1
|
Tachibana S, Hayashi S, Ikuta K, Anjiki K, Onoi Y, Suda Y, Wada K, Maeda T, Saito A, Tsubosaka M, Kamenaga T, Kuroda Y, Nakano N, Matsumoto T, Hosooka T, Ogawa W, Kuroda R. Downregulation of Krüppel-like factor 15 expression delays endochondral bone ossification during fracture healing. Bone 2024; 190:117302. [PMID: 39437873 DOI: 10.1016/j.bone.2024.117302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE The role of Krüppel-like zinc finger transcription factor 15 (KLF15) in endochondral ossification during fracture healing remains unexplored. In this study, we aimed to elucidate the impact of KLF15 in a mouse model of tibial transverse fracture. METHODS We created tamoxifen-inducible, cartilage-specific KLF15 knockout mice (KLF15 KO). KLF15 fl/fl Col2-CreERT mice from the same litters as the KLF15 KO mice, but not treated with 4-hydroxytamoxifen, were used as controls (CT). At 10 weeks, male KLF15 KO and CT mice underwent tibial fracture followed by intramedullary nailing. Both groups were administered tamoxifen at days 0, 3, and 7 after surgery. The tibiae were harvested on post-surgery days 7, 10, and 14 for radiological assessment using micro-computed tomography. Histological staining (Safranin-O) and immunohistochemistry for KLF15, SOX9, Indian hedgehog (IHH), RUNX2, and Osterix were performed. Additionally, cartilage from mouse fetus was cultured for qRT-PCR and western blot analyses of KLF15, SOX9, IHH, Col2, RUNX2, Osterix, TGF-β, SMAD3, and phosphor-SMAD3. RESULTS The radiological assessment revealed that immature callus formation was delayed in KLF15 KO, compared with that in CT, peaking on day 14 compared with that on day 10 in CT. KLF15 KO mice exhibited delayed fracture healing and reduced Safranin-O staining at days 7 and 10 post-surgery. The ratio of cells positive for KLF15 and SOX9 was significantly lower in KLF15 KO than in CT, whereas the ratios for IHH, RUNX2, and Osterix showed no significant difference. RT-PCR revealed reduced expression of KLF15, SOX9, and COL2, with no significant changes in IHH, Osterix, RUNX2, TGF-β, and SMAD3. Western blot analysis indicated decreased SMAD3 phosphorylation in KLF15 KO mice. CONCLUSION KLF15 regulates SOX9 via the TGF-β-SMAD3-SOX9 pathway, independent of IHH, in endochondral ossification. The KLF15 deficiency decreases SOX9 expression through reduced SMAD3 phosphorylation, subsequently delaying fracture healing.
Collapse
Affiliation(s)
- Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Kemmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuma Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Saito
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tetsuya Hosooka
- Laboratory of Nutritional Physiology, Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
2
|
Xiang Q, Li L, Ji W, Gawlitta D, Walboomers XF, van den Beucken JJJP. Beyond resorption: osteoclasts as drivers of bone formation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:22. [PMID: 39392536 PMCID: PMC11469995 DOI: 10.1186/s13619-024-00205-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Emerging evidence illustrates that osteoclasts (OCs) play diverse roles beyond bone resorption, contributing significantly to bone formation and regeneration. Despite this, OCs remain mysterious cells, with aspects of their lifespan-from origin, fusion, alterations in cellular characteristics, to functions-remaining incompletely understood. Recent studies have identified that embryonic osteoclastogenesis is primarily driven by osteoclast precursors (OCPs) derived from erythromyeloid progenitors (EMPs). These precursor cells subsequently fuse into OCs essential for normal bone development and repair. Postnatally, hematopoietic stem cells (HSCs) become the primary source of OCs, gradually replacing EMP-derived OCs and assuming functional roles in adulthood. The absence of OCs during bone development results in bone structure malformation, including abnormal bone marrow cavity formation and shorter long bones. Additionally, OCs are reported to have intimate interactions with blood vessels, influencing bone formation and repair through angiogenesis regulation. Upon biomaterial implantation, activation of the innate immune system ensues immediately. OCs, originating from macrophages, closely interact with the immune system. Furthermore, evidence from material-induced bone formation events suggests that OCs are pivotal in these de novo bone formation processes. Nevertheless, achieving a pure OC culture remains challenging, and interpreting OC functions in vivo faces difficulties due to the presence of other multinucleated cells around bone-forming biomaterials. We here describe the fusion characteristics of OCPs and summarize reliable markers and morphological changes in OCs during their fusion process, providing guidance for researchers in identifying OCs both in vitro and in vivo. This review focuses on OC formation, characterization, and the roles of OCs beyond resorption in various bone pathophysiological processes. Finally, therapeutic strategies targeting OCs are discussed.
Collapse
Affiliation(s)
- Qianfeng Xiang
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
| | - Lei Li
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, GA, 3508, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, CT, 3584, The Netherlands
| | - X Frank Walboomers
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- Research Institute for Medical Innovation, Radboudumc, Nijmegen, the Netherlands
| | - Jeroen J J P van den Beucken
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands.
| |
Collapse
|
3
|
Xu H, Yan S, Gerhard E, Xie D, Liu X, Zhang B, Shi D, Ameer GA, Yang J. Citric Acid: A Nexus Between Cellular Mechanisms and Biomaterial Innovations. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2402871. [PMID: 38801111 PMCID: PMC11309907 DOI: 10.1002/adma.202402871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/07/2024] [Indexed: 05/29/2024]
Abstract
Citrate-based biodegradable polymers have emerged as a distinctive biomaterial platform with tremendous potential for diverse medical applications. By harnessing their versatile chemistry, these polymers exhibit a wide range of material and bioactive properties, enabling them to regulate cell metabolism and stem cell differentiation through energy metabolism, metabonegenesis, angiogenesis, and immunomodulation. Moreover, the recent US Food and Drug Administration (FDA) clearance of the biodegradable poly(octamethylene citrate) (POC)/hydroxyapatite-based orthopedic fixation devices represents a translational research milestone for biomaterial science. POC joins a short list of biodegradable synthetic polymers that have ever been authorized by the FDA for use in humans. The clinical success of POC has sparked enthusiasm and accelerated the development of next-generation citrate-based biomaterials. This review presents a comprehensive, forward-thinking discussion on the pivotal role of citrate chemistry and metabolism in various tissue regeneration and on the development of functional citrate-based metabotissugenic biomaterials for regenerative engineering applications.
