the bone marrow can differentiate into osteoblasts or adipocytes in response to microenvironmental signalling input, including hormonal signalling. Glucocorticoids (GC) are corticosteroid hormones that promote adipogenic differentiation and are endogenously increased in patients with Cushing´s syndrome (CS). Here, we investigate bone marrow adiposity changes in response to endogenous or exogenous GC increases. For that, we characterize bone biopsies from patients with CS and post-menopausal women with glucocorticoid-induced osteoporosis (GC-O), compared to age-matched controls, including postmenopausal osteoporotic patients (PM-O). Methods Transiliac crest bone biopsies from CS patients and healthy controls, and from postmenopausal women with GC-O and matched controls were analysed; an additional cohort included biopsies from women with PM-O. Plastic-embedded biopsies were sectioned for histomorphometric characterization and quantification of adipocytes. The fraction of adipocyte area per tissue (Ad.Ar/T.Ar) and marrow area (Ad.Ar/Ma.Ar), mean adipocyte profile area (Ad.Pf.Ar) and adipocyte profile density (N.Ad.Pf/Ma.Ar) were determined and correlated to steroid levels. Furthermore, the spatial distribution of adipocytes in relation to trabecular bone was characterized and correlations between bone marrow adiposity and bone remodeling parameters investigated. Results Biopsies from patients with CS and GC-O presented increased Ad.Ar/Ma.Ar, along with adipocyte hypertrophy and hyperplasia. In patients with CS, both Ad.Ar/Ma.Ar and Ad.Pf.Ar significantly correlated with serum cortisol levels. Spatial distribution analyses revealed that, in CS, the increase in Ad.Ar/Ma.Ar near to trabecular bone (<100 µm) was mediated by both adipocyte hypertrophy and hyperplasia, while N.Ad.Pf/Ma.Ar further into the marrow (>100 µm) remained unchanged. In contrast, patients with GC-O only presented increased Ad.Ar/Ma.Ar and mean Ad.Pf.Ar>100 µm from trabecular bone surface, highlighting the differential effect of increased endogenous steroid accumulation. Finally, the Ad.Ar/Ma.Ar and Ad.Ar/T.Ar correlated with the canopy coverage above remodeling events. Conclusion Increased cortisol production in patients with CS induces increased bone marrow adiposity, primarily mediated by adipocyte hypertrophy. This adiposity is particularly evident near trabecular bone surfaces, where hyperplasia also occurs. The differential pattern of adiposity in patients with CS and GC-O highlights that bone marrow adipocytes and their progenitors may respond differently in these two GC-mediated bone diseases.
Collapse
Affiliation(s)
- Nina N. Sørensen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina M. Andreasen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Danish Spatial Imaging Consortium (DanSIC), Denmark
| | - Pia R. Jensen
- Clinical Cell Biology (KCB), Vejle/Lillebaelt Hospital, Institute of Regional Health Research (IRS), University of Southern Denmark, Vejle, Denmark
| | - Ellen M. Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jean-Marie Delaissé
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Marta Diaz-delCastillo
- Danish Spatial Imaging Consortium (DanSIC), Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas L. Andersen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Danish Spatial Imaging Consortium (DanSIC), Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Shen F, Huang X, He G, Shi Y. The emerging studies on mesenchymal progenitors in the long bone. Cell Biosci 2023; 13:105. [PMID: 37301964 DOI: 10.1186/s13578-023-01039-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/01/2023] [Indexed: 06/12/2023] Open
Abstract
Mesenchymal progenitors (MPs) are considered to play vital roles in bone development, growth, bone turnover, and repair. In recent years, benefiting from advanced approaches such as single-cell sequence, lineage tracing, flow cytometry, and transplantation, multiple MPs are identified and characterized in several locations of bone, including perichondrium, growth plate, periosteum, endosteum, trabecular bone, and stromal compartment. However, although great discoveries about skeletal stem cells (SSCs) and progenitors are present, it is still largely obscure how the varied landscape of MPs from different residing sites diversely contribute to the further differentiation of osteoblasts, osteocytes, chondrocytes, and other stromal cells in their respective destiny sites during development and regeneration. Here we discuss recent findings on MPs' origin, differentiation, and maintenance during long bone development and homeostasis, providing clues and models of how the MPs contribute to bone development and repair.
Collapse
Affiliation(s)
- Fangyuan Shen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobin Huang
- Department of Oral and Maxillofacial Surgery/Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guangxu He
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, NO. 139 Middle Renmin Road, Changsha, Hunan, China.
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Everts PA, Panero AJ. Basic Science of Autologous Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:25-47. [DOI: 10.1016/j.pmr.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
13
|
Tonk CH, Shoushrah SH, Babczyk P, El Khaldi-Hansen B, Schulze M, Herten M, Tobiasch E. Therapeutic Treatments for Osteoporosis-Which Combination of Pills Is the Best among the Bad? Int J Mol Sci 2022; 23:1393. [PMID: 35163315 PMCID: PMC8836178 DOI: 10.3390/ijms23031393] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is a chronical, systemic skeletal disorder characterized by an increase in bone resorption, which leads to reduced bone density. The reduction in bone mineral density and therefore low bone mass results in an increased risk of fractures. Osteoporosis is caused by an imbalance in the normally strictly regulated bone homeostasis. This imbalance is caused by overactive bone-resorbing osteoclasts, while bone-synthesizing osteoblasts do not compensate for this. In this review, the mechanism is presented, underlined by in vitro and animal models to investigate this imbalance as well as the current status of clinical trials. Furthermore, new therapeutic strategies for osteoporosis are presented, such as anabolic treatments and catabolic treatments and treatments using biomaterials and biomolecules. Another focus is on new combination therapies with multiple drugs which are currently considered more beneficial for the treatment of osteoporosis than monotherapies. Taken together, this review starts with an overview and ends with the newest approaches for osteoporosis therapies and a future perspective not presented so far.
Collapse
Affiliation(s)
- Christian Horst Tonk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Sarah Hani Shoushrah
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Patrick Babczyk
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Basma El Khaldi-Hansen
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Margit Schulze
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| | - Monika Herten
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Edda Tobiasch
- Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, von-Liebig-Str. 20, 53359 Rheinbach, Germany; (C.H.T.); (S.H.S.); (P.B.); (B.E.K.-H.); (M.S.); (E.T.)
| |
Collapse
|
14
|
Loopmans S, Stockmans I, Carmeliet G, Stegen S. Isolation and in vitro characterization of murine young-adult long bone skeletal progenitors. Front Endocrinol (Lausanne) 2022; 13:930358. [PMID: 35979436 PMCID: PMC9376626 DOI: 10.3389/fendo.2022.930358] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Skeletal stem and progenitor cells (SSPCs) constitute a reservoir of bone-forming cells necessary for bone development, modeling and remodeling, as well as for fracture healing. Recent advances in tools to identify and isolate SSPCs have revealed that cells with multipotent properties are present not only in neonatal bone, but also in adult bone marrow and periosteum. The long bone metaphysis and endosteum have been proposed as an additional SSPC niche, although in vitro approaches to study their cellular and molecular characteristics are still limited. Here, we describe a comprehensive procedure to isolate and culture SSPCs derived from the metaphysis and endosteum of young-adult mice. Based on flow cytometry analysis of known SSPC markers, we found the presence of putative multipotent SSPCs, similar to neonatal bone tissue. In vitro, metaphyseal/endosteal SSPCs possess self-renewing capacity, and their multipotency is underscored by the ability to differentiate into the osteogenic and adipogenic lineage, while chondrogenic potential is limited. Expansion of metaphyseal/endosteal SSPCs under low oxygen conditions increases their proliferation capacity, while progenitor properties are maintained, likely reflecting their hypoxic niche in vivo. Collectively, we propose a validated isolation and culture protocol to study metaphyseal/endosteal SSPC biology in vitro.
Collapse
|
15
|
Markina EA, Alekseeva OY, Andreeva ER, Buravkova LB. Short-Term Reloading After Prolonged Unloading Ensures Restoration of Stromal but Not Hematopoietic Precursor Activity in Tibia Bone Marrow of C57Bl/6N Mice. Stem Cells Dev 2021; 30:1228-1240. [PMID: 34714129 DOI: 10.1089/scd.2021.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Bone and muscle tissues are mostly susceptible to different kinds of hypodynamia, including real and simulated microgravity (sμg). To evaluate the effect of sμg on bone marrow (BM), male C57Bl/6N mice were divided into three groups: vivarium control (VC), 30-day hindlimb suspension (HS), and subsequent 12-h short-term support reloading (RL). The effects on BM total mononucleated cells (MNCs) as well as stromal and hematopoietic progenitors from murine tibia were studied. The number of BM MNCs, immunophenotype, proliferation, colony-forming units (CFUs), differentiation and secretory activity of hematopoietic and stromal BM cells were determined. HS led to a twofold decrease in MNCs, alteration of surface molecule expression profiles, suppression of proliferative activity of BM cells, and change of soluble mediators' levels. The stromal compartment was characterized by a decrease of CFU of fibroblasts and suppression of spontaneous osteo-commitment after HS. Among the hematopoietic precursors, a decrease in the total number of CFUs was found mainly at the expense of suppression of CFU-GM and CFU-GEMM. After RL, restoration of the stromal precursor's functional activity to control levels and overabundance of paracrine mediator's production were detected, whereas the complete recovery of hematopoietic precursor's activity did not occur. These data demonstrate the fast functional reaction of the stromal compartment on restoration of loading support.
