1
|
Zhou H, Zhang YF, Zhang QQ, Liu F, Zhang JY, Chen Y. Cathepsin K inhibition alleviates periodontal bone resorption by promoting type H vessel formation through PDGF-BB/PDGFR-β axis. Oral Dis 2024; 30:5335-5348. [PMID: 38462960 DOI: 10.1111/odi.14920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVES To explore the effects of cathepsin K (CTSK) inhibition on type H vessel formation and alveolar bone resorption within periodontitis. METHODS Conditioned media derived from preosteoclasts pretreated with the CTSK inhibitor odanacatib (ODN), ODN supplemented small interfering RNA targeting PDGF-BB (si-PDGF-BB), or PBS were prepared, to assess their proangiogenic effects on endothelial cells (HUVECs). A series of angiogenic-related assays were conducted to evaluate HUVEC proliferation, migration, and tube formation abilities in vitro. In addition, qRT-PCR and Western blot assays were employed to examine the expression levels of genes/proteins related to PDGF-BB/PDGFR-β axis components. A mouse periodontitis model was established to evaluate the effects of CTSK inhibition on type H vessel formation. RESULTS CTSK inhibition promoted PDGF-BB secretion from preosteoclasts and proliferation, migration, and tube formation activities of HUVECs in vitro. However, the conditioned medium from preosteoclasts pretreated by si-PDGF-BB impaired the angiogenic activities of HUVECs. This promoted angiogenesis function by CTSK inhibition may be mediated by the PDGF-BB/PDGFR-β axis. Functionally, in vivo studies demonstrated that CTSK inhibition significantly accelerated type H vessel formation and alleviated bone loss within periodontitis. CONCLUSION CTSK inhibition promotes type H vessel formation and attenuates alveolar bone resorption within periodontitis via PDGF-BB/PDGFR-β axis.
Collapse
Affiliation(s)
- Huan Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yi-Fan Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Qian-Qian Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Fen Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Jia-Yu Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yue Chen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
2
|
Liu W, Wang Q, Luo H, Luo B, Zhao F, Kang Y, Zhang Y, Shao L. Nanographene Oxide Promotes Angiogenesis by Regulating Osteoclast Differentiation and Platelet-Derived Growth Factor Secretion. ACS NANO 2024; 18:22390-22403. [PMID: 39105734 DOI: 10.1021/acsnano.4c06979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
An imbalanced system of angiogenesis-osteoblasts-osteoclasts is regarded as the main factor in bone remodeling dysfunction diseases or osseointegration loss. Osteoclast precursors are the key cells that accelerate bone-specific angiogenesis and maintain normal osteoblast and osteoclast function. Graphene oxide is an effective scaffold surface modification agent with broad application prospects in bone tissue engineering. However, the effect of graphene oxide on the interaction between osteoclasts and angiogenesis has not yet been elucidated. In this study, a rat calvarial defect model was established and treated with an electrochemically derived nanographene oxide (ENGO) hydrogel. Higher angiogenesis and platelet-derived growth factor (PDGF) B in preosteoclasts were observed in the ENGO group compared with that in the control group. Moreover, in vitro experiments demonstrate the efficacy of ENGO in substantially reducing the expression of the receptor activator of nuclear factor-kappaB ligand (RANKL)-induced osteoclast-associated markers and inhibiting bone resorption activity. Additionally, ENGO enhances the secretion of the osteoclast-derived coupling factor PDGF-BB and promotes angiogenesis. Our investigation revealed the crucial role of isocitrate dehydrogenase 1 (IDH1) in the ENGO-mediated regulation of osteoclast differentiation and PDGF-BB secretion. The decreased expression of IDH1 reduces the level of histone lysine demethylase 7A (KDM7A) and subsequently increases the H3K9me2 level in the cathepsin K promoter region. In summary, we found that ENGO promotes angiogenesis by inhibiting the maturity of RANKL-induced osteoclasts and enhancing PDGF-BB secretion. These results indicate that ENGO holds promise for the application in fostering osteoclast-endothelial cell crosstalk, providing an effective strategy for treating bone resorption and osteoclast-related bone loss diseases.