Collapse
Affiliation(s)
- Hui Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Ethan Gerhard
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Denghui Xie
- Department of Histology and Embryology, School of Basic Medical Sciences, Department of Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, 510515, P. R. China
- Academy of Orthopedics of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, 510630, P. R. China
| | - Xiaodong Liu
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310030, P. R. China
- Westlake Institute for Advanced Study, Hangzhou, Zhejiang, 310030, P. R. China
| | - Bing Zhang
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310030, P. R. China
- Westlake Institute for Advanced Study, Hangzhou, Zhejiang, 310030, P. R. China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, Jiangsu, 210008, P. R. China
| | - Guillermo A Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jian Yang
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
- Biomedical Engineering Program, School of Engineering, Westlake University, Hangzhou, Zhejiang, 310030, P. R. China
| |
Collapse
|
4
|
Wu C, Yan J, Ge C, Xie L, He Y, Zhao Z, Deng Y, Dong Q, Yin L. Macrophage membrane-reversibly camouflaged nanotherapeutics accelerate fracture healing by fostering MSCs recruitment and osteogenic differentiation. J Nanobiotechnology 2024; 22:411. [PMID: 38997706 PMCID: PMC11241938 DOI: 10.1186/s12951-024-02679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/30/2024] [Indexed: 07/14/2024] Open
Abstract
The fracture healing outcome is largely dependent on the quantities as well as osteogenic differentiation capacities of mesenchymal stem cells (MSCs) at the lesion site. Herein, macrophage membrane (MM)-reversibly cloaked nanocomplexes (NCs) are engineered for the lesion-targeted and hierarchical co-delivery of short stromal derived factor-1α peptide (sSDF-1α) and Ckip-1 small interfering RNA (Ckip-1 siRNA, siCkip-1) to promote bone repair by concurrently fostering recruitment and osteogenic differentiation of endogenous MSCs. To construct the NCs, a membrane-penetrating α-helical polypeptide first assembles with siCkip-1, and the cationic NCs are sequentially coated with catalase and an outer shell of sSDF-1α-anchored MM. Due to MM-assisted inflammation homing, intravenously injected NCs could efficiently accumulate at the fractured femur, where catalase decomposes the local hydrogen peroxide to generate oxygen bubbles that drives the shedding of sSDF-1α-anchored MM in the extracellular compartment. The exposed, cationic inner core thus enables robust trans-membrane delivery into MSCs to induce Ckip-1 silencing. Consequently, sSDF-1α-guided MSCs recruitment cooperates with siCkip-1-mediated osteogenic differentiation to facilitate bone formation and accelerate bone fracture healing. This study provides an enlightened strategy for the hierarchical co-delivery of macromolecular drugs into different cellular compartments, and it also renders a promising modality for the management of fracture healing.
Collapse
Affiliation(s)
- Cheng Wu
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
- Department of Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, China
| | - Jing Yan
- Department of Gastroenterology, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Chenglong Ge
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Lucheng Xie
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yunjie He
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China
| | - Yekun Deng
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China
| | - Qirong Dong
- Department of Orthopedics, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China.
| | - Lichen Yin
- Department of Gastroenterology, The Second Affiliated Hospital, Soochow University, Suzhou, 215004, China.
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
5
|
Inoue S, Li C, Hatakeyama J, Jiang H, Kuroki H, Moriyama H. Higher-intensity ultrasound accelerates fracture healing via mechanosensitive ion channel Piezo1. Bone 2023; 177:116916. [PMID: 37777037 DOI: 10.1016/j.bone.2023.116916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/08/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Osteoporosis-related fractures are a major public health problem. Mechanobiological stimulation utilizing low-intensity pulsed ultrasound (LIPUS) is the most widely accepted modality for accelerating fracture healing. However, recent evidence has demonstrated the ineffectiveness of LIPUS, and the biophysical mechanisms of ultrasound-induced bone formation also remain elusive. Here, we demonstrate that ultrasound at a higher intensity than LIPUS effectively accelerates fracture healing in a mouse osteoporotic fracture model. Higher-intensity ultrasound promoted chondrogenesis and hypertrophic differentiation of chondrocytes in the fracture callus. Higher-intensity ultrasound also increased osteoblasts and newly formed bone in the callus, resulting in accelerated endochondral ossification during fracture healing. In addition, we found that accelerated fracture healing by ultrasound exposure was attenuated when the mechanosensitive ion channel Piezo1 was inhibited by GsMTx4. Ultrasound-induced new bone formation in the callus was attenuated in fractured mice treated with GsMTx4. Similar results were also confirmed in a 3D osteocyte-osteoblast co-culture system, where osteocytic Piezo1 knockdown attenuated the expression of osteoblastic genes after ultrasound exposure. Together these results demonstrate that higher-intensity ultrasound than clinically used LIPUS can accelerate endochondral ossification after fractures. Furthermore, our results suggest that mechanotransduction via Piezo1 mediates ultrasound-stimulated fracture healing and bone formation.
Collapse
Affiliation(s)
- Shota Inoue
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Changxin Li
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Junpei Hatakeyama
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan; Research Fellowship of the Japan Society for the Promotion of Science, Japan
| | - Hanlin Jiang
- Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Japan
| | - Hiroshi Kuroki
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Moriyama
- Life and Medical Sciences Area, Health Sciences Discipline, Kobe University, Kobe, Japan.
| |
Collapse
|
6
|
Dong WC, Guo JL, Jiang XH, Xu L, Wang H, Ni XY, Zhang YZ, Zhang ZQ, Jiang Y. A more accurate indicator to evaluate oxidative stress in rat plasma with osteoporosis. RSC Adv 2023; 13:1267-1277. [PMID: 36686958 PMCID: PMC9813688 DOI: 10.1039/d2ra05572d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/08/2022] [Indexed: 01/06/2023] Open
Abstract
Background: oxidative stress is linked to various human diseases which developed into the idea of "disrupted redox signaling". Osteoporosis (OP) is a chronic skeletal disorder characterized by low bone mineral density and deterioration of bone microarchitecture among which estrogen deficiency is the main cause. Lack of estrogen leads to the imbalance between oxidation and anti-oxidation in patients, and oxidative stress is an important link in the pathogenesis of OP. The ratio of the reduced to the oxidized thiols can characterize the redox status. However, few methods have been reported for the simultaneous determination of reduced forms and their oxidized forms of thiols in plasma. Methods: we developed a hollow fiber centrifugal ultrafiltration (HFCF-UF) method for sample preparation and validated a high-performance liquid chromatography tandem mass spectrometry (HPLC-MS/MS) method to determine two reduced forms of thiols-homocysteine (Hcy), cysteine (Cys) levels and their respective oxidized compounds, homocystine (HHcy) and cystine (Cyss) in rat plasma simultaneously for the first time. Thirty-six female rats were randomly divided into three groups: normal control (NC), oxidative stress (ovariectomy, OVX) and ovariectomy with hydrogen-rich saline administration (OVX + HRS). Results: the validation parameters for the methodological results were within the acceptance criteria. There were both significant differences of Hcy/HHcy (Hcy reduced/oxidized) and Cys/Cyss (Cys reduced/oxidized) in rat plasma between three groups with both p < 0.05 and meanwhile, the p values of malondialdehyde, superoxide dismutase and glutathione peroxidase were all less than 0.01. The value of both Hcy/HHcy and Cys/Cyss were significantly decreased with the change of Micro-CT scan result of femoral neck in OVX group (both the trabecular thickness and trabecular number significantly decreased with a significant increase of trabecular separation) which demonstrate OP occurs. The change of Hcy/HHcy is more obvious and prominent than Cys/Cyss. Conclusions: the Hcy/HHcy and Cys/Cyss could be suitable biomarkers for oxidative stress and especially Hcy/HHcy is more sensitive. The developed method is simple and accurate. It can be easily applied in clinical research to further evaluate the oxidative stress indicator for disease risk factors.