Collapse
Affiliation(s)
- Elena A Markina
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russian Federation
| | - Olga Y Alekseeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russian Federation
| | - Elena R Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russian Federation
| | - Ludmila B Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
16
|
Inoue S, Takito J, Nakamura M. Site-Specific Fracture Healing: Comparison between Diaphysis and Metaphysis in the Mouse Long Bone. Int J Mol Sci 2021; 22:ijms22179299. [PMID: 34502206 PMCID: PMC8430651 DOI: 10.3390/ijms22179299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
The process of fracture healing varies depending upon internal and external factors, such as the fracture site, mode of injury, and mechanical environment. This review focuses on site-specific fracture healing, particularly diaphyseal and metaphyseal healing in mouse long bones. Diaphyseal fractures heal by forming the periosteal and medullary callus, whereas metaphyseal fractures heal by forming the medullary callus. Bone healing in ovariectomized mice is accompanied by a decrease in the medullary callus formation both in the diaphysis and metaphysis. Administration of estrogen after fracture significantly recovers the decrease in diaphyseal healing but fails to recover the metaphyseal healing. Thus, the two bones show different osteogenic potentials after fracture in ovariectomized mice. This difference may be attributed to the heterogeneity of the skeletal stem cells (SSCs)/osteoblast progenitors of the two bones. The Hox genes that specify the patterning of the mammalian skeleton during embryogenesis are upregulated during the diaphyseal healing. Hox genes positively regulate the differentiation of osteoblasts from SSCs in vitro. During bone grafting, the SSCs in the donor’s bone express Hox with adaptability in the heterologous bone. These novel functions of the Hox genes are discussed herein with reference to the site-specificity of fracture healing.
Collapse
|
17
|
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
|
18
|
Mullis BH, Gudeman AS, Borrelli J, Crist BD, Lee MA, Evans AR. Bone healing: Advances in biology and technology. OTA Int 2021; 4:e100(1-5). [PMID: 37608854 PMCID: PMC10441680 DOI: 10.1097/oi9.0000000000000100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 08/24/2023]
Abstract
Fracture healing is a complex cascade of cellular and molecular processes. These processes require the appropriate cellular and molecular environment to ensure the restoration of skeletal stability and resolution of inflammation. In order for fracture healing to occur, the necessary building blocks for bone metabolism and synthesis must be supplied through proper nutrition. Pharmacologic therapies aimed at modulating the inflammatory response to fractures have the potential to interfere with the synthesis of molecules needed for the production of bone. Infection can interfere with, and even prevent normal fracture healing from occurring. Cellular and genetic treatment strategies are actively being developed to target deficiencies, and bridge gaps that can influence how fractures heal. Evolving technologies, including nutritional supplementation, pharmacotherapies, antibiotics, surgical techniques, as well as genetic and cellular therapies, have the potential to enhance, optimize, and even revolutionize the process of fracture healing.
Collapse
Affiliation(s)
- Brian H Mullis
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew S Gudeman
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN
| | - Joseph Borrelli
- Department of Orthopaedic Surgery and Sports Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Brett D Crist
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO
| | - Mark A Lee
- Department of Orthopaedic Surgery, University of California - Davis, CA
| | - Andrew R Evans
- Department of Orthopedics, The Warren Alpert School of Medicine, Brown University/Rhode Island Hospital, Providence, RI
| |
Collapse
|
19
|
Muscarella AM, Aguirre S, Hao X, Waldvogel SM, Zhang XHF. Exploiting bone niches: progression of disseminated tumor cells to metastasis. J Clin Invest 2021; 131:143764. [PMID: 33720051 PMCID: PMC7954594 DOI: 10.1172/jci143764] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Many solid cancers metastasize to the bone and bone marrow (BM). This process may occur even before the diagnosis of primary tumors, as evidenced by the discovery of disseminated tumor cells (DTCs) in patients without occult malignancies. The cellular fates and metastatic progression of DTCs are determined by complicated interactions between cancer cells and BM niches. Not surprisingly, these niches also play important roles in normal biology, including homeostasis and turnover of skeletal and hematopoiesis systems. In this Review, we summarize recent findings on functions of BM niches in bone metastasis (BoMet), particularly during the early stage of colonization. In light of the rich knowledge of hematopoiesis and osteogenesis, we highlight how DTCs may progress into overt BoMet by taking advantage of niche cells and their activities in tissue turnover, especially those related to immunomodulation and bone repair.
Collapse
Affiliation(s)
- Aaron M. Muscarella
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah M. Waldvogel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
20
|
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
|
21
|
Zhong L, Yao L, Qin L. A Novel Enzymatic Digestion Approach for Isolation and Culture of Rodent Bone Marrow Mesenchymal Progenitors. Methods Mol Biol 2021; 2221:29-39. [PMID: 32979196 DOI: 10.1007/978-1-0716-0989-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Bone marrow mesenchymal stem cells (MSCs) are promising therapeutic tools for tissue repair and treatment of a number of human diseases. As a result, there is substantial interest in characterizing and expanding these cells to uncover their therapeutic potential. Bone marrow mesenchymal progenitors, containing both MSCs and their proliferative progeny, are commonly isolated from the central region of rodent long bones. However, challenges exist in expanding these central mesenchymal progenitors in culture. We have designed an enzymatic digestion protocol to isolate mesenchymal progenitors within rodent long bones that resides close to the bone surface, which we termed endosteal mesenchymal progenitors. These cells are more metabolically active and more responsive to external stimuli compared to central mesenchymal progenitors. Therefore, they represent a biologically important target for MSC research. This chapter describes the approach in detail how to isolate and culture endosteal mesenchymal progenitors as well as their central counterparts from rodent long bones.
Collapse
Affiliation(s)
- Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Rossetti R, Rós FA, Souza LEBD, Maçonetto JDM, Costa PNMD, Ferreira FU, Borges JS, Carvalho JVD, Morotti NP, Kashima S, Covas DT. Hypoxia-cultured mouse mesenchymal stromal cells from bone marrow and compact bone display different phenotypic traits. Exp Cell Res 2020; 399:112434. [PMID: 33340494 DOI: 10.1016/j.yexcr.2020.112434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/10/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022]
Abstract
It has been suggested that the bone marrow microenvironment harbors two distinct populations of mesenchymal stromal cells (MSC), one with a perivascular location and other present in the endosteum. A better understanding of the biology of these MSC subsets has been pursued in order to refine its clinical application. However, most comparative characterizations of mouse MSC have been performed in normoxia. This can result in misleading interpretations since mouse MSC subsets with low/defective p53 activity are known to be selected during culture in normoxia. Here, we report a comprehensive in vitro characterization of mouse MSC isolated from bone marrow (BM-MSC) and compact bone (CB-MSC) expanded and assayed under hypoxia for their morphology, clonogenic efficiency and differentiation capacity. We found that, under hypoxia, compact bone is richer in absolute numbers of MSC and isolation of MSC from compact bone is associated with a reduced risk of hematopoietic cell carryover. In addition, CB-MSC have higher in vitro osteogenic capacity than BM-MSC, while adipogenic differentiation potential is similar. These findings reinforce the hypothesis of the existence of MSC in bone marrow and compact bone representing functionally distinct cell populations and highlight the compact bone as an efficient source of murine MSC under physiological oxygen concentrations.
Collapse
Affiliation(s)
- Rafaela Rossetti
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil.
| | - Felipe Augusto Rós
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Eduardo Botelho de Souza
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Juliana de Matos Maçonetto
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Péricles Natan Mendes da Costa
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Fernanda Ursoli Ferreira
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Josiane Serrano Borges
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Julianne Vargas de Carvalho
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Nayara Patrícia Morotti
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Simone Kashima
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Blood Center of Ribeirão Preto - Ribeirão Preto Medical School, University of São Paulo, 2501 Tenente Catão Roxo Avenue, 14051-060, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
23
|
Etani Y, Ebina K, Hirao M, Kitaguchi K, Kashii M, Ishimoto T, Nakano T, Okamura G, Miyama A, Takami K, Goshima A, Kanamoto T, Nakata K, Yoshikawa H. Combined effect of teriparatide and an anti-RANKL monoclonal antibody on bone defect regeneration in mice with glucocorticoid-induced osteoporosis. Bone 2020; 139:115525. [PMID: 32645445 DOI: 10.1016/j.bone.2020.115525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/21/2020] [Accepted: 07/02/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE The purpose of this study was to examine the effect of single or combination therapy of teriparatide (TPTD) and a monoclonal antibody against the murine receptor activator of nuclear factor κB ligand (anti-RANKL Ab) on cancellous and cortical bone regeneration in a mouse model of glucocorticoid-induced osteoporosis (GIOP). METHODS C57BL/6 J mice (24 weeks of age) were divided into five groups: (1) the SHAM group: sham operation + saline; (2) the prednisolone (PSL) group: PSL + saline; (3) the TPTD group: PSL + TPTD; (4) the Ab group: PSL + anti-RANKL Ab; and (5) the COMB group: PSL + TPTD + anti-RANKL Ab (n = 8 per group). With the exception of the SHAM group, 7.5 mg of PSL was inserted subcutaneously into mice, to generate a mouse model of GIOP. Four weeks after insertion, bone defects with a diameter of 0.9 mm were created to assess bone regeneration on both femoral metaphysis (cancellous bone) and diaphysis (cortical bone). After surgery, therapeutic intervention was continued for 4 weeks. Saline (200 μl) or TPTD (40 μg/kg) was injected subcutaneously five times per week, whereas the anti-RANKL Ab (5 mg/kg) was injected subcutaneously once on the day after surgery. Subsequently, the following analyses were performed: microstructural assessment of bone regeneration and bone mineral density (BMD) measurement via micro-computed tomography, and histological, histomorphometrical, and biomechanical analyses with nanoindentation. RESULTS The COMB group showed the highest lumbar spine BMD increase (vs. the PSL, TPTD, and Ab groups). The volume of regenerated cancellous bone at the bone defect site was higher in the COMB group compared with the PSL, TPTD, and Ab group. The volume of the regenerated cortical bone was significantly higher in the COMB group compared with the PSL group, and its hardness was significantly higher in the COMB group compared with the PSL and TPTD groups. CONCLUSION In a mouse model of glucocorticoid-induced osteoporosis, the combination therapy of TPTD plus the anti-RANKL Ab increased bone mineral density in the lumbar spine and regenerated cancellous bone volume compared with single administration of each agent, and also increased regenerated cortical bone strength compared with single administration of TPTD.