Collapse
Affiliation(s)
- Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Qinying Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Haiyun Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Bichong Luo
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Yiyuan Kang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
3
|
Zhang D, Wang Y, Zhou Z, Wang L, Liu C, Jiang Y. Role of miRNA-regulated type H vessel formation in osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1394785. [PMID: 38883597 PMCID: PMC11176424 DOI: 10.3389/fendo.2024.1394785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Osteoporosis (OP) is a chronic systemic bone metabolism disease characterized by decreased bone mass, microarchitectural deterioration, and fragility fractures. With the demographic change caused by long lifespans and population aging, OP is a growing health problem. The role of miRNA in the pathogenesis of OP has also attracted widespread attention from scholars in recent years. Type H vessels are unique microvessels of the bone and have become a new focus in the pathogenesis of OP because they play an essential role in osteogenesis-angiogenesis coupling. Previous studies found some miRNAs regulate type H vessel formation through the regulatory factors, including platelet-derived growth factor-BB (PDGF-BB), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and so on. These findings help us gain a more in-depth understanding of the relationship among miRNAs, type H vessels, and OP to find a new perspective on treating OP. In the present mini-review, we will introduce the role of type H vessels in the pathogenesis of OP and the regulation of miRNAs on type H vessel formation by affecting regulatory factors to provide some valuable insights for future studies of OP treatment.
Collapse
Affiliation(s)
- Dailiang Zhang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yongjing Wang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Zunzhen Zhou
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Limei Wang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Chongzhi Liu
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yuan Jiang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Ma T, Wang Y, Ma J, Cui H, Feng X, Ma X. Research progress in the pathogenesis of hormone-induced femoral head necrosis based on microvessels: a systematic review. J Orthop Surg Res 2024; 19:265. [PMID: 38671500 PMCID: PMC11046814 DOI: 10.1186/s13018-024-04748-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Hormonal necrosis of the femoral head is caused by long-term use of glucocorticoids and other causes of abnormal bone metabolism, lipid metabolism imbalance and blood microcirculation disorders in the femoral head, resulting in bone trabecular fracture, bone tissue necrosis collapse, and hip dysfunction. It is the most common type of non-traumatic necrosis of the femoral head, and its pathogenesis is complex, while impaired blood circulation is considered to be the key to its occurrence. There are a large number of microvessels in the femoral head, among which H-type vessels play a decisive role in the "angiogenesis and osteogenesis coupling", and thus have an important impact on the occurrence and development of femoral head necrosis. Glucocorticoids can cause blood flow injury of the femoral head mainly through coagulation dysfunction, endothelial dysfunction and impaired angiogenesis. Glucocorticoids may inhibit the formation of H-type vessels by reducing the expression of HIF-1α, PDGF-BB, VGEF and other factors, thus causing damage to the "angiogenesis-osteogenesis coupling" and reducing the ability of necrosis reconstruction and repair of the femoral head. Leads to the occurrence of hormonal femoral head necrosis. Therefore, this paper reviewed the progress in the study of the mechanism of hormone-induced femoral head necrosis based on microvascular blood flow at home and abroad, hoping to provide new ideas for the study of the mechanism of femoral head necrosis and provide references for clinical treatment of femoral head necrosis.
Collapse
Affiliation(s)
- Tiancheng Ma
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Yan Wang
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Jianxiong Ma
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China.
- Tianjin Orthopedic Institute, Tianjin, 300050, China.