Collapse
Affiliation(s)
- Wei-Chong Dong
- Department of Pharmacy, The Second Hospital of Hebei Medical University215# Heping West RoadShijiazhuangHebei Province 050051China,Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University361# East Zhongshan RoadShijiazhuangHebei Province 050017China
| | - Jia-Liang Guo
- Department of Orthopaedics, The Third Hospital of Hebei Medical University139# Ziqiang RoadShijiazhuangHebei Province 050000China
| | - Xin-Hui Jiang
- Department of Obstetrics and Gynecology, Aerospace Central HospitalBeijing 100049China
| | - Lei Xu
- Department of Neurology, The Second Hospital of Hebei Medical UniversityShijiazhuangHebei Province 050051China
| | - Huan Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University361# East Zhongshan RoadShijiazhuangHebei Province 050017China
| | - Xiao-yu Ni
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University361# East Zhongshan RoadShijiazhuangHebei Province 050017China
| | - Ying-Ze Zhang
- Department of Orthopaedics, The Third Hospital of Hebei Medical University139# Ziqiang RoadShijiazhuangHebei Province 050000China
| | - Zhi-Qing Zhang
- Department of Pharmacy, The Second Hospital of Hebei Medical University215# Heping West RoadShijiazhuangHebei Province 050051China
| | - Ye Jiang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University361# East Zhongshan RoadShijiazhuangHebei Province 050017China
| |
Collapse
|
7
|
Kikuchi K, Haneda M, Hayashi S, Maeda T, Nakano N, Kuroda Y, Tsubosaka M, Kamenaga T, Fujita M, Ikuta K, Anjiki K, Tachibana S, Onoi Y, Matsumoto T, Kuroda R. P21 deficiency exhibits delayed endochondral ossification during fracture healing. Bone 2022; 165:116572. [PMID: 36180020 DOI: 10.1016/j.bone.2022.116572] [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: 05/21/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Endochondral ossification is a complex biological phenomenon involving a variety of factors and cells. Cyclin-dependent kinase inhibitor 1 (p21) inhibits cell cycle progression and is affected by external stress. We recently reported that embryonic endochondral ossification is unaffected by endogenous p21 deficiency. In this study, we evaluated whether p21 expression affects endochondral ossification during fracture healing. METHODS Tibial fractures were introduced into p21 knockout (p21-/-) (n = 24) and wild-type C57BL/6 (p21+/+) (n = 24) mice at age 10 weeks. Fracture healing was evaluated using radiological, histological, and immunohistochemical (IHC) analyses. The effect of p21 small interfering RNA (siRNA) on ATDC5 cells was assessed in vitro. RESULTS The Allen score for fracture healing was lower in p21-/- mice than in p21+/+ mice. In addition, p21-/- mice exhibited larger calluses and lower bone mineral density. IHC analyses showed that p21-/- mice exhibited delayed endochondral ossification via the Ihh-Runx2-Osterix pathway in vivo. Down-regulation of p21 expression in ATDC5 cells delayed endochondral ossification in vitro. CONCLUSIONS p21 deficiency leads to delayed endochondral ossification by attenuating the Ihh-Runx2-Osterix pathway in vivo, and p21 deficiency in hypertrophic chondrocytes causes delayed differentiation of hypertrophic chondrocytes in vitro. p21 plays a role in endochondral ossification during fracture healing.
Collapse
Affiliation(s)
- Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiko Haneda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Toshihisa Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
8
|
Kaune H, Montiel JF, Fenwick M, Williams SA. Rapid ovarian transcript changes during the onset of premature ovarian insufficiency. REPRODUCTION AND FERTILITY 2022; 3:RAF-22-0036. [PMID: 35972313 PMCID: PMC9513667 DOI: 10.1530/raf-22-0036] [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] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022] Open
Abstract
The manuscript has been submitted without altering abstract in line with Reproduction's Flexible Submission Process. The abstract is extended and thus does not fit this space.
Collapse
Affiliation(s)
- Heidy Kaune
- Laboratory of Reproduction, Centre for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, Santiago, Chile
- Program of Ethics and Public Policies in Human Reproduction, Universidad Diego Portales, Santiago, Chile
| | - Juan F Montiel
- Laboratory of Integrative Neuroscience, Centre for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, Santiago, Chile
| | - Mark Fenwick
- Academic Unit of Reproductive and Developmental Medicine, University of Sheffield, Sheffield, UK
| | - Suzannah A Williams
- Nuffield Department of Women’s and Reproductive Health, Women’s Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Zou N, Liu R, Li C. Cathepsin K+ Non-Osteoclast Cells in the Skeletal System: Function, Models, Identity, and Therapeutic Implications. Front Cell Dev Biol 2022; 10:818462. [PMID: 35912093 PMCID: PMC9326176 DOI: 10.3389/fcell.2022.818462] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Cathepsin K (Ctsk) is a cysteine protease of the papain superfamily initially identified in differentiated osteoclasts; it plays a critical role in degrading the bone matrix. However, subsequent in vivo and in vitro studies based on animal models elucidate novel subpopulations of Ctsk-expressing cells, which display markers and properties of mesenchymal stem/progenitor cells. This review introduces the function, identity, and role of Ctsk+ cells and their therapeutic implications in related preclinical osseous disorder models. It also summarizes the available in vivo models for studying Ctsk+ cells and their progeny. Further investigations of detailed properties and mechanisms of Ctsk+ cells in transgenic models are required to guide potential therapeutic targets in multiple diseases in the future.
Collapse
Affiliation(s)
- Nanyu Zou
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Ran Liu
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, The Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- *Correspondence: Changjun Li,
| |
Collapse
|
10
|
Lu J, Hu D, Ma C, Shuai B. Advances in Our Understanding of the Mechanism of Action of Drugs (including Traditional Chinese Medicines) for the Intervention and Treatment of Osteoporosis. Front Pharmacol 2022; 13:938447. [PMID: 35774616 PMCID: PMC9237325 DOI: 10.3389/fphar.2022.938447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis (OP) is known as a silent disease in which the loss of bone mass and bone density does not cause obvious symptoms, resulting in insufficient treatment and preventive measures. The losses of bone mass and bone density become more severe over time and an only small percentage of patients are diagnosed when OP-related fractures occur. The high disability and mortality rates of OP-related fractures cause great psychological and physical damage and impose a heavy economic burden on individuals and society. Therefore, early intervention and treatment must be emphasized to achieve the overall goal of reducing the fracture risk. Anti-OP drugs are currently divided into three classes: antiresorptive agents, anabolic agents, and drugs with other mechanisms. In this review, research progress related to common anti-OP drugs in these three classes as well as targeted therapies is summarized to help researchers and clinicians understand their mechanisms of action and to promote pharmacological research and novel drug development.
Collapse
|
11
|
Gao Y, Patil S, Jia J. The Development of Molecular Biology of Osteoporosis. Int J Mol Sci 2021; 22:8182. [PMID: 34360948 PMCID: PMC8347149 DOI: 10.3390/ijms22158182] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is one of the major bone disorders that affects both women and men, and causes bone deterioration and bone strength. Bone remodeling maintains bone mass and mineral homeostasis through the balanced action of osteoblasts and osteoclasts, which are responsible for bone formation and bone resorption, respectively. The imbalance in bone remodeling is known to be the main cause of osteoporosis. The imbalance can be the result of the action of various molecules produced by one bone cell that acts on other bone cells and influence cell activity. The understanding of the effect of these molecules on bone can help identify new targets and therapeutics to prevent and treat bone disorders. In this article, we have focused on molecules that are produced by osteoblasts, osteocytes, and osteoclasts and their mechanism of action on these cells. We have also summarized the different pharmacological osteoporosis treatments that target different molecular aspects of these bone cells to minimize osteoporosis.