Collapse
Affiliation(s)
- Yuki Etani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | - Makoto Hirao
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kazuma Kitaguchi
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| | - Masafumi Kashii
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| | - Takuya Ishimoto
- Division of Materials and Manufacturing Science, Osaka University Graduate School of Engineering, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Osaka University Graduate School of Engineering, 2-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Gensuke Okamura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Akira Miyama
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kenji Takami
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Atsushi Goshima
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Takashi Kanamoto
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka 560-8565, Japan
| |
Collapse
|
24
|
Johnstone BH, Miller HM, Beck MR, Gu D, Thirumala S, LaFontaine M, Brandacher G, Woods EJ. Identification and characterization of a large source of primary mesenchymal stem cells tightly adhered to bone surfaces of human vertebral body marrow cavities. Cytotherapy 2020; 22:617-628. [PMID: 32873509 PMCID: PMC8919862 DOI: 10.1016/j.jcyt.2020.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/12/2020] [Accepted: 07/05/2020] [Indexed: 12/13/2022]
Abstract
Background: Therapeutic allogeneic mesenchymal stromal cells (MSCs) are currently in clinical trials to evaluate their effectiveness in treating many different disease indications. Eventual commercialization for broad distribution will require further improvements in manufacturing processes to economically manufacture MSCs at scales sufficient to satisfy projected demands. A key contributor to the present high cost of goods sold for MSC manufacturing is the need to create master cell banks from multiple donors, which leads to variability in large-scale manufacturing runs. Therefore, the availability of large single donor depots of primary MSCs would greatly benefit the cell therapy market by reducing costs associated with manufacturing. Methods: We have discovered that an abundant population of cells possessing all the hallmarks of MSCs is tightly associated with the vertebral body (VB) bone matrix and only liberated by proteolytic digestion. Here we demonstrate that these vertebral bone-adherent (vBA) MSCs possess all the International Society of Cell and Gene Therapy-defined characteristics (e.g., plastic adherence, surface marker expression and trilineage differentiation) of MSCs, and we have therefore termed them vBA-MSCs to distinguish this population from loosely associated MSCs recovered through aspiration or rinsing of the bone marrow compartment. Results: Pilot banking and expansion were performed with vBA-MSCs obtained from 3 deceased donors, and it was demonstrated that bank sizes averaging 2.9 × 108 ± 1.35 × 108 vBA-MSCs at passage 1 were obtainable from only 5 g of digested VB bone fragments. Each bank of cells demonstrated robust proliferation through a total of 9 passages, without significant reduction in population doubling times. The theoretical total cell yield from the entire amount of bone fragments (approximately 300 g) from each donor with limited expansion through 4 passages is 100 trillion (1 × 1014) vBA-MSCs, equating to over 105 doses at 10 × 106 cells/kg for an average 70-kg recipient. Discussion: Thus, we have established a novel and plentiful source of MSCs that will benefit the cell therapy market by overcoming manufacturing and regulatory inefficiencies due to donor-to-donor variability.
Collapse
Affiliation(s)
- Brian H Johnstone
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA.
| | - Hannah M Miller
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Madelyn R Beck
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dongsheng Gu
- Ossium Health, Inc, Indianapolis, Indiana, USA; Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sreedhar Thirumala
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael LaFontaine
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erik J Woods
- Ossium Health, Inc, Indianapolis, Indiana, USA; Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, Indiana, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
25
|
α-Hemolysin suppresses osteogenesis by inducing lipid rafts accumulation in bone marrow stromal cells. Food Chem Toxicol 2020; 145:111689. [PMID: 32810588 DOI: 10.1016/j.fct.2020.111689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 07/20/2020] [Accepted: 08/11/2020] [Indexed: 01/15/2023]
Abstract
α-hemolysin (Hla) is considered an essential virulent factor for Staphylococcus aureus (S. aureus) toxicity, the mechanism by which Hla affect bone metabolism is poorly understood. In this study, 2-month-old C57BL/6 mice were treated with Hla (40 μg/kg, i.p.) or S. aureus (1 × 106 CFU/ml, 100 μl, i.v.) with the presence or absence of methyl-β-cyclodextrin (MβCD) (300 mg/kg, i.p.). MicroCT analysis showed progressive bone loss from week 2 to week 4 after Hla treatment, accompanied by a decreased osteoblasts and increased osteoclasts in femoral metaphysis in mice. Further, Hla stimulated the expression of Caveolin-1 in vivo and in vitro, activated lipid rafts accumulation in cell membrane of bone marrow stromal cells (BMSCs), and suppressed osteogenesis of BMSCs. Destruction of lipid rafts with MβCD or inhibition of Caveolin-1 with Daidzein blocked the detrimental effect of Hla on osteogenesis of BMSCs. Importantly, treating mice with MβCD rescued the loss of osteoblasts and increased osteoclastogenesis induced by Hla as well as the bone loss induced by S. aureus infection. Together, we demonstrate that Hla induces bone destruction directly by suppressing osteogenesis and indirectly by stimulating osteoclastogenesis, and that lipid rafts may mediate the detrimental effect of Hla and S. aureus on osteogenesis and bone formation.
Collapse
|
26
|
Zhang X, Wang S, Ding X, Guo J, Tian Z. Potential methods for improving the efficacy of mesenchymal stem cells in the treatment of inflammatory bowel diseases. Scand J Immunol 2020; 92:e12897. [PMID: 32443180 DOI: 10.1111/sji.12897] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic recurrent gastrointestinal inflammatory diseases, including ulcerative colitis (UC), Crohn's disease (CD) and IBD unclassified. The pathogenesis may be related to the mucosal immune dysfunction in genetically susceptible hosts affected by environmental factors. Current therapeutic agents mainly include aminosalicylates, corticosteroids, immunosuppressive drugs and novel biological agents. The purpose of treatment is to suppress inflammation and prevent irreversible structural damage. However, long-term application of these drugs may lead to multiple adverse effects and is not always effective. Mesenchymal stem cells (MSCs) are multipotent progenitors with low immunogenicity, which can be obtained and expanded easily. They play an important role in regulating immune responses and repairing damaged tissues in vivo. Therefore, MSCs are considered to be a promising option for the treatment of IBD. Nonetheless, there are many factors that can reduce the efficacy of MSCs, such as gradual deterioration of functional stem cells with age, low recruitment and persistence in vivo and different routes of administration. In recent years, researchers have been able to improve the efficacy of MSCs by pretreatment, genetic modification or co-application with other substances, as well as using different tissue-derived cells, administration methods or doses. This article reviews these methods to provide references for more effective application of MSCs in the treatment of IBD in the future.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shaojun Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueli Ding
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Guo
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
27
|
Chang H, Xiang H, Yao Z, Yang S, Tu M, Zhang X, Yu B. Strontium-substituted calcium sulfate hemihydrate/hydroxyapatite scaffold enhances bone regeneration by recruiting bone mesenchymal stromal cells. J Biomater Appl 2020; 35:97-107. [PMID: 32233720 DOI: 10.1177/0885328220915816] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fabrication of osteoconductive scaffold with osteoinductive capability and appropriate resorption rate is of great significance for treating bone defects. To achieve this aim, strontium-substituted calcium sulfate hemihydrate (Sr-CSH) and hydroxyapatite (HA) were mixed to develop a novel composite. Sr-CSH containing 5% and 10% strontium was mixed with HA at the weight ratio of 6:4, respectively. Female Sprague-Dawley rats underwent bone defect surgery in left tibia were randomly assigned to three different treatment groups filled with CSH/HA, 5% and 10% Sr-CSH/HA. Micro-CT analysis showed increased new bone formation in 10% Sr-CSH/HA group compared to CSH/HA group. In addition, histological analysis showed large amounts of chondrocytes and osteoblasts within the pores of Sr-CSH/HA composites as a result of the CSH resorption. Further, CFU-F assay demonstrated the increased amount of bone marrow mesenchymal stromal cells (BMSCs) colonies in 10% Sr-CSH/HA group. In primary BMSCs, extraction from Sr-CSH/HA composite significantly increased the migration of cells, up-regulated the expression of osteoblastic marker genes, and increased the area of mineralized nodules. Together, Sr-CSH/HA may promote bone formation by recruiting and stimulating osteogenic differentiation of BMSCs. Therefore, this composite may be proposed as an ideal substitute to repair bone defects.