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China.
| | - Hongwei Cui
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Xiaotian Feng
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| | - Xinlong Ma
- Tianjin Hospital of Tianjin University, Tianjin, 300211, China
- Tianjin Orthopedic Institute, Tianjin, 300050, China
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Tianjin, 300050, China
| |
Collapse
|
5
|
Qiu J, Liu J, Tian L, Yu J, Duan Q, Liu Y, Zhao W, Si H, Lu X, Zhang Q. Knockdown of LOX-1 ameliorates bone quality and generation of type H blood vessels in diabetic mice. Arch Biochem Biophys 2024; 752:109870. [PMID: 38141905 DOI: 10.1016/j.abb.2023.109870] [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: 07/29/2023] [Revised: 11/02/2023] [Accepted: 12/15/2023] [Indexed: 12/25/2023]
Abstract
Our previous studies have shown that lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) is expressed in liver sinusoidal endothelial cells, and oxidized low-density lipoprotein induces liver sinusoidal dysfunction and defenestration through the LOX-1/ROS/NF-kB pathway, revealing that LOX-1 can mediate liver sinusoidal barrier function, involved in the regulation of non-alcoholic fatty liver disease. Here, we investigated whether, in the context of bone metabolic diseases, LOX-1 could affect bone quality and type H blood vessels in diabetic mice. We used db/db mice as model and found that LOX-1 knockdown can ameliorate bone quality and type H blood vessel generation in db/db mice. This further verifies our hypothesis that LOX-1 is involved in the regulation of bone quality and type H blood vessel homeostasis, thus inhibiting osteoporosis progression in db/db mice.
Collapse
Affiliation(s)
- Jumei Qiu
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Jing Liu
- Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China; Clinical Research Center for Metabolic Disease, Lanzhou, 730000, Gansu Province, China
| | - Limin Tian
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China; Clinical Research Center for Metabolic Disease, Lanzhou, 730000, Gansu Province, China
| | - Jing Yu
- Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China; Clinical Research Center for Metabolic Disease, Lanzhou, 730000, Gansu Province, China
| | - Qidang Duan
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Yaqian Liu
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Wenshu Zhao
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Huiling Si
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China
| | - Xun Lu
- Ningxia Medical University, Yinchuan, 750000, Ningxia Hui Autonomous Region, China
| | - Qi Zhang
- First Clinical School of Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu Province, China; Department of Geriatrics, Gansu Provincial Hospital, Lanzhou, 730000, Gansu Province, China; Clinical Research Center for Metabolic Disease, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
6
|
Xu J, He S, Xia T, Shan Y, Wang L. Targeting type H vessels in bone-related diseases. J Cell Mol Med 2024; 28:e18123. [PMID: 38353470 PMCID: PMC10865918 DOI: 10.1111/jcmm.18123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/01/2024] [Accepted: 01/09/2024] [Indexed: 02/16/2024] Open
Abstract
Blood vessels are essential for bone development and metabolism. Type H vessels in bone, named after their high expression of CD31 and Endomucin (Emcn), have recently been reported to locate mainly in the metaphysis, exhibit different molecular properties and couple osteogenesis and angiogenesis. A strong correlation between type H vessels and bone metabolism is now well-recognized. The crosstalk between type H vessels and osteoprogenitor cells is also involved in bone metabolism-related diseases such as osteoporosis, osteoarthritis, fracture healing and bone defects. Targeting the type H vessel formation may become a new approach for managing a variety of bone diseases. This review highlighted the roles of type H vessels in bone-related diseases and summarized the research attempts to develop targeted intervention, which will help us gain a better understanding of their potential value in clinical application.