Collapse
Affiliation(s)
- Yongguang Gao
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China;
| | - Suryaji Patil
- Lab for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China;
| | - Jingxian Jia
- Tangshan Key Laboratory of Green Speciality Chemicals, Department of Chemistry, Tangshan Normal University, Tangshan 063000, China;
| |
Collapse
|
12
|
Yahara Y, Ma X, Gracia L, Alman BA. Monocyte/Macrophage Lineage Cells From Fetal Erythromyeloid Progenitors Orchestrate Bone Remodeling and Repair. Front Cell Dev Biol 2021; 9:622035. [PMID: 33614650 PMCID: PMC7889961 DOI: 10.3389/fcell.2021.622035] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/12/2021] [Indexed: 12/21/2022] Open
Abstract
A third of the population sustains a bone fracture, and the pace of fracture healing slows with age. The slower pace of repair is responsible for the increased morbidity in older individuals who sustain a fracture. Bone healing progresses through overlapping phases, initiated by cells of the monocyte/macrophage lineage. The repair process ends with remodeling. This last phase is controlled by osteoclasts, which are bone-specific multinucleated cells also of the monocyte/macrophage lineage. The slower rate of healing in aging can be rejuvenated by macrophages from young animals, and secreted proteins from macrophage regulate undifferentiated mesenchymal cells to become bone-forming osteoblasts. Macrophages can derive from fetal erythromyeloid progenitors or from adult hematopoietic progenitors. Recent studies show that fetal erythromyeloid progenitors are responsible for the osteoclasts that form the space in bone for hematopoiesis and the fetal osteoclast precursors reside in the spleen postnatally, traveling through the blood to participate in fracture repair. Differences in secreted proteins between macrophages from old and young animals regulate the efficiency of osteoblast differentiation from undifferentiated mesenchymal precursor cells. Interestingly, during the remodeling phase osteoclasts can form from the fusion between monocyte/macrophage lineage cells from the fetal and postnatal precursor populations. Data from single cell RNA sequencing identifies specific markers for populations derived from the different precursor populations, a finding that can be used in future studies. Here, we review the diversity of macrophages and osteoclasts, and discuss recent finding about their developmental origin and functions, which provides novel insights into their roles in bone homeostasis and repair.
Collapse
Affiliation(s)
- Yasuhito Yahara
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Toyama, Toyama, Japan.,Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Xinyi Ma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Liam Gracia
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Benjamin A Alman
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States.,Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
13
|
Peres-Ueno MJ, Fernandes F, Brito VGB, Nicola ÂC, Stringhetta-Garcia CT, Castoldi RC, Menezes AP, Ciarlini PC, Louzada MJQ, Oliveira SHP, Ervolino E, Chaves-Neto AH, Dornelles RCM. Effect of pre-treatment of strength training and raloxifene in periestropause on bone healing. Bone 2020; 134:115285. [PMID: 32097761 DOI: 10.1016/j.bone.2020.115285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/31/2020] [Accepted: 02/14/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND There is evidence that strength training (ST) and raloxifene (Ral) treatment during periestropause promotes better bone quality. We wanted to determine whether the skeletal benefits of ST or Ral treatment, performed during periestropause, would persist after fracture. Therefore, the present study aimed to analyze the influence of pre-treatment with ST and administration of Ral during periestropause on bone healing after total unilateral osteotomy. METHODS Senescent female Wistar rats between 18 and 21 months of age, performed ST on a ladder three times per week, were administered Ral by gavage (2.3 mg/kg/day), or an association of both. After 120 days, the treatments were interrupted, and a total osteotomy was performed on the left tibia in all animals. They were euthanized 1 and 8 weeks post-osteotomy. RESULTS The administration of Ral during periestropause worsened the biochemical and oxidative profile, decreased gene expression of markers related to bone resorption and remodeling, which negatively affected the physicochemical properties; this lead to changes in the bone callus microarchitecture and mass, as well as a decrease in callus resistance to torsional deformation, resulting in lower tissue quality during bone healing. In contrast, ST performed prior to the osteotomy resulted in better bone healing, improvement of the biochemical and oxidative profile, alteration of the genetic profile in favor of bone formation and resorption, as well as the physic-ochemical properties of the callus. These changes led to better microarchitecture and bone mass and increased callus resistance to torsional deformation, confirming its beneficial effect on the quality of bone tissue, providing acceleration of bone consolidation. The combination of therapies at this exercise intensity and drug dosage showed a negative interaction, where the negative effect of Ral overcame the positive effect of ST, leading to decreased tissue quality in the bone healing process. CONCLUSIONS This study indicates that in addition to excellent non-pharmacological therapy and action in the prevention of osteoporosis, ST performed during the aging period may increase bone quality at the onset of healing and provide improved bone consolidation. Furthermore, the anti-osteoclastogenic effect of Ral shown in this model delayed the bone repair process, resulting in considerable clinical concern.
Collapse
Affiliation(s)
- Melise Jacon Peres-Ueno
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil.
| | - Fernanda Fernandes
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Victor Gustavo Balera Brito
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Ângela Cristina Nicola
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Camila Tami Stringhetta-Garcia
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Robson Chacon Castoldi
- Faculty of Science and Technology, São Paulo State University (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Amanda Pinatti Menezes
- Department of Support, Production and Animal Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Paulo Cézar Ciarlini
- Department of Support, Production and Animal Health, School of Veterinary Medicine, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Mário Jeferson Quirino Louzada
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Sandra Helena Penha Oliveira
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil; Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Edilson Ervolino
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Antonio Hernandes Chaves-Neto
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil; Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Rita Cassia Menegati Dornelles
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, SBFis, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil; Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil.
| |
Collapse
|
14
|
Paglia DN, Diaz-Hernandez ME, Roberts JL, Kalinowski J, Lorenzo J, Drissi H. Deletion of Runx1 in osteoclasts impairs murine fracture healing through progressive woven bone loss and delayed cartilage remodeling. J Orthop Res 2020; 38:1007-1015. [PMID: 31769548 DOI: 10.1002/jor.24537] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/24/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023]
Abstract
Conditional deletion of the transcription factor Runt-related transcription factor 1 (Runx1) in myeloid osteoclast precursors promotes osteoclastogenesis and subsequent bone loss. This study posits whether Runx1 regulates clastic cell-mediated bone and cartilage resorption in the fracture callus. We first generated mice, in which Runx1 was conditionally abrogated in osteoclast precursors (LysM-Cre;Runx1F/F ; Runx1 cKO). Runx1 cKO and control mice were then subjected to experimental mid-diaphyseal femoral fractures. Our study found differential resorption of bony and calcified cartilage callus matrix by osteoclasts and chondroclasts within Runx1 cKO calluses, with increased early bony callus resorption and delayed calcified cartilage resorption. There was an increased number of osteoclasts and chondroclasts in the chondro-osseous junction of Runx1 cKO calluses starting at day 11 post-fracture, with minimal woven bone occupying the callus at day 18 post-fracture. LysM-Cre;Runx1F/F mutant mice had increased bone compliance at day 28, but their strength and work to failure were comparable with controls. Taken together, these results indicate that Runx1 is a critical transcription factor in controlling osteoclastogenesis that negatively regulates bone and cartilage resorption in the fracture callus. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:1007-1015, 2020.