Collapse
Affiliation(s)
- Hong Chang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopaedics, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Haibo Xiang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Orthopaedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zilong Yao
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shenyu Yang
- Laboratory of Materials Science and Engineering, Academy of Chemistry and Materials, Jinan University, Guangzhou, China
| | - Mei Tu
- Laboratory of Materials Science and Engineering, Academy of Chemistry and Materials, Jinan University, Guangzhou, China
| | - Xianrong Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
28
|
Hu Y, Wu H, Xu T, Wang Y, Qin H, Yao Z, Chen P, Xie Y, Ji Z, Yang K, Chai Y, Zhang X, Yu B, Cui Z. Defactinib attenuates osteoarthritis by inhibiting positive feedback loop between H-type vessels and MSCs in subchondral bone. J Orthop Translat 2020; 24:12-22. [PMID: 32518750 PMCID: PMC7261948 DOI: 10.1016/j.jot.2020.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/24/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022] Open
Abstract
Background Abnormal bone formation in subchondral bone resulting from uncoupled bone remodeling is considered a central feature in osteoarthritis (OA) pathogenesis. H-type vessels can couple angiogenesis and osteogenesis. We previously revealed that elevated H-type vessels in subchondral bone were correlated with OA and focal adhesion kinase (FAK) in MSCs is critical for H-type vessel formation in osteoporosis. The aim of this study was to explore the correlation between H-type vessels and MSCs in OA pathogenesis through regulation of H-type vessel formation using defactinib (an FAK inhibitor). Methods In vivo: 3-month-old male C57BL/6J (WT) mice were randomly divided into three groups: sham controls, vehicle-treated ACLT mice, and defactinib-treated ACLT mice (25 mg/kg, intraperitoneally weekly). In vitro: we explored the role of conditioned medium (CM) of MSCs from subchondral bone of different groups on the angiogenesis of endothelial cells (ECs). Flow cytometry, Western blotting, ELISA, real time (RT)-PCR, immunostaining, CT-based microangiography, and bone micro-CT (μCT) were used to detect changes in relative cells and tissues. Results This study demonstrated that inhibition of H-type vessels with defactinib alleviated OA by inhibiting H-type vessel-linked MSCs in subchondral bone. During OA pathogenesis, H-type vessels and MSCs formed a positive feedback loop contributing to abnormal bone formation in subchondral bone. Elevated H-type vessels provided indispensable MSCs for abnormal bone formation in subchondral bone. Flow cytometry and immunostaining results confirmed that the amount of MSCs in subchondral bone was obviously higher in vehicle-treated ACLT mice than that in sham controls and defactinib-treated ACLT mice. In vitro, p-FAK in MSCs from subchondral bone of vehicle-treated ALCT mice increased significantly relative to other groups. Further, the CM from MSCs of vehicle-treated ACLT mice enhanced angiogenesis of ECs through FAK-Grb2-MAPK-linked VEGF expression. Conclusions Our results demonstrate that defactinib inhibits OA by suppressing the positive feedback loop between H-type vessels and MSCs in subchondral bone. The translational potential of this article Our results provide a mechanistic rationale for the use of defactinib as an effective candidate for OA treatment.
Collapse
Affiliation(s)
- Yanjun Hu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hangtian Wu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Ting Xu
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yutian Wang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hanjun Qin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zilong Yao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Peisheng Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian 350007, China
| | - Yongheng Xie
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhiguo Ji
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Kaifan Yang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu Chai
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Department of Orthopedic Surgery, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xianrong Zhang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Bin Yu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhuang Cui
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.,Key Laboratory of Bone and Cartilage Regeneration Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| |
Collapse
|
29
|
Wu H, Xu T, Chen Z, Wang Y, Li K, Chen PS, Yao Z, Su J, Cheng C, Wu X, Zhang H, Chai Y, Zhang X, Hu Y, Yu B, Cui Z. Specific inhibition of FAK signaling attenuates subchondral bone deterioration and articular cartilage degeneration during osteoarthritis pathogenesis. J Cell Physiol 2020; 235:8653-8666. [PMID: 32324278 DOI: 10.1002/jcp.29709] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA), a disease of the entire joint, is characterized by abnormal bone remodeling and coalescent degradation of articular cartilage. We have previously found that elevated levels of H-type vessels in subchondral bone correlate with OA and that focal adhesion kinase (FAK) is critical for H-type vessel formation in osteoporosis. However, the potential role of FAK in OA remains unexplored. Here, we demonstrate that the p-FAK level was dramatically elevated in subchondral bone following anterior cruciate ligament transection (ACLT) in rats. Specific inhibition of FAK signaling with Y15 in subchondral bone resulted in the suppression of subchondral bone deterioration and this effect was mediated by H-type vessel-induced ectopic bone formation. Further, articular cartilage degeneration was also alleviated after Y15 treatment. In vitro, the p-FAK level was significantly elevated in mesenchymal stem cells (MSCs) from vehicle-treated ACLT rats as compared to that in MSCs from sham controls and Y15-treated ACLT rats. Elevated p-FAK level in MSCs promoted vascular endothelial growth factor (VEGF) expression, as demonstrated from the high VEGF level in the blood, subchondral bone, and conditioned medium (CM) of MSCs from vehicle-treated ACLT rats. The CM of MSCs from vehicle-treated ACLT rats might promote the angiogenesis of endothelial cells and the catabolic response of chondrocytes through the FAK-growth factor receptor-bound protein 2-mitogen-activated protein kinase-mediated expression of VEGF. The effect of the CM from MSCs of Y15-treated ACLT rats or that treated with a VEGF-neutralizing antibody on vessel formation and the catabolic response was lowered. Thus, the specific inhibition of FAK signaling may be a promising avenue for the prevention or early treatment of OA.
Collapse
Affiliation(s)
- Hangtian Wu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Xu
- Department of Sleep Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhigang Chen
- Department of Orthopaedics and Traumatology, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yutian Wang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaiqun Li
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pei-Sheng Chen
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| | - Zilong Yao
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianwen Su
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Caiyu Cheng
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaohu Wu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongan Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Chai
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedic Surgery, Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Xianrong Zhang
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanjun Hu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Yu
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhuang Cui
- Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Zhong L, Yao L, Tower RJ, Wei Y, Miao Z, Park J, Shrestha R, Wang L, Yu W, Holdreith N, Huang X, Zhang Y, Tong W, Gong Y, Ahn J, Susztak K, Dyment N, Li M, Long F, Chen C, Seale P, Qin L. Single cell transcriptomics identifies a unique adipose lineage cell population that regulates bone marrow environment. eLife 2020; 9:e54695. [PMID: 32286228 PMCID: PMC7220380 DOI: 10.7554/elife.54695] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
Bone marrow mesenchymal lineage cells are a heterogeneous cell population involved in bone homeostasis and diseases such as osteoporosis. While it is long postulated that they originate from mesenchymal stem cells, the true identity of progenitors and their in vivo bifurcated differentiation routes into osteoblasts and adipocytes remain poorly understood. Here, by employing large scale single cell transcriptome analysis, we computationally defined mesenchymal progenitors at different stages and delineated their bi-lineage differentiation paths in young, adult and aging mice. One identified subpopulation is a unique cell type that expresses adipocyte markers but contains no lipid droplets. As non-proliferative precursors for adipocytes, they exist abundantly as pericytes and stromal cells that form a ubiquitous 3D network inside the marrow cavity. Functionally they play critical roles in maintaining marrow vasculature and suppressing bone formation. Therefore, we name them marrow adipogenic lineage precursors (MALPs) and conclude that they are a newly identified component of marrow adipose tissue.
Collapse
Affiliation(s)
- Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, The First Hospital of China Medical UniversityShenyangChina
| | - Robert J Tower
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yulong Wei
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhen Miao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Rojesh Shrestha
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Shandong University Qilu Hospital, Shandong UniversityJinanChina
| | - Wei Yu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Nicholas Holdreith
- Division of Hematology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Xiaobin Huang
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Yejia Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaUnited States
| | - Wei Tong
- Division of Hematology, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Yanqing Gong
- Division of Transnational Medicine and Human Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jaimo Ahn
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of PennsylvaniaPhiladelphiaUnited States
| | - Nathanial Dyment
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Fanxin Long
- Translational Research Program in Pediatric Orthopaedics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery/Pharmacology, University of Pennsylvania, School of Dental MedicinePhiladelphiaUnited States
| | - Patrick Seale
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
31
|
Root SH, Wee NKY, Novak S, Rosen CJ, Baron R, Matthews BG, Kalajzic I. Perivascular osteoprogenitors are associated with transcortical channels of long bones. Stem Cells 2020; 38:769-781. [PMID: 32053258 DOI: 10.1002/stem.3159] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022]
Abstract
Bone remodeling and regeneration are dependent on resident stem/progenitor cells with the ability to replenish mature osteoblasts and repair the skeleton. Using lineage tracing approaches, we identified a population of Dmp1+ cells that reside within cortical bone and are distinct from osteocytes. Our aims were to characterize this stromal population of transcortical perivascular cells (TPCs) in their resident niche and evaluate their osteogenic potential. To distinguish this population from osteoblasts/osteocytes, we crossed mice containing inducible DMP1CreERT2/Ai9 Tomato reporter (iDMP/T) with Col2.3GFP reporter (ColGFP), a marker of osteoblasts and osteocytes. We observed iDMP/T+;ColGFP- TPCs within cortical bone following tamoxifen injection. These cells were perivascular and located within transcortical channels. Ex vivo bone outgrowth cultures showed TPCs migrated out of the channels onto the plate and expressed stem cell markers such as Sca1, platelet derived growth factor receptor beta (PDGFRβ), and leptin receptor. In a cortical bone transplantation model, TPCs migrate from their vascular niche within cortical bone and contribute to new osteoblast formation and bone tube closure. Treatment with intermittent parathyroid hormone increased TPC number and differentiation. TPCs were unable to differentiate into adipocytes in the presence of rosiglitazone in vitro or in vivo. Altogether, we have identified and characterized a novel stromal lineage-restricted osteoprogenitor that is associated with transcortical vessels of long bones. Functionally, we have demonstrated that this population can migrate out of cortical bone channels, expand, and differentiate into osteoblasts, therefore serving as a source of progenitors contributing to new bone formation.