Collapse
Affiliation(s)
- Juan Xu
- Outpatient DepartmentChildren's Hospital of Soochow UniversitySuzhouChina
| | - Shuang‐jian He
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Ting‐ting Xia
- Clinical Research InstituteSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
| | - Yu Shan
- Department of OrthopeadicsSuzhou Ninth Hospital Affiliated to Soochow UniversitySuzhouChina
| | - Liang Wang
- Department of OrthopaedicsSuzhou Hospital, Affiliated Hospital of Medical School, Nanjing UniversitySuzhouChina
- Department of OrthopeadicsThe Fourth Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
7
|
Liu X, Zhang P, Gu Y, Guo Q, Liu Y. Type H vessels: functions in bone development and diseases. Front Cell Dev Biol 2023; 11:1236545. [PMID: 38033859 PMCID: PMC10687371 DOI: 10.3389/fcell.2023.1236545] [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: 06/09/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Type H vessels are specialized blood vessels found in the bone marrow that are closely associated with osteogenic activity. They are characterized by high expression of endomucin and CD31. Type H vessels form in the cancellous bone area during long bone development to provide adequate nutritional support for cells near the growth plate. They also influence the proliferation and differentiation of osteoprogenitors and osteoclasts in a paracrine manner, thereby creating a suitable microenvironment to facilitate new bone formation. Because of the close relationship between type H vessels and osteogenic activity, it has been found that type H vessels play a role in the physiological and pathological processes of bone diseases such as fracture healing, osteoporosis, osteoarthritis, osteonecrosis, and tumor bone metastasis. Moreover, experimental treatments targeting type H vessels can improve the outcomes of these diseases. Here, we reviewed the molecular mechanisms related to type H vessels and their associated osteogenic activities, which are helpful in further understanding the role of type H vessels in bone metabolism and will provide a theoretical basis and ideas for comprehending bone diseases from the vascular perspective.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Colorectal and Anal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| | - Peilin Zhang
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiaoyue Guo
- Endocrinology Research Center, Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yonggan Liu
- Department of Colorectal and Anal Surgery, Zhongshan City People’s Hospital, Zhongshan, Guangdong, China
| |
Collapse
|
8
|
Qin X, Xi Y, Jiang Q, Chen C, Yang G. Type H vessels in osteogenesis, homeostasis, and related disorders. Differentiation 2023; 134:20-30. [PMID: 37774549 DOI: 10.1016/j.diff.2023.09.005] [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: 07/05/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023]
Abstract
The vascular system plays a crucial role in bone tissue. Angiogenic and osteogenic processes are coupled through a spatial-temporal connection. Recent studies have identified three types of capillaries in the skeletal system. Compared with type L and E vessels, type H vessels express high levels of CD31 and endomucin, and function to couple angiogenesis and osteogenesis. Endothelial cells in type H vessels interact with osteolineage cells (e.g., osteoblasts, osteoclasts, and osteocytes) through cytokines or signaling pathways to maintain bone growth and homeostasis. In imbalanced bone homeostases, such as osteoporosis and osteoarthritis, it may be a new therapeutic strategy to regulate the endothelial cell activity in type H vessels to repair the imbalance. Here, we reviewed the latest progress in relevant factors or signaling pathways in coupling angiogenesis and osteogenesis. This review would contribute to further understanding the role and mechanisms of type H vessels in coupling angiogenic and osteogenic processes. Furthermore, it will facilitate the development of therapeutic approaches for bone disorders by targeting type H vessels.
Collapse
Affiliation(s)
- Xiaoru Qin
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Yue Xi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Qifeng Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Chaozhen Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
9
|
Khoswanto C. Role of matrix metalloproteinases in bone regeneration: Narrative review. J Oral Biol Craniofac Res 2023; 13:539-543. [PMID: 37351418 PMCID: PMC10282173 DOI: 10.1016/j.jobcr.2023.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/14/2023] [Accepted: 06/07/2023] [Indexed: 06/24/2023] Open
Abstract
Background Matrix metalloproteinases (MMPs) not only work as enzymes but also as degrading enzymes that have been shown to play an important function in extracellular matrix (ECM) regeneration, including bone regeneration. To generate new bone tissue, bone regeneration or repair relies on a series of regulated processes in which MMPs play an important role. Bone cells express the MMPs in an active state, and these MMPs are assumed to have a crucial role, not only for the viability and functionality of osteoclasts, osteoblasts, and osteocytes but also for the formation and development of chondrocytes. Objective This study aimed to review and present the roles of matrix metalloproteinases in bone regeneration. Methods An analysis of the scientific literature on the topics of matrix metalloproteinases in bone regeneration was done on PubMed and Google Scholar. Search results were screened for articles that described or investigated the impacts matrix metalloproteinases have on bones in relation to dentistry. The journals' cited papers were also assessed for relevance and included if they complied with the criteria for inclusion. Accessibility to the full document was one of the prerequisites for admission. Result Bone regeneration are intricate ongoing processes involving numerous MMPs, especially MMP 2, 9 and 13. MMP-2 appears to alter bone growth through influencing osteoclast and osteoblast activity and proliferation, MMP-9-deficient animals have abnormal bone formation exclusively during endochondral ossification, MMP 13 is responsible for osteoclast receptor activation, has been linked to the breakdown bone resorption. Conclusions MMP 2, 9, and 13 play a major protective role in osteogenesis and bone regeneration.