Collapse
Affiliation(s)
- David N Paglia
- Department of Orthopaedics, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | | | - Joseph L Roberts
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, Georgia
| | - Judy Kalinowski
- Department of Medicine and Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Joseph Lorenzo
- Department of Medicine and Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut
| | - Hicham Drissi
- Department of Orthopaedics, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
15
|
Zhang T, Han W, Zhao K, Yang W, Lu X, Jia Y, Qin A, Qian Y. Psoralen accelerates bone fracture healing by activating both osteoclasts and osteoblasts. FASEB J 2019; 33:5399-5410. [PMID: 30702934 DOI: 10.1096/fj.201801797r] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Bone fracture healing is a complex, dynamic process that involves various cell types, with osteoclasts and osteoblasts playing indispensable roles. In this study, we found that psoralen, the main active ingredient in Psoralea corylifolia L. fruit extract, enhanced bone fracture healing through activation of osteoclast and osteoblast activity via the ERK signaling pathway. In detail, psoralen promoted receptor activator of nuclear factor-κB ligand-induced osteoclastogenesis, mRNA expression of osteoclast-specific genes, and osteoclastic bone resorption in primary bone marrow-derived macrophages. Meanwhile, psoralen induced osteogenic differentiation by promoting the mRNA expression of the osteoblast differentiation markers alkaline phosphatase, runt-related transcription factor 2, osterix, and osteocalcin. At the molecular level, psoralen preferentially activated ERK1/2 but not JNK or p38 MAPKs. Further experiments revealed that psoralen-induced osteoclast and osteoblast differentiation was abrogated by a specific inhibitor of phosphorylated ERK. In addition, psoralen accelerated bone fracture healing in a rat tibial fracture model, and the numbers of osteoclasts and osteoblasts were increased in psoralen-treated fracture callus. Taken together, our findings indicate that psoralen accelerates bone fracture healing through activation of osteoclasts and osteoblasts via ERK signaling and has potential as a novel drug in the orthopedic clinic for the treatment of bone fractures.-Zhang, T., Han, W., Zhao, K., Yang, W., Lu, X., Jia, Y., Qin, A., Qian, Y. Psoralen accelerates bone fracture healing by activating both osteoclasts and osteoblasts.
Collapse
Affiliation(s)
- Tan Zhang
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Weiqi Han
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Kangxian Zhao
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Wanlei Yang
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Xuanyuan Lu
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Yewei Jia
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - An Qin
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implants, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Qian
- Department of Orthopaedics, Shaoxing People's Hospital, Zhejiang University School of Medicine, Shaoxing, China
| |
Collapse
|
16
|
Bahney CS, Zondervan RL, Allison P, Theologis A, Ashley JW, Ahn J, Miclau T, Marcucio RS, Hankenson KD. Cellular biology of fracture healing. J Orthop Res 2019; 37:35-50. [PMID: 30370699 PMCID: PMC6542569 DOI: 10.1002/jor.24170] [Citation(s) in RCA: 339] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
The biology of bone healing is a rapidly developing science. Advances in transgenic and gene-targeted mice have enabled tissue and cell-specific investigations of skeletal regeneration. As an example, only recently has it been recognized that chondrocytes convert to osteoblasts during healing bone, and only several years prior, seminal publications reported definitively that the primary tissues contributing bone forming cells during regeneration were the periosteum and endosteum. While genetically modified animals offer incredible insights into the temporal and spatial importance of various gene products, the complexity and rapidity of healing-coupled with the heterogeneity of animal models-renders studies of regenerative biology challenging. Herein, cells that play a key role in bone healing will be reviewed and extracellular mediators regulating their behavior discussed. We will focus on recent studies that explore novel roles of inflammation in bone healing, and the origins and fates of various cells in the fracture environment. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Chelsea S. Bahney
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Robert L. Zondervan
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Patrick Allison
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Alekos Theologis
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Jason W. Ashley
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Jaimo Ahn
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
17
|
Tian L, Sheng Y, Huang L, Chow DHK, Chau WH, Tang N, Ngai T, Wu C, Lu J, Qin L. An innovative Mg/Ti hybrid fixation system developed for fracture fixation and healing enhancement at load-bearing skeletal site. Biomaterials 2018; 180:173-183. [PMID: 30041069 DOI: 10.1016/j.biomaterials.2018.07.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 07/05/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
Magnesium (Mg) is a potential biomaterial suitable for developing biodegradable orthopaedic implants, especially as internal fixators for fracture fixation at non-load bearing skeletal sites. However, Mg alone cannot provide sufficient mechanical support for stable fracture fixation at load bearing sites due to its rapid degradation in the early stage after implantation. In consideration of the strengths and weaknesses of Mg, we developed an innovative magnesium/titanium (Mg/Ti) hybrid fixation system for long bone fracture fixation and investigated the fixation efficacy. The finite element analysis (FEA) results indicated that the Mg/Ti hybrid fixation system provided sufficient mechanical support for fracture fixation at load-bearing skeletal site. As a proof-of-concept, we performed a "Z-shaped" open osteotomy at the mid-shaft of rabbit tibia. For comparison, the animals were divided into two groups: Mg/Ti group (fixated with Mg screws and Ti fixators) and Ti control group (fixated with Ti screws and Ti fixators). The radiographic, four-point bending mechanical test, histological and histomorphometric analysis were postoperatively performed in a temporal manner up to 12 weeks. Both X-ray and micro-CT images of the Mg/Ti group showed a larger callus (14.7% at 3rd week and 24.8% at 6th week, n = 5-7, p < 0.05) in the regions of interest (ROIs) over time, especially at the opposite cortex of the fixation plate. At the 12th week post-operation, the biomechanical test result indicated that the rabbit tibia in the Mg/Ti group healed better and the overall mechanical strength was approximately 3-fold higher (n = 8, p < 0.05) than that at 6th week. Furthermore, the FEA revealed that the Mg/Ti group had a higher mechanical strength (19.5% at week 6 and 31.5% at week 12) at the specified ROI and resulted in an earlier and faster endochondral ossification (68.0% at week 3 and 71.4% at week 6) with a higher expression of osteocalcin (54.0%) and collagen I (34.2%) than the Ti control group (n = 4, p < 0.05). Further evaluation suggested that a higher expression of calcitonin gene-related peptide (CGRP), a known osteogenic neuron peptide, in the fracture callus of the Mg/Ti group might be a major underlying mechanism of enhanced fracture healing attributed to the release of Mg ions during the degradation of Mg screws.