Collapse
Affiliation(s)
- Sierra H Root
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, USA
| | - Natalie K Y Wee
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, USA
| | - Sanja Novak
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, USA
| | - Clifford J Rosen
- Department of Medicine, Tufts University School of Medicine, Maine Medical Center Research Institute, Scarborough, Maine, USA
| | - Roland Baron
- Department of Oral Medicine, Infection and Immunity, Division of Bone and Mineral Research, Harvard School of Dental Medicine, Boston, Massachusetts, USA.,Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Brya G Matthews
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, USA.,Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
32
|
Perlbach R, Palm L, Mohaddes M, Ivarsson I, Schilcher J. Good implant survival after acetabular revision with extensive impaction bone grafting and uncemented components. Bone Joint J 2020; 102-B:198-204. [DOI: 10.1302/0301-620x.102b2.bjj-2019-0584.r2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Aims This single-centre observational study aimed to describe the results of extensive bone impaction grafting of the whole acetabular cavity in combination with an uncemented component in acetabular revisions performed in a standardized manner since 1993. Methods Between 1993 and 2013, 370 patients with a median age of 72 years (interquartile range (IQR) 63 to 79 years) underwent acetabular revision surgery. Of these, 229 were more than ten years following surgery and 137 were more than 15 years. All revisions were performed with extensive use of morcellized allograft firmly impacted into the entire acetabular cavity, followed by insertion of an uncemented component with supplementary screw fixation. All types of reoperation were captured using review of radiographs and medical charts, combined with data from the local surgical register and the Swedish Hip Arthroplasty Register. Results Among patients with possible follow-up of ten and 15 years, 152 and 72 patients remained alive without revision of the acetabular component. The number of deaths was 61 and 50, respectively. Of those who died, six patients in each group had a reoperation performed before death. The number of patients with a reoperation was 22 for those with ten-year follow-up and 21 for those with 15 years of follow-up. The Kaplan-Meier implant survival rate for aseptic loosening among all 370 patients in the cohort was 96.3% (95% confidence interval (CI) 94.1 to 98.5) after ten years and 92.8% (95% CI 89.2 to 96.6) after 15 years. Conclusion Extensive bone impaction grafting combined with uncemented revision components appears to be a reliable method with favourable long-term survival. This technique offers the advantage of bone stock restoration and disputes the long-standing perception that uncemented components require > 50% of host bone contact for successful implant survival. Cite this article: Bone Joint J 2020;102-B(2):198–204.
Collapse
Affiliation(s)
- Rico Perlbach
- Department of Orthopedic Surgery and Department of Clinical and Experimental Medicine, University Hospital Linköping, Linköping University, Linköping, Sweden
| | - Lars Palm
- Department of Orthopedic Surgery and Department of Clinical and Experimental Medicine, University Hospital Linköping, Linköping University, Linköping, Sweden
| | - Maziar Mohaddes
- Department of Orthopedics, Institute of Clinical Sciences, The Sahlgrenska Academy
- University of Gothenburg, Swedish Hip Arthroplasty Register, Registercentrum VGR, Gothenburg, Sweden
| | - Ingemar Ivarsson
- Department of Orthopedic Surgery and Department of Clinical and Experimental Medicine, University Hospital Linköping, Linköping University, Linköping, Sweden
| | - Jörg Schilcher
- Department of Orthopedic Surgery and Department of Clinical and Experimental Medicine, University Hospital Linköping, Linköping University, Linköping, Sweden
| |
Collapse
|
33
|
Kitaguchi K, Kashii M, Ebina K, Kaito T, Okada R, Makino T, Etani Y, Ishimoto T, Nakano T, Yoshikawa H. The combined effects of teriparatide and anti-RANKL monoclonal antibody on bone defect regeneration in ovariectomized mice. Bone 2020; 130:115077. [PMID: 31622773 DOI: 10.1016/j.bone.2019.115077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/23/2019] [Accepted: 09/23/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVE The purpose of this study was to investigate the combined effects of teriparatide (TPTD) and anti-murine receptor activator of nuclear factor-κB ligand monoclonal antibody (anti-RANKL Ab) on both cancellous and cortical bone healing in ovariectomized mice. METHODS Thirteen-week-old mice were divided into the sham-operated group (n=11) or the ovariectomized group (n=44). At 1 month post-operation, all mice underwent bone defect surgery on the left femoral metaphysis (cancellous bone healing model) and right femoral mid-diaphysis (cortical bone healing model). After creating the bone defects, all ovariectomized mice were assigned to one of four groups to receive 1) saline (5 times a week; CNT group), 2) TPTD (40μg/kg 5 times a week; TPTD group), 3) anti-RANKL Ab (5mg/kg once; Ab group), or 4) a combination of TPTD and anti-RANKL Ab (COMB group). The following analyses were performed: Time-course microstructural analysis of healing in both cancellous and cortical bone in the bone defect, measuring the volumetric bone mineral density and the cortical bone thickness of the tibia as a representative of whole body bone with the use of micro-computed tomography, and histological analysis. RESULTS Regeneration of cancellous bone volume in the COMB group was the highest among the four groups, and combined treatment accelerated the formation of medullary callus during the early phase of bone regeneration. On the other hand, there were no significant differences in the regeneration of cortical bone volume during the early phase of bone regeneration among the four groups. Furthermore, lamellar bone was not well identified in the all four groups. Volumetric bone mineral density in the tibia in the COMB group was significantly higher compared with that in the CNT and TPTD groups and tended to be higher compared with that in the Ab group. The mean values of cortical bone thickness in the TPTD and COMB groups were significantly higher than that in the CNT group. CONCLUSION In a mouse model of postmenopausal osteoporosis, combination therapy of TPTD and anti-RANKL Ab accelerates regeneration of cancellous bone more effectively than either agent alone during the early phase of bone regeneration.
Collapse
Affiliation(s)
- Kazuma Kitaguchi
- Department of Orthopedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharamachi, Toyonaka, 560-8565, Japan; Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Masafumi Kashii
- Department of Orthopedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharamachi, Toyonaka, 560-8565, Japan; Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kosuke Ebina
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Rintaro Okada
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takahiro Makino
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Yuki Etani
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takuya Ishimoto
- Division of Materials and Manufacturing Science, Graduate School Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Takayoshi Nakano
- Division of Materials and Manufacturing Science, Graduate School Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Hideki Yoshikawa
- Department of Orthopedic Surgery, Toyonaka Municipal Hospital, 4-14-1 Shibaharamachi, Toyonaka, 560-8565, Japan; Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
34
|
Yang M, Arai A, Udagawa N, Zhao L, Nishida D, Murakami K, Hiraga T, Takao-Kawabata R, Matsuo K, Komori T, Kobayashi Y, Takahashi N, Isogai Y, Ishizuya T, Yamaguchi A, Mizoguchi T. Parathyroid Hormone Shifts Cell Fate of a Leptin Receptor-Marked Stromal Population from Adipogenic to Osteoblastic Lineage. J Bone Miner Res 2019; 34:1952-1963. [PMID: 31173642 DOI: 10.1002/jbmr.3811] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/28/2022]
Abstract
Intermittent parathyroid hormone (iPTH) treatment induces bone anabolic effects that result in the recovery of osteoporotic bone loss. Human PTH is usually given to osteoporotic patients because it induces osteoblastogenesis. However, the mechanism by which PTH stimulates the expansion of stromal cell populations and their maturation toward the osteoblastic cell lineage has not be elucidated. Mouse genetic lineage tracing revealed that iPTH treatment induced osteoblastic differentiation of bone marrow (BM) mesenchymal stem and progenitor cells (MSPCs), which carried the leptin receptor (LepR)-Cre. Although these findings suggested that part of the PTH-induced bone anabolic action is exerted because of osteoblastic commitment of MSPCs, little is known about the in vivo mechanistic details of these processes. Here, we showed that LepR+ MSPCs differentiated into type I collagen (Col1)+ mature osteoblasts in response to iPTH treatment. Along with osteoblastogenesis, the number of Col1+ mature osteoblasts increased around the bone surface, although most of them were characterized as quiescent cells. However, the number of LepR-Cre-marked lineage cells in a proliferative state also increased in the vicinity of bone tissue after iPTH treatment. The expression levels of SP7/osterix (Osx) and Col1, which are markers for osteoblasts, were also increased in the LepR+ MSPCs population in response to iPTH treatment. In contrast, the expression levels of Cebpb, Pparg, and Zfp467, which are adipocyte markers, decreased in this population. Consistent with these results, iPTH treatment inhibited 5-fluorouracil- or ovariectomy (OVX)-induced LepR+ MSPC-derived adipogenesis in BM and increased LepR+ MSPC-derived osteoblasts, even under the adipocyte-induced conditions. Treatment of OVX rats with iPTH significantly affected the osteoporotic bone tissue and expansion of the BM adipose tissue. These results indicated that iPTH treatment induced transient proliferation of the LepR+ MSPCs and skewed their lineage differentiation from adipocytes toward osteoblasts, resulting in an expanded, quiescent, and mature osteoblast population. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mengyu Yang
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| | - Atsushi Arai
- Department of Orthodontics, Matsumoto Dental University, Nagano, Japan
| | - Nobuyuki Udagawa
- Department of Oral Biochemistry, Matsumoto Dental University, Nagano, Japan
| | - Lijuan Zhao
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| | - Daisuke Nishida
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| | - Kohei Murakami
- Department of Oral Biochemistry, Matsumoto Dental University, Nagano, Japan
| | - Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, Nagano, Japan
| | - Ryoko Takao-Kawabata
- Laboratory for Pharmacology, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Koichi Matsuo
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, Tokyo, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Unit of Basic Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Naoyuki Takahashi
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan
| | - Yukihiro Isogai
- Laboratory for Pharmacology, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Toshinori Ishizuya
- Laboratory for Pharmacology, Pharmaceutical Research Center, Asahi Kasei Pharma Corporation, Shizuoka, Japan
| | - Akira Yamaguchi
- Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| | - Toshihide Mizoguchi
- Institute for Oral Science, Matsumoto Dental University, Nagano, Japan.,Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| |
Collapse
|
35
|
Coutel X, Delattre J, Marchandise P, Falgayrac G, Béhal H, Kerckhofs G, Penel G, Olejnik C. Mandibular bone is protected against microarchitectural alterations and bone marrow adipose conversion in ovariectomized rats. Bone 2019; 127:343-352. [PMID: 31276849 DOI: 10.1016/j.bone.2019.06.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 06/02/2019] [Accepted: 06/29/2019] [Indexed: 02/05/2023]
Abstract
Osteoporosis is a disease that leads to a loss of bone mass and to alterations in the bone microarchitecture that occur in a site-specific manner; however it remains controversial in the jaw. The involvement of bone marrow adipose tissue (BMAT) in the bone metabolism has been suggested in several physiopathological contexts, such as in aging and osteoporosis. To test whether the BMAT content is related to mandibular bone loss, this study aimed to investigate the potential correlations between the trabecular bone microarchitecture on one hand and BMAT content and its spatial distribution in relation to bone surface on the other hand during aging and ovariectomy (OVX) during a long-term follow-up in a mature rat model. No age-related microarchitectural or BMAT changes were observed in the mandible. The OVX-induced bone loss was three-fold lower in the mandible than in the tibia and was observed only in the alveolar bone (not in the condyle). We also report a delayed increase in the mandibular BMAT content that remained 4-6-fold lower compared to tibia. This low BMAT content in the mandible was located at a distance from the trabecular bone surface (only 5% in contact with the bone surface versus 87% in the tibia). These findings highlight a specific mandibular response to OVX, in particular fewer microarchitectural alterations compared to that in the tibia. For the latter, the trabecular bone thickness and surface were correlated with the BMAT content. Oral functions may have a protective effect on the mandibular BMAT conversion in an OVX context.