Collapse
Affiliation(s)
- Christian Khoswanto
- Department of Oral Biology Faculty of Dentistry, Airlangga University. Jln. Mayjend. Prof. Dr. Moestopo No. 47, Surabaya, 60132, Indonesia
| |
Collapse
|
10
|
Watanabe H, Maishi N, Hoshi-Numahata M, Nishiura M, Nakanishi-Kimura A, Hida K, Iimura T. Skeletal-Vascular Interactions in Bone Development, Homeostasis, and Pathological Destruction. Int J Mol Sci 2023; 24:10912. [PMID: 37446097 DOI: 10.3390/ijms241310912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bone is a highly vascularized organ that not only plays multiple roles in supporting the body and organs but also endows the microstructure, enabling distinct cell lineages to reciprocally interact. Recent studies have uncovered relevant roles of the bone vasculature in bone patterning, morphogenesis, homeostasis, and pathological bone destruction, including osteoporosis and tumor metastasis. This review provides an overview of current topics in the interactive molecular events between endothelial cells and bone cells during bone ontogeny and discusses the future direction of this research area to find novel ways to treat bone diseases.
Collapse
Affiliation(s)
- Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Nako Maishi
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| |
Collapse
|
11
|
Liu X, Gu Y, Kumar S, Amin S, Guo Q, Wang J, Fang CL, Cao X, Wan M. Oxylipin-PPARγ-initiated adipocyte senescence propagates secondary senescence in the bone marrow. Cell Metab 2023; 35:667-684.e6. [PMID: 37019080 PMCID: PMC10127143 DOI: 10.1016/j.cmet.2023.03.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/12/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023]
Abstract
The chronic use of glucocorticoids decreases bone mass and quality and increases bone-marrow adiposity, but the underlying mechanisms remain unclear. Here, we show that bone-marrow adipocyte (BMAd) lineage cells in adult mice undergo rapid cellular senescence upon glucocorticoid treatment. The senescent BMAds acquire a senescence-associated secretory phenotype, which spreads senescence in bone and bone marrow. Mechanistically, glucocorticoids increase the synthesis of oxylipins, such as 15d-PGJ2, for peroxisome proliferator-activated receptor gamma (PPARγ) activation. PPARγ stimulates the expression of key senescence genes and also promotes oxylipin synthesis in BMAds, forming a positive feedback loop. Transplanting senescent BMAds into the bone marrow of healthy mice is sufficient to induce the secondary spread of senescent cells and bone-loss phenotypes, whereas transplanting BMAds harboring a p16INK4a deletion did not show such effects. Thus, glucocorticoid treatment induces a lipid metabolic circuit that robustly triggers the senescence of BMAd lineage cells that, in turn, act as the mediators of glucocorticoid-induced bone deterioration.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yiru Gu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Surendra Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sahran Amin
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jiekang Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ching-Lien Fang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Cao H, Shi K, Long J, Liu Y, Li L, Ye T, Huang C, Lai Y, Bai X, Qin L, Wang X. PDGF-BB prevents destructive repair and promotes reparative osteogenesis of steroid-associated osteonecrosis of the femoral head in rabbits. Bone 2023; 167:116645. [PMID: 36539110 DOI: 10.1016/j.bone.2022.116645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/28/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
Destructive repair characterized by inadequate angiogenesis and osteogenesis is the main pathological progression in steroid-associated osteonecrosis of the femoral head (SONFH). Platelet-derived growth factor-BB (PDGF-BB) is an "angiogenesis and osteogenesis coupling" factor that has been used for the treatment of bone defects in clinic. This study was designed to analyze the ability of PDGF-BB for preventing destructive repair and promoting reparative osteogenesis in SONFH. Steroid-associated osteonecrosis (SAON) was induced and triggered destructive repair of the femoral head by repeated lipopolysaccharide (LPS) and methylprednisolone (MPS) injections in rabbits. At 2, 4, and 6 weeks after induction, recombinant human PDGF-BB, neutralizing PDGF-BB antibody, or saline was intramedullary injected into the proximal femora. At week 6 after SAON induction, the proximal femora were dissected for bone architecture and histological analysis. C3H10T1/2 cells and HUVECs were used for further mechanistic investigation. After PDGF-BB treatment, type H vessels and leptin receptor-positive (LepR+) mesenchymal stem cells (MSCs) increased in the affected femoral head, and more osteoblastic osteogenesis along the bone surfaces but scattered adipocytes in bone marrow tissue than that in the SAON group. PDGF-BB treatment prevented destructive repair progression and led to 50-70 % of osteonecrotic femoral heads undergoing reparative osteogenesis. In particular, we found that PDGF-BB could mediate MSC self-renewal and maintain their osteogenic potency by activating PDGFR/Akt/GSK3β/CERB signaling in the presence of steroids. Moreover, PDGF-BB also stabled the newly formed vascular tubes by recruiting MSCs for improving intraosseous vascular integration. PDGF-BB may be a candidate for the promotion of reparative osteogenesis in SONFH.