Collapse
Affiliation(s)
- Li Tian
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yifeng Sheng
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Le Huang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Dick Ho-Kiu Chow
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Wing Ho Chau
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Ning Tang
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Wu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jian Lu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region
| | - Ling Qin
- Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
18
|
Soki FN, Yoshida R, Paglia DN, Duong LT, Hansen MF, Drissi H. Articular cartilage protection in Ctsk -/- mice is associated with cellular and molecular changes in subchondral bone and cartilage matrix. J Cell Physiol 2018; 233:8666-8676. [PMID: 29781506 DOI: 10.1002/jcp.26745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) is a degenerative disease and a major cause of chronic disability in aging individuals. Cathepsin K (CatK), encoded by the Ctsk gene, has been implicated in the pathogenesis of pycnodysostosis and osteoporosis. The use of a selective inhibitor of CatK was recently shown to delay OA progression in rabbits. However, the cellular mechanisms underlying these protective effects remain unexplored. We examined articular cartilage maintenance and joint bone remodeling using Ctsk null mice (Ctsk-/- ) which underwent destabilization of the medial meniscus (DMM). We found that Ctsk-/- mice displayed delayed remodeling of subchondral and calcified cartilage by osteoclasts and chodroclasts respectively in DMM-induced osteoarthritis. While WT mice displayed a more severe OA phenotype than Ctsk-/- mice at 16 weeks, higher subchondral bone volume and lower trabecular spacing were also observed in surgically-induced OA joints of Ctsk-/- mice. However, no differences were seen in non-surgical controls. During OA progression, TRAP+ osteoclast numbers were increased in both WT and Ctsk-/- mice. However, Ctsk-/- mice had fewer physis-derived chondroclasts than WT when OA was present. These data suggest that CatK may differentially regulate chondroclastogenesis in the growth plate. Targeted PCR arrays of RNA harvested from laser captured osteoclasts in the subchondral bone and chondroclasts in the growth plate demonstrated differential expression of Atp6v0d2, Tnfrsf11a, Ca2, Calcr, Ccr1, Gpr68, Itgb3, Nfatc1, and Syk genes between WT and Ctsk-/- mice at 8- and 16-weeks post-DMM. Our data provide insight into the cellular mechanisms by which cathepsin K deletion delays OA progression in mice.
Collapse
Affiliation(s)
- Fabiana N Soki
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Ryu Yoshida
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - David N Paglia
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, Connecticut
| | - Le T Duong
- Bone Biology, Merck & Co., Inc., West Point, Pennsylvania
| | - Marc F Hansen
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Hicham Drissi
- Department of Orthopaedics, Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
Walia B, Lingenheld E, Duong L, Sanjay A, Drissi H. A novel role for cathepsin K in periosteal osteoclast precursors during fracture repair. Ann N Y Acad Sci 2018; 1415:57-68. [PMID: 29479711 DOI: 10.1111/nyas.13629] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/04/2018] [Accepted: 01/11/2018] [Indexed: 11/29/2022]
Abstract
Osteoporosis management is currently centered around bisphosphonates, which inhibit osteoclast (OC) bone resorption but do not affect bone formation. This reduces fracture risk, but fails to restore healthy bone remodeling. Studies in animal models showed that cathepsin K (CatK) inhibition by genetic deletion or chemical inhibitors maintained bone formation while abrogating resorption during bone remodeling and stimulated periosteal bone modeling. Recently, periosteal mononuclear tartrate-resistant acid phosphatase-positive (TRAP+ ) osteoclast precursors (OCPs) were shown to augment angiogenesis-coupled osteogenesis. CatK gene deletion increased osteoblast differentiation via enhanced OCP and OC secretion of platelet-derived growth factor (PDGF)-BB and sphingosine 1 phosphate. The effects of periosteum-derived OCPs on bone remodeling are unknown, particularly with regard to fracture repair. We hypothesized that periosteal OCPs derived from CatK-null (Ctsk-/- ) mice may enhance periosteal bone formation during fracture repair. We found fewer periosteal OCPs in Ctsk-/- mice under homeostatic conditions; however, after fracture, this population increased in number relative to that seen in wild-type (WT) mice. Enhanced TRAP staining and greater expression of PDGF-BB were observed in fractured Ctsk-/- femurs relative to WT femurs. This early pattern of augmented PDGF-BB expression in Ctsk-/- mice may contribute to improved fracture healing by enhancing callus mineralization in Ctsk-/- mice.
Collapse
Affiliation(s)
- Bhavita Walia
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut
| | | | - Le Duong
- Bone Biology Group, Merck Research Laboratories, West Point, Pennsylvania
| | - Archana Sanjay
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
20
|
Mirza F, Lorenzo J, Drissi H, Lee FY, Soung DY. Dried plum alleviates symptoms of inflammatory arthritis in TNF transgenic mice. J Nutr Biochem 2017; 52:54-61. [PMID: 29149648 DOI: 10.1016/j.jnutbio.2017.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 07/24/2017] [Accepted: 10/14/2017] [Indexed: 12/20/2022]
Abstract
Dried plum (DP), a rich source of polyphenols has been shown to have bone-preserving properties in both animal models of osteoporosis and postmenopausal women. We evaluated if DP alleviated the destruction of joints in transgenic mice (TG) that overexpress human tumor necrosis factor (TNF), a genetic model of rheumatoid arthritis (RA). A four-week treatment of 20% DP diet in TG slowed the onset of arthritis and reduced bone erosions in the joints compared to TG on a regular diet. This was associated with fewer tartrate-resistant acid phosphatase (TRAP) positive cells, suggesting decreased osteoclastogenesis. A DP diet also produced significant protection of articular cartilage and reduction of synovitis. Cultures of human synovial fibroblast in the presence of TNF showed a significant increase in inflammatory interleukin (IL)-1β, chemokines (monocyte chemoattractant protein-1: MCP1 & macrophage inflammatory protein-1 alpha: MIP1α), cartilage matrix metalloproteinases (MMP1&3), and an osteoclastogenic cytokine (receptor activator of nuclear factor-κB ligand: RANKL) compared to controls. Addition of neochlorogenic acid (NC), a major polyphenol in DP to these cultures resulted in down-regulation of these genes. In the cultures of mouse bone marrow macrophage, NC also repressed TNF-induced formation of osteoclasts and mRNA levels of cathepsin K and MMP9 through inhibition of nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) expression and nuclear factor kappa B (NF-κB) activation. Our data suggested that dietary supplementation with DP inhibited TNF singling; leading to decreased erosions of bone and articular cartilage as well as synovitis.
Collapse
Affiliation(s)
- Faryal Mirza
- Department of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Joseph Lorenzo
- Department of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA; Orthopaedic Surgery, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Hicham Drissi
- Genetics and Genome Sciences, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA; Orthopaedic Surgery, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, 800 Howard Avenue, New Haven, CT 06519, USA
| | - Do Y Soung
- Department of Orthpaedic Surgery, Columbia University, 650 W. 168th Street, Black Building 14-1410, New York, NY 10032, USA.