Collapse
Affiliation(s)
- Xavier Coutel
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France.
| | - Jérôme Delattre
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France
| | - Pierre Marchandise
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France
| | - Guillaume Falgayrac
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France
| | - Hélène Béhal
- Univ. Lille, CHU Lille, EA 2694 - Santé publique: épidémiologie et qualité des soins, Unité de Méthodologie et Biostatistiques, F-59000 Lille, France
| | - Greet Kerckhofs
- Biomechanics Lab, Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium; Institute of Experimental and Clinical Research, UCLouvain, Woluwe, Belgium; Department Materials Engineering, KU Leuven, Leuven, Belgium; Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Guillaume Penel
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France
| | - Cécile Olejnik
- Univ. Lille, Univ. Littoral Côte d'Opale, CHU Lille, EA 4490 - PMOI, F-59000 Lille, France
| |
Collapse
|
36
|
Gerace D, Martiniello-Wilks R, Habib R, Simpson AM. Luciferase-based reporting of suicide gene activity in murine mesenchymal stem cells. PLoS One 2019; 14:e0220013. [PMID: 31318955 PMCID: PMC6638968 DOI: 10.1371/journal.pone.0220013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/05/2019] [Indexed: 12/24/2022] Open
Abstract
Due to their ease of isolation, gene modification and tumor-homing properties, mesenchymal stem cells (MSCs) are an attractive cellular vehicle for the delivery of toxic suicide genes to a variety of cancers in pre-clinical models. In addition, the incorporation of suicide genes in stem cell-derived cell replacement therapies improves their safety profile by permitting graft destruction in the event of unexpected tumorigeneses or unwanted differentiation. Due to the functional requirement of ATP for the Firefly luciferase gene Luc2 to produce light, luciferase-based reporting of cytotoxicity can be engineered into potential cell therapies. Consequently, we nucleofected mammalian expression plasmids containing both the Luc2 and the yeast fusion cytosine deaminase uracil phosphoribosyltransferase (CDUPRT) genes for expression in murine MSCs to assess luciferase as a reporter of suicide gene cytotoxicity, and MSC as vehicles of suicide gene therapy. In vitro bioluminescence imaging (BLI) showed that following the addition of the non-toxic prodrug fluorocytosine (5-FC), CDUPRT-expressing MSCs displayed enhanced cytotoxicity in comparison to Luc2 reporter MSC controls. This study demonstrates the utility of luciferase as a reporter of CDUPRT-mediated cytotoxicity in murine MSC using BLI.
Collapse
Affiliation(s)
- Dario Gerace
- The School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, Sydney, Australia
| | - Rosetta Martiniello-Wilks
- The School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, Sydney, Australia
- Translational Cancer Research Group, University of Technology Sydney, Sydney, Australia
| | - Rosaline Habib
- The School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, Sydney, Australia
| | - Ann Margaret Simpson
- The School of Life Sciences and the Centre for Health Technologies, University of Technology Sydney, Sydney, Australia
- * E-mail:
| |
Collapse
|
37
|
Pineault KM, Song JY, Kozloff KM, Lucas D, Wellik DM. Hox11 expressing regional skeletal stem cells are progenitors for osteoblasts, chondrocytes and adipocytes throughout life. Nat Commun 2019; 10:3168. [PMID: 31320650 PMCID: PMC6639390 DOI: 10.1038/s41467-019-11100-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 06/18/2019] [Indexed: 12/31/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are required for skeletal formation, maintenance, and repair throughout life; however, current models posit that postnatally arising long-lived adult MSCs replace transient embryonic progenitor populations. We previously reported exclusive expression and function of the embryonic patterning transcription factor, Hoxa11, in adult skeletal progenitor-enriched MSCs. Here, using a newly generated Hoxa11-CreERT2 lineage-tracing system, we show Hoxa11-lineage marked cells give rise to all skeletal lineages throughout the life of the animal and persist as MSCs. Hoxa11 lineage-positive cells give rise to previously described progenitor-enriched MSC populations marked by LepR-Cre and Osx-CreER, placing them upstream of these populations. Our studies establish that Hox-expressing cells are skeletal stem cells that arise from the earliest stages of skeletal development and self-renew throughout the life of the animal.
Collapse
Affiliation(s)
- Kyriel M Pineault
- Department of Cell & Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jane Y Song
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109-2200, USA
| | - Kenneth M Kozloff
- Department of Orthopedic Surgery, University of Michigan, Ann Arbor, MI, 48109-2200, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Research, Cincinnati Children's Medical Center, Cincinnati, OH, 45229-2842, USA
| | - Deneen M Wellik
- Department of Cell & Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
38
|
Liu G, Xie Y, Su J, Qin H, Wu H, Li K, Yu B, Zhang X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone. FASEB J 2019; 33:11137-11147. [PMID: 31298955 DOI: 10.1096/fj.201900436rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
So far, there has been no effective cure for osteoporotic cortical bone, the most significant change in long bone structure during aging and the main cause of bone fragility fractures, because its underlying molecular and cellular mechanisms remain largely unknown. We used 3- and 15-mo-old mice as well as 15-mo-old mice treated with vehicle and gefitinib to evaluate structural, cellular, and molecular changes in cortical bone. We found that the senescence of osteoprogenitors was increased, whereas the expression of phosphorylated epidermal growth factor receptor (EGFR) on the endosteal surface of cortical bone down-regulated in middle-aged 15-mo-old mice compared with young 3-mo-old mice. Further decreasing EGFR signaling by gefitinib treatment in middle-aged mice resulted in promoted senescence of osteoprogenitors and accelerated cortical bone degeneration. Moreover, inhibiting EGFR signaling suppressed the expression of enhancer of zeste homolog 2 (Ezh2), the repressor of cell senescence-inducer genes, through ERK1/2 pathway, thereby promoting senescence in osteoprogenitors. Down-regulated EGFR signaling plays a physiologically significant role during aging by reducing Ezh2 expression, leading to the senescence of osteoprogenitors and the decline in bone formation on the endosteal surface of cortical bone.-Liu, G., Xie, Y., Su, J., Qin, H., Wu, H., Li, K., Yu, B., Zhang, X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone.
Collapse
Affiliation(s)
- Guanqiao Liu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongheng Xie
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianwen Su
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanjun Qin
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hangtian Wu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kaiqun Li
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yu
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianrong Zhang
- Department of Orthopaedics and Traumatology Nanfang Hospital, Southern Medical University, Guangzhou, China.,Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
High-Efficiency Lentiviral Gene Modification of Primary Murine Bone-Marrow Mesenchymal Stem Cells. Methods Mol Biol 2019. [PMID: 31273744 DOI: 10.1007/978-1-4939-9631-5_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Lentiviral vectors are the method of choice for stable gene modification of a variety of cell types. However, the efficiency with which they transduce target cells varies significantly, in particular their typically poor capacity to transduce primary stem cells. Here we describe the isolation and enrichment of murine bone-marrow mesenchymal stem cells (MSCs) via fluorescence-activated cell sorting (FACS); the cloning, production, and concentration of high-titer second generation lentiviral vectors via combined tangential flow filtration (TFF) and ultracentrifugation; and the subsequent high-efficiency gene modification of MSCs into insulin-producing cells via overexpression of the furin-cleavable human insulin (INS-FUR) gene.