Collapse
Affiliation(s)
- Huijuan Cao
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Keda Shi
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Jing Long
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Yanzhi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, PR China
| | - Lingli Li
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Tianluo Ye
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Cuishan Huang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Xueling Bai
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Ling Qin
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong.
| | - Xinluan Wang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China; University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
13
|
Gao Z, Gao Z, Zhang H, Hou S, Zhou Y, Liu X. Targeting STING: From antiviral immunity to treat osteoporosis. Front Immunol 2023; 13:1095577. [PMID: 36741390 PMCID: PMC9891206 DOI: 10.3389/fimmu.2022.1095577] [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: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
The cGAS-STING signaling pathway can trigger innate immune responses by detecting dsDNA from outside or within the host. In addition, the cGAS-STING signaling pathway has emerged as a critical mediator of the inflammatory response and a new target for inflammatory diseases. STING activation leads to dimerization and translocation to the endoplasmic reticulum Golgi intermediate compartment or Golgi apparatus catalyzed by TBK1, triggers the production of IRF3 and NF-κB and translocates to the nucleus to induce a subsequent interferon response and pro-inflammatory factor production. Osteoporosis is a degenerative bone metabolic disease accompanied by chronic sterile inflammation. Activating the STING/IFN-β signaling pathway can reduce bone resorption by inhibiting osteoclast differentiation. Conversely, activation of STING/NF-κB leads to the formation of osteoporosis by increasing bone resorption and decreasing bone formation. In addition, activation of STING inhibits the generation of type H vessels with the capacity to osteogenesis, thereby inhibiting bone formation. Here, we outline the mechanism of action of STING and its downstream in osteoporosis and discuss the role of targeting STING in the treatment of osteoporosis, thus providing new ideas for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhonghua Gao
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongguo Gao
- Department of Medical Laboratory Technology, School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hao Zhang
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shoubo Hou
- Department of General Practice, General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Yunhua Zhou
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Yunhua Zhou, ; Xiangjie Liu,
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Yunhua Zhou, ; Xiangjie Liu,
| |
Collapse
|
14
|
Li Z, Liu C, Liu X, Wang N, Gao L, Bao X, Liu S, Xue P. Aucubin Impeded Preosteoclast Fusion and Enhanced CD31 hi EMCN hi Vessel Angiogenesis in Ovariectomized Mice. Stem Cells Int 2022; 2022:5226771. [PMID: 36406003 PMCID: PMC9668463 DOI: 10.1155/2022/5226771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/27/2022] [Indexed: 08/31/2023] Open
Abstract
Osteogenesis is tightly correlated with angiogenesis during the process of bone development, regeneration, and remodeling. In addition to providing nutrients and oxygen for bone tissue, blood vessels around bone tissue also secrete some factors to regulate bone formation. Type H vessels which were regulated by platelet-derived growth factor-BB (PDGF-BB) were confirmed to couple angiogenesis and osteogenesis. Recently, preosteoclasts have been identified as the most important source of PDGF-BB. Therefore, inhibiting osteoclast maturation, improving PDGF-BB secretion, stimulating type H angiogenesis, and subsequently accelerating bone regeneration may be potent treatments for bone loss disease. In the present study, aucubin, an iridoid glycoside extracted from Aucuba japonica and Eucommia ulmoides, was found to inhibit bone loss in ovariectomized mice. We further confirmed that aucubin could inhibit the fusion of tartrate-resistant acid phosphatase (TRAP)+ preosteoclasts into mature osteoclasts and indirectly increasing angiogenesis of type H vessel. The underlying mechanism is the aucubin-induced inhibition of MAPK/NF-κB signaling, which increases the preosteoclast number and subsequently promotes angiogenesis via PDGF-BB. These results prompted that aucubin could be an antiosteoporosis drug candidate, which needs further research.