| |
Collapse
|
21
|
Both T, van de Peppel HJ, Zillikens MC, Koedam M, van Leeuwen JPTM, van Hagen PM, van Daele PLA, van der Eerden BCJ. Hydroxychloroquine decreases human MSC-derived osteoblast differentiation and mineralization in vitro. J Cell Mol Med 2017; 22:873-882. [PMID: 28975700 PMCID: PMC5783866 DOI: 10.1111/jcmm.13373] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/09/2017] [Indexed: 01/03/2023] Open
Abstract
We recently showed that patients with primary Sjögren Syndrome (pSS) have significantly higher bone mineral density (BMD) compared to healthy controls. The majority of those patients (69%) was using hydroxychloroquine (HCQ), which may have favourable effects on BMD. To study the direct effects of HCQ on human MSC‐derived osteoblast activity. Osteoblasts were cultured from human mesenchymal stromal cells (hMSCs). Cultures were treated with different HCQ doses (control, 1 and 5 µg/ml). Alkaline phosphatase activity and calcium measurements were performed to evaluate osteoblast differentiation and activity, respectively. Detailed microarray analysis was performed in 5 µg/ml HCQ‐treated cells and controls followed by qPCR validation. Additional cultures were performed using the cholesterol synthesis inhibitor simvastatin (SIM) to evaluate a potential mechanism of action. We showed that HCQ inhibits both MSC‐derived osteoblast differentiation and mineralization in vitro. Microarray analysis and additional PCR validation revealed a highly significant up‐regulation of the cholesterol biosynthesis, lysosomal and extracellular matrix pathways in the 5 µg/ml HCQ‐treated cells compared to controls. Besides, we demonstrated that 1 µM SIM also decreases MSC‐derived osteoblast differentiation and mineralization compared to controls. It appears that the positive effect of HCQ on BMD cannot be explained by a stimulating effect on the MSC‐derived osteoblast. The discrepancy between high BMD and decreased MSC‐derived osteoblast function due to HCQ treatment might be caused by systemic factors that stimulate bone formation and/or local factors that reduce bone resorption, which is lacking in cell cultures.
Collapse
Affiliation(s)
- Tim Both
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Jeroen van de Peppel
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - M Carola Zillikens
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johannes P T M van Leeuwen
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Paul L A van Daele
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Lin H, O'Connor JP. Osteoclast depletion with clodronate liposomes delays fracture healing in mice. J Orthop Res 2017; 35:1699-1706. [PMID: 27653179 PMCID: PMC7582234 DOI: 10.1002/jor.23440] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 09/15/2016] [Indexed: 02/04/2023]
Abstract
Osteoclasts are abundant within the fracture callus and also localize at the chondro-osseous junction. However, osteoclast functions during fracture healing are not well defined. Inhibition of osteoclast formation or resorptive activity impairs callus remodeling but does not prevent callus formation. Interestingly, though anti-osteoclast therapies differentially affect resolution of callus cartilage into bone. Treatments that inhibit osteoclast formation or viability tend to impair callus cartilage resolution, while treatments that target inhibition of bone resorption generally do not affect callus cartilage resolution. Here, we tested whether depletion of osteoclasts by systemic treatment with clodronate liposomes would similarly impair callus cartilage resolution. ICR mice were treated by intraperitoneal injections of clodronate-laden liposomes or control liposomes and subjected to closed femur fracture. Femurs were resected at multiple times after fracture and analyzed by radiography, histology, and mechanical testing to determine effects on healing. Clodronate liposome treatment did not prevent callus formation. However, radiographic scoring indicated that clodronate liposome treatment impaired healing. Clodronate liposome treatment significantly reduced callus osteoclast populations and delayed resolution of callus cartilage. Consistent with continued presence of callus cartilage, torsional mechanical testing found significant decreases in callus material properties after 28 days of healing. The results support a role for osteoclasts in the resolution of callus cartilage into bone. Whether the cartilage resolution role for osteoclasts is limited to simply resorbing cartilage at the chondro-osseous junction or in promoting bone formation at the chondro-osseous junction through another mechanism, perhaps similar to the reversal process in bone remodeling, will require further experimentation. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1699-1706, 2017.
Collapse
Affiliation(s)
- Hsuan‐Ni Lin
- Department of OrthopaedicsGraduate School of Biomedical Sciences and New Jersey Medical School, RutgersThe State University of New JerseyNewark185 South Orange AvenueNew Jersey07103
| | - J. Patrick O'Connor
- Department of OrthopaedicsGraduate School of Biomedical Sciences and New Jersey Medical School, RutgersThe State University of New JerseyNewark185 South Orange AvenueNew Jersey07103
| |
Collapse
|
23
|
Whole grape alleviates inflammatory arthritis through inhibition of tumor necrosis factor. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
24
|
Drake MT, Clarke BL, Oursler MJ, Khosla S. Cathepsin K Inhibitors for Osteoporosis: Biology, Potential Clinical Utility, and Lessons Learned. Endocr Rev 2017; 38:325-350. [PMID: 28651365 PMCID: PMC5546879 DOI: 10.1210/er.2015-1114] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 06/20/2017] [Indexed: 12/24/2022]
Abstract
Cathepsin K is a cysteine protease member of the cathepsin lysosomal protease family. Although cathepsin K is highly expressed in osteoclasts, lower levels of cathepsin K are also found in a variety of other tissues. Secretion of cathepsin K from the osteoclast into the sealed osteoclast-bone cell interface results in efficient degradation of type I collagen. The absence of cathepsin K activity in humans results in pycnodysostosis, characterized by increased bone mineral density and fractures. Pharmacologic cathepsin K inhibition leads to continuous increases in bone mineral density for ≤5 years of treatment and improves bone strength at the spine and hip. Compared with other antiresorptive agents, cathepsin K inhibition is nearly equally efficacious for reducing biochemical markers of bone resorption but comparatively less active for reducing bone formation markers. Despite multiple efforts to develop cathepsin K inhibitors, potential concerns related to off-target effects of the inhibitors against other cathepsins and cathepsin K inhibition at nonbone sites, including skin and perhaps cardiovascular and cerebrovascular sites, prolonged the regulatory approval process. A large multinational randomized, double-blind phase III study of odanacatib in postmenopausal women with osteoporosis was recently completed. Although that study demonstrated clinically relevant reductions in fractures at multiple sites, odanacatib was ultimately withdrawn from the regulatory approval process after it was found to be associated with an increased risk of cerebrovascular accidents. Nonetheless, the underlying biology and clinical effects of cathepsin K inhibition remain of considerable interest and could guide future therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Matthew T. Drake
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Bart L. Clarke
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Merry Jo Oursler
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Sundeep Khosla
- Division of Endocrinology and Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| |
Collapse
|
25
|
He LH, Liu M, He Y, Xiao E, Zhao L, Zhang T, Yang HQ, Zhang Y. TRPV1 deletion impaired fracture healing and inhibited osteoclast and osteoblast differentiation. Sci Rep 2017; 7:42385. [PMID: 28225019 PMCID: PMC5320507 DOI: 10.1038/srep42385] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/09/2017] [Indexed: 12/24/2022] Open
Abstract
Fracture healing, in which osteoclasts and osteoblasts play important roles, has drawn much clinical attention. Osteoclast deficiency or decreased osteoblast activity will impair fracture healing. TRPV1 is a member of the Ca2+ permeable cation channel subfamily, and pharmacological inhibition of TRPV1 prevents ovariectomy-induced bone loss, which makes TRPV1 a potential target for osteoporosis. However, whether long term TRPV1 inhibition or TRPV1 deletion will affect the fracture healing process is unclear. In this study, we found that the wild-type mice showed a well-remodeled fracture callus, whereas TRPV1 knockout mice still had an obvious fracture gap with unresorbed soft-callus 4 weeks post-fracture. The number of osteoclasts was reduced in the TRPV1 knockout fracture callus, and osteoclast formation and resorption activity were also impaired in vitro. TRPV1 deletion decreased the calcium oscillation frequency and peak cytoplasmic concentration in osteoclast precursors, subsequently reducing the expression and nuclear translocation of NFATc1 and downregulating DC-stamp, cathepsin K, and ATP6V. In addition, TRPV1 deletion caused reduced mRNA and protein expression of Runx2 and ALP in bone marrow stromal cells (BMSCs) and reduced calcium deposition in vitro. Our results suggest that TRPV1 deletion impairs fracture healing, and inhibited osteoclastogenesis and osteogenesis.