Collapse
|
40
|
Mesenchymal Progenitors Derived from Different Locations in Long Bones Display Diverse Characteristics. Stem Cells Int 2019; 2019:5037578. [PMID: 31089329 PMCID: PMC6476036 DOI: 10.1155/2019/5037578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 11/20/2022] Open
Abstract
Mesenchymal progenitors within bone marrow have multiple differentiation potential and play an essential role in the maintenance of adult skeleton homeostasis. Mesenchymal progenitors located in bone regions other than the bone marrow also display bone-forming properties. However, owing to the differences in each distinct microenvironment, the mesenchymal characteristics of skeletal progenitor cells within different regions of long bones may show some differences. In order to clearly elucidate these differences, we performed a comparative study on mesenchymal progenitors from different regions of long bones. Here, we isolated mesenchymal progenitors from the periosteum, endosteum, and bone marrow of rat long bones. The three groups exhibited similar cellular morphologies and expressed the typical surface markers associated with mesenchymal stem cells. Interestingly, after cell proliferation assays and bidirectional differentiation analysis, periosteal mesenchymal progenitors showed a higher proliferative ability and adipogenic differentiation potential. In contrast, endosteal mesenchymal progenitors were more prone to osteogenic differentiation. Using in vitro osteoclast culture systems, conditioned media from different mesenchymal progenitor cultures were used to induce osteoclastic differentiation. Osteoclast formation was found to be significantly promoted by the secretion of RANKL and IL-6 by endosteal progenitors. Overall, our results provide strong evidence for the importance of selecting the appropriate source of skeletal progenitors for applications in future skeleton regeneration therapies.
Collapse
|
41
|
Bernhardsson M, Dietrich-Zagonel F, Tätting L, Eliasson P, Aspenberg P. Depletion of cytotoxic (CD8+) T cells impairs implant fixation in rat cancellous bone. J Orthop Res 2019; 37:805-811. [PMID: 30737834 DOI: 10.1002/jor.24246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/29/2019] [Indexed: 02/04/2023]
Abstract
As cytotoxic (CD8+ ) T cells seem to impair shaft fracture healing, we hypothesized that depletion of CD8+ cells would instead improve healing of cancellous bone. Additionally, we also tested if CD8-depletion would influence the healing of ruptured Achilles tendons. Rats received a single injection of either anti-CD8 antibodies or saline and put through surgery 24 h later. Three different surgical interventions were performed as follows: (1) a drill hole in the proximal tibia with microCT (BV/TV) to assess bone formation; (2) a screw in the proximal tibia with mechanical evaluation (pull-out force) to assess fracture healing; (3) Achilles tendon transection with mechanical evaluation (force-at-failure) to assess tendon healing. Furthermore, CD8-depletion was confirmed with flow cytometry on peripheral blood. Flow cytometric analysis confirmed depletion of CD8+ cells (p < 0.001). Contrary to our hypothesis, depletion of CD8+ cells reduced the implant pull-out force by 19% (p < 0.05) and stiffness by 34% (p < 0.01), although the bone formation in the drill holes was the same as in the controls. Tendon healing was unaffected by CD8-depletion. Our results suggest that CD8+ cells have an important part in cancellous bone healing. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Magnus Bernhardsson
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Franciele Dietrich-Zagonel
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Love Tätting
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Pernilla Eliasson
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Per Aspenberg
- Orthopaedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
42
|
Yao Z, Chen P, Wang S, Deng G, Hu Y, Lin Q, Zhang X, Yu B. Reduced PDGF-AA in subchondral bone leads to articular cartilage degeneration after strenuous running. J Cell Physiol 2019; 234:17946-17958. [PMID: 30834523 DOI: 10.1002/jcp.28427] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/10/2019] [Accepted: 02/14/2019] [Indexed: 12/24/2022]
Abstract
To identify the effects of running on articular cartilage and subchondral bone remodeling, C57BL/6 mice were randomly divided into three groups: control, moderate-, and strenuous running. Magnetic resonance imaging showed bone marrow lesions in the knee subchondral bone in the strenuous-running group in contrast with the other two groups. The microcomputed tomography analysis showed promoted bone formation in the subchondral bone in mice subjected to strenuous running. Histological and immunohistochemistry results indicated that terminal differentiation of chondrocytes and degeneration of articular cartilage were enhanced but, synthesis of platelet-derived growth factor-AA (PDGF-AA) in the subchondral bone was suppressed after strenuous running. In vitro, excessive mechanical treatments suppressed the expression of PDGF-AA in osteoblasts, and the condition medium from mechanical-treated osteoblasts stimulated maturation and terminal differentiation of chondrocytes. These results indicate that strenuous running suppresses the synthesis of PDGF-AA in subchondral bone, leading to downregulated PDGF/Akt signal in articular cartilage and thus cartilage degeneration.
Collapse
Affiliation(s)
- Zilong Yao
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Peisheng Chen
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Orthopedics, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, Fujian, China
| | - Shengnan Wang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ganming Deng
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Department of Bone and Joint Surgery, Southern Medical University Affiliated Shenzhen Baoan Hospital, Shenzhen, China
| | - Yanjun Hu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qingrong Lin
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xianrong Zhang
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Bin Yu
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.,Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
43
|
Tätting L, Sandberg O, Bernhardsson M, Ernerudh J, Aspenberg P. Different composition of leucocytes in cortical and cancellous bone healing in a mouse model. Bone Joint Res 2019; 7:620-628. [PMID: 30662708 PMCID: PMC6318753 DOI: 10.1302/2046-3758.712.bjr-2017-0366.r2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objectives Cortical and cancellous bone healing processes appear to be histologically different. They also respond differently to anti-inflammatory agents. We investigated whether the leucocyte composition on days 3 and 5 after cortical and cancellous injuries to bone was different, and compared changes over time using day 3 as the baseline. Methods Ten-week-old male C56/Bl6J mice were randomized to either cancellous injury in the proximal tibia or cortical injury in the femoral diaphysis. Regenerating tissues were analyzed with flow cytometry at days 3 and 5, using panels with 15 antibodies for common macrophage and lymphocyte markers. The cellular response from day 3 to 5 was compared in order to identify differences in how cancellous and cortical bone healing develop. Results Between day 3 and 5, the granulocytes increased in the cancellous model, whereas the lymphocytes (T cells, B cells, NK cells) and monocytes (CD11b+, F4/80+, CD206+, CD14+) increased in the cortical model. Conclusion These results suggest an acute type of inflammation in cancellous bone healing, and a more chronic inflammation in cortical healing. This might explain, in part, why cancellous healing is faster and more resistant to anti-inflammatory drugs than are diaphyseal fractures. Cite this article: L. Tätting, O. Sandberg, M. Bernhardsson, J. Ernerudh, P. Aspenberg. Different composition of leucocytes in cortical and cancellous bone healing in a mouse model. Bone Joint Res 2018;7:620–628. DOI: 10.1302/2046-3758.712.BJR-2017-0366.R2.
Collapse
Affiliation(s)
- L Tätting
- Department of Clinical and Experimental Medicine, Orthopaedics, Linköping University, Linköping, Sweden
| | - O Sandberg
- Principal Research Engineer, Department of Clinical and Experimental Medicine, Orthopaedics, Linköping University, Linköping, Sweden
| | - M Bernhardsson
- Department of Clinical and Experimental Medicine, Orthopaedics, Linköping University, Linköping, Sweden
| | - J Ernerudh
- Department of Clinical and Experimental Medicine and Department of Clinical Immunology and Transfusion Medicine, Linköping University, Linköping, Sweden
| | | |
Collapse
|
44
|
Wang X, Chu W, Zhuang Y, Shi D, Tao H, Jin C, Dai K, Zhao J, Gan Y. Bone Mesenchymal Stem Cell-Enriched β-Tricalcium Phosphate Scaffold Processed by the Screen-Enrich-Combine Circulating System Promotes Regeneration of Diaphyseal Bone Non-Union. Cell Transplant 2018; 28:212-223. [PMID: 30554525 PMCID: PMC6362520 DOI: 10.1177/0963689718818096] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bone non-union after fracture, considered a therapeutic challenge for orthopedics, always needs a reversion surgery, including autograft transplantation (AGT). However, adverse events related to autograft harvest cannot be ignored. Our group designed a novel system called the bone marrow stem cell Screen-Enrich-Combine Circulating System (SECCS) by seeding mesenchymal stem cells (MSCs) into β-tricalcium phosphate (β-TCP) during surgery to thereafter rapidly process bioactive bone implantation. In this retrospective case-control study, 30 non-union patients who accepted SECCS therapy and 20 non-union patients who accepted AGT were enrolled. By SECCS therapy, the MSC-enriched β-TCP particles were implanted into the non-union gap. During the enrichment procedure, a significant proportion of MSCs were screened and enriched from bone marrow into porous β-TCP particles, and the cells possessed the capacity for three-line differentiation and were CD90+/CD105+/CD34-/CD45-. Approximately 82.0±10.7% of MSCs were enriched from 60 mL bone marrow without damaging cell viability, and approximately 11,444.0±6,018 MSCs were transplanted per patient. No implant-related infections occurred in any case. After 9 months of follow-up, 27 patients (90%) in the SECCS group acquired clinical union, compared with 18 patients (90%) in the AGT group (clinical union time, P = 0.064), and postoperative radiographic union score at 9 months post-operation was similar between the two groups. In conclusion, the SECCS could concentrate a large proportion of MSCs from bone marrow to acquire enough effective cells for therapy without in vitro cell culture. Bone substitutes processed by SECCS demonstrated encouraging promotion of bone regeneration and showed a satisfactory clinical curative effect for diaphyseal bone non-union, which was non-inferior to AGT.
Collapse
Affiliation(s)
- Xin Wang
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China.,Both these authors contributed equally to this work as co-first authors
| | - WenXiang Chu
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China.,Both these authors contributed equally to this work as co-first authors
| | - YiFu Zhuang
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - DingWei Shi
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - HaiRong Tao
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Chen Jin
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - KeRong Dai
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Jie Zhao
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - YaoKai Gan
- 1 Shanghai Key Laboratory of Orthopaedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shang Hai, P.R. China.,2 Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
45
|
Liu J, Li X, Zhang D, Jiao J, Wu L, Hao F, Qin YX. Acceleration of Bone Defect Healing and Regeneration by Low-Intensity Ultrasound Radiation Force in a Rat Tibial Model. ULTRASOUND IN MEDICINE & BIOLOGY 2018; 44:2646-2654. [PMID: 30286949 DOI: 10.1016/j.ultrasmedbio.2018.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/20/2018] [Accepted: 08/02/2018] [Indexed: 06/08/2023]
Abstract
The objective was to evaluate the effect of low-intensity pulsed ultrasound (LIPUS)-induced acoustic radiation force on trabecular bone defect repair and healing in a rat tibial model. A uniform surgical defect, 3.5 mm in diameter, was generated in the proximal bilateral tibial region of rats (N = 20). LIPUS was applied to the defects in the left tibia for 20 min every day for 2 wk. Contralateral defects in the right tibia served as a control without active LIPUS treatment. The micro-computed tomography data revealed that LIPUS-treated tibia exhibited higher bone volume/total volume, connectivity density, trabecular number, and bone mineral density and significantly lower trabecular separation. Histomorphometry analysis indicated a similar trend. Mechanical testing data revealed that LIPUS treatment significantly increased bone stiffness relative to that of the control group. Short-term (2-wk) LIPUS therapy initiated trabecular bone repair and regeneration in large trabecular bone defects, whereas cortical bone remained in the initial non-mineralization stage.
Collapse
Affiliation(s)
- Jingbo Liu
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA; School of Stomatology, China Medical University, Shenyang, China
| | - Xiaofei Li
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Dongye Zhang
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Jian Jiao
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Lin Wu
- School of Stomatology, China Medical University, Shenyang, China
| | - Fengyu Hao
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA; School of Stomatology, China Medical University, Shenyang, China
| | - Yi-Xian Qin
- Orthopaedic Bioengineering Research Laboratory, Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
46
|
Li CJ, Xiao Y, Yang M, Su T, Sun X, Guo Q, Huang Y, Luo XH. Long noncoding RNA Bmncr regulates mesenchymal stem cell fate during skeletal aging. J Clin Invest 2018; 128:5251-5266. [PMID: 30352426 DOI: 10.1172/jci99044] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 09/11/2018] [Indexed: 12/22/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) exhibit an age-related lineage switch between osteogenic and adipogenic fates, which contributes to bone loss and adiposity. Here we identified a long noncoding RNA, Bmncr, which regulated the fate of BMSCs during aging. Mice depleted of Bmncr (Bmncr-KO) showed decreased bone mass and increased bone marrow adiposity, whereas transgenic overexpression of Bmncr (Bmncr-Tg) alleviated bone loss and bone marrow fat accumulation. Bmncr regulated the osteogenic niche of BMSCs by maintaining extracellular matrix protein fibromodulin (FMOD) and activation of the BMP2 pathway. Bmncr affected local 3D chromatin structure and transcription of Fmod. The absence of Fmod modified the bone phenotype of Bmncr-Tg mice. Further analysis revealed that Bmncr would serve as a scaffold to facilitate the interaction of TAZ and ABL, and thus facilitate the assembly of the TAZ and RUNX2/PPARG transcriptional complex, promoting osteogenesis and inhibiting adipogenesis. Adeno-associated viral-mediated overexpression of Taz in osteoprogenitors alleviated bone loss and marrow fat accumulation in Bmncr-KO mice. Furthermore, restoring BMNCR levels in human BMSCs reversed the age-related switch between osteoblast and adipocyte differentiation. Our findings indicate that Bmncr is a key regulator of the age-related osteogenic niche alteration and cell fate switch of BMSCs.
Collapse
Affiliation(s)
- Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan, China.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xi Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan, China
| |
Collapse
|
47
|
Abstract
Bones provide both skeletal scaffolding and space for hematopoiesis in its marrow. Previous work has shown that these functions were tightly regulated by the nervous system. The central and peripheral nervous systems tightly regulate compact bone remodeling, its metabolism, and hematopoietic homeostasis in the bone marrow (BM). Accumulating evidence indicates that the nervous system, which fine-tunes inflammatory responses and alterations in neural functions, may regulate autoimmune diseases. Neural signals also influence the progression of hematological malignancies such as acute and chronic myeloid leukemias. Here, we review the interplay of the nervous system with bone, BM, and immunity, and discuss future challenges to target hematological diseases through modulation of activity of the nervous system.
Collapse
Affiliation(s)
- Maria Maryanovich
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Shoichiro Takeishi
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
48
|
Bernhardsson M, Aspenberg P. Osteoblast precursors and inflammatory cells arrive simultaneously to sites of a trabecular-bone injury. Acta Orthop 2018; 89:457-461. [PMID: 29865916 PMCID: PMC6066776 DOI: 10.1080/17453674.2018.1481682] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background and purpose - Fracture healing in the shaft is usually described as a sequence of events, starting with inflammation, which triggers mesenchymal tissue formation in successive steps. Most clinical fractures engage cancellous bone. We here describe fracture healing in cancellous bone, focusing on the timing of inflammatory and mesenchymal cell type appearance at the site of injury Material and methods - Rats received a proximal tibial drill hole. A subgroup received clodronate-containing liposomes before or after surgery. The tibiae were analyzed with micro-CT and immunohistochemistry 1 to 7 days after injury. Results - Granulocytes (myeloperoxidase) appeared in moderate numbers within the hole at day 1 and then gradually disappeared. Macrophage expression (CD68) was seen on day 1, increased until day 3, and then decreased. Mesenchymal cells (vimentin) had already accumulated in the periphery of the hole on day 1. Mesenchymal cells dominated in the entire lesion on day 3, now producing extracellular matrix. A modest number of preosteoblasts (RUNX2) were seen on day 1 and peaked on day 4. Osteoid was seen on day 4 in the traumatized region, with a distinct border to the uninjured surrounding marrow. Clodronate liposomes given before the injury reduced the volume of bone formation at day 7, but no reduction in macrophage numbers could be detected. Interpretation - The typical sequence of events in shaft fractures was not seen. Mesenchymal cells appeared simultaneously with granulocyte and macrophage arrival. Clodronate liposomes, known to reduce macrophage numbers, seemed to be associated with the delineation of the volume of tissue to be replaced by bone. Most fracture healing studies in animal models concern cortical bone in shafts. However, most fractures in patients occur in cancellous bone in the metaphysis, such as the distal radius or in the vertebrae. A growing body of evidence suggests that there are important differences between the healing processes in cortical and cancellous bone.
Collapse
Affiliation(s)
- Magnus Bernhardsson
- Orthopedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.,Correspondence:
| | - Per Aspenberg
- Orthopedics, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
49
|
Abstract
Purpose of Review Functional decline of hematopoiesis that occurs in the elderly, or in patients who receive therapies that trigger cellular senescence effects, results in a progressive reduction in the immune response and an increased incidence of myeloid malignancy. Intracellular signals in hematopoietic stem cells and progenitors (HSC/P) mediate systemic, microenvironment, and cell-intrinsic effector aging signals that induce their decline. This review intends to summarize and critically review our advances in the understanding of the intracellular signaling pathways responsible for HSC decline during aging and opportunities for intervention. Recent Findings For a long time, aging of HSC has been thought to be an irreversible process imprinted in stem cells due to the cell intrinsic nature of aging. However, recent murine models and human correlative studies provide evidence that aging is associated with molecular signaling pathways, including oxidative stress, metabolic dysfunction, loss of polarity and an altered epigenome. These signaling pathways provide potential targets for prevention or reversal of age-related changes. Summary Here we review our current understanding of the signalling pathways that are differentially activated or repressed during HSC/P aging, focusing on the oxidative, metabolic, biochemical and structural consequences downstream, and cell-intrinsic, systemic, and environmental influences.
Collapse
|
50
|
Abstract
A substantial proportion of fractures can present with nonunion, and the management of nonunion continues to present a challenge for orthopaedic surgeons. A variety of biological, mechanical, patient, and injury factors can contribute to the occurrence of nonunion, and often the cause of nonunion may be multifactorial. Successful management often requires assessment and treatment of more than one of these factors. This article reviews common factors that may contribute to nonunion including infection, impaired biology, and metabolic disorders. In addition, new and evolving strategies for diagnosing the cause and effectively treating nonunion including the diagnosis of infection, metabolic workup, bone grafting, cell-based therapies, and biological adjuvants are reviewed and discussed.
Collapse
|