Collapse
Affiliation(s)
- Lin-Hai He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Meng Liu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - E. Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Lu Zhao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Ting Zhang
- Center for Craniofacial Stem Cell Research and Regeneration, Department of Orthodontics, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| | - Hua-Qian Yang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of digital Stomatology, Beijing, China
| |
Collapse
|
26
|
Drissi H, Sanjay A. The Multifaceted Osteoclast; Far and Beyond Bone Resorption. J Cell Biochem 2016; 117:1753-6. [PMID: 27019318 DOI: 10.1002/jcb.25560] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/24/2016] [Indexed: 01/21/2023]
Abstract
The accepted function of the bone resorbing cell, osteoclast, has been linked to bone remodeling and pathological osteolysis. Emerging evidence points to novel functions of osteoclasts in controlling bone formation and angiogenesis. Thus, while the concept of a "clastokine" with the potential to regulate osteogenesis during remodeling did not come as a surprise, new evidence provided unique insight into the mechanisms underlying osteoclastic control of bone formation. The question still remains as to whether osteoclast precursors or a unique trap positive mononuclear cell, can govern any aspect of bone formation. The novel paradigm eloquently proposed by leaders in the field brings together the concept of clastokines and osteoclast precursor-mediated bone formation, potentially though enhanced angiogenesis. These fascinating advances in osteoclast biology have motivated this short review, in which we discuss these new roles of osteoclasts. J. Cell. Biochem. 117: 1753-1756, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hicham Drissi
- University of Connecticut Health Center, Orthopaedics Surgery, 263 Farmington Ave, Farmington 06034, Connecticut
| | - Archana Sanjay
- University of Connecticut Health Center, Orthopaedics Surgery, 263 Farmington Ave, Farmington 06034, Connecticut
| |
Collapse
|
27
|
Brömme D, Panwar P, Turan S. Cathepsin K osteoporosis trials, pycnodysostosis and mouse deficiency models: Commonalities and differences. Expert Opin Drug Discov 2016; 11:457-72. [DOI: 10.1517/17460441.2016.1160884] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Dieter Brömme
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Preety Panwar
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
- Centre for Blood Research, University of British Columbia, Vancouver, Canada
| | - Serap Turan
- Department of Pediatric Endocrinology, Marmara University, Istanbul, Turkey
| |
Collapse
|
28
|
Pennypacker BL, Gilberto D, Gatto NT, Samadfam R, Smith SY, Kimmel DB, Duong LT. Odanacatib increases mineralized callus during fracture healing in a rabbit ulnar osteotomy model. J Orthop Res 2016; 34:72-80. [PMID: 26178170 PMCID: PMC6680236 DOI: 10.1002/jor.22982] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 07/04/2015] [Indexed: 02/04/2023]
Abstract
The effects of the cathepsin K inhibitor odanacatib (ODN) on fracture healing were monitored for ~6 and 15 weeks post-fracture in two separate studies using the unilateral transverse mid-ulnar osteotomy model in skeletally mature female rabbits. Rabbits were pre-treated for 3-4 weeks with vehicle (Veh), ODN (2 mg/kg, po, daily), or alendronate (ALN) (0.3 mg/kg, sc, twice-weekly) prior to osteotomy. In Study 1, the animals were maintained on the same respective treatment for ~6 weeks. In Study 2, the animals were also continued on the same therapy or switched from Veh to ODN or ODN to Veh for 15 weeks. No treatment-related impairment of fracture union was seen by qualitative histological assessments in the first study. Cartilage retention was detected in the calluses of ALN-treated rabbits at week-6, while calluses in the ODN and Veh groups contained bony tissue with significantly less residual cartilage. ODN treatment also markedly increased the number of cathepsin K-(+) osteoclasts in the callus, indicating enhanced callus remodeling. From the second study, ex vivo DXA and pQCT confirmed that ODN treatment pre- and post-osteotomy increased callus bone mineral content and bone mineral density (BMD) versus Veh (p < 0.001) and discontinuation of ODN post-surgery returned callus BMD to Veh. Peak load of ODN- or ALN-treated calluses were comparable to Veh. ODN increased callus yield load (20%, p = 0.056) and stiffness (26%, p < 0.05) versus Veh. These studies demonstrated that ODN increased mineralized callus during the early phase of fracture repair without impairing callus formation or biomechanical integrity at the fracture site.
Collapse
Affiliation(s)
| | - David Gilberto
- Safety Assessment and Laboratory Animal ResourcesMerck Research LaboratoriesWest PointPennsylvania
| | - Nicholas T. Gatto
- Safety Assessment and Laboratory Animal ResourcesMerck Research LaboratoriesWest PointPennsylvania
| | - Rana Samadfam
- Charles River LaboratoriesPreclinical ServicesSennevilleQuebecCanada
| | - Susan Y. Smith
- Charles River LaboratoriesPreclinical ServicesSennevilleQuebecCanada
| | | | - Le Thi Duong
- Bone BiologyMerck Research LaboratoriesWest PointPennsylvania
| |
Collapse
|
29
|
Drissi H, Paglia DN, Alaee F, Yoshida R. Constructing the toolbox: Patient-specific genetic factors of altered fracture healing. Genes Dis 2014; 1:140-148. [PMID: 25558470 PMCID: PMC4280851 DOI: 10.1016/j.gendis.2014.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/18/2014] [Indexed: 01/10/2023] Open
Abstract
The multifaceted sequence of events that follow fracture repair can be further complicated when considering risk factors for impaired union, present in a large and growing percentage of the population. Risk factors such as diabetes, substance abuse, and poor nutrition affect both the young and old alike, and have been shown to dramatically impair the body's natural healing processes. To this end, biotherapeudic interventions such as ultrasound, electrical simulation, growth factor treatment (BMP-2, BMP-7, PDGF-BB, FGF-2) have been evaluated in preclinical models and in some cases are used widely for patients with established non-union or risk/indication or impaired healing (ie. ultrasound, BMP-2, etc.). Despite the promise of these interventions, they have been shown to be reliant on patient compliance and can produce adverse side-effects such as heterotopic ossification. Gene and cell therapy approaches have attempted to apply controlled regimens of these factors and have produced promising results. However, there are safety and efficacy concerns that may limit the translation of these approaches. In addition, none of the above mentioned approaches consider genetic variation between individual patients. Several clinical and preclinical studies have demonstrated a genetic component to fracture repair and that SNPs and genetic background variation play major roles in the determination of healing outcomes. Despite this, there is a need for preclinical data to dissect the mechanism underlying the influence of specific gene loci on the processes of fracture healing, which will be paramount in the future of patient-centered interventions for fracture repair.
Collapse
Affiliation(s)
- Hicham Drissi
- New England Musculoskeletal Institute and Department of Orthopaedic Surgery, United States
| | | | | | | |
Collapse
|