Collapse
Affiliation(s)
- Ziyi Li
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| | - Chang Liu
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| | - Xiaoli Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology & Hebei Key Laboratory of Stomatology, Hebei Medical University, Shijiazhuang 050017, China
| | - Na Wang
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| | - Liu Gao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| | - Xiaoxue Bao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| | - Sijing Liu
- Editorial Department of Hebei Medical University, Hebei Medical University, Shijiazhuang 050017, China
| | - Peng Xue
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
- Key Orthopaedic Biomechanics Laboratory of Hebei Province, Shijiazhuang 050051, China
| |
Collapse
|
15
|
Zheng G, Ma HW, Xiang GH, He GL, Cai HC, Dai ZH, Chen YL, Lin Y, Xu HZ, Ni WF, Xu C, Liu HX, Wang XY. Bone-targeting delivery of platelet lysate exosomes ameliorates glucocorticoid-induced osteoporosis by enhancing bone-vessel coupling. J Nanobiotechnology 2022; 20:220. [PMID: 36310171 PMCID: PMC9620632 DOI: 10.1186/s12951-022-01400-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Glucocorticoids (GCs) overuse is associated with decreased bone mass and osseous vasculature destruction, leading to severe osteoporosis. Platelet lysates (PL) as a pool of growth factors (GFs) were widely used in local bone repair by its potent pro-regeneration and pro-angiogenesis. However, it is still seldom applied for treating systemic osteopathia due to the lack of a suitable delivery strategy. The non-targeted distribution of GFs might cause tumorigenesis in other organs. RESULTS In this study, PL-derived exosomes (PL-exo) were isolated to enrich the platelet-derived GFs, followed by conjugating with alendronate (ALN) grafted PEGylated phospholipid (DSPE-PEG-ALN) to establish a bone-targeting PL-exo (PL-exo-ALN). The in vitro hydroxyapatite binding affinity and in vivo bone targeting aggregation of PL-exo were significantly enhanced after ALN modification. Besides directly modulating the osteogenic and angiogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and endothelial progenitor cells (EPCs), respectively, PL-exo-ALN also facilitate their coupling under GCs' stimulation. Additionally, intravenous injection of PL-exo-ALN could successfully rescue GCs induced osteoporosis (GIOP) in vivo. CONCLUSIONS PL-exo-ALN may be utilized as a novel nanoplatform for precise infusion of GFs to bone sites and exerts promising therapeutic potential for GIOP.
Collapse
Affiliation(s)
- Gang Zheng
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hai-Wei Ma
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Guang-Heng Xiang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Gao-Lu He
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Han-Chen Cai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zi-Han Dai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Yan-Lin Chen
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang Province, China
| | - Yan Lin
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hua-Zi Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Wen-Fei Ni
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Cong Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Hai-Xiao Liu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Xiang-Yang Wang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
16
|
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
|
17
|
Li Z, Wang H, Zhang K, Yang B, Xie X, Yang Z, Kong L, Shi P, Zhang Y, Ho YP, Zhang ZY, Li G, Bian L. Bisphosphonate-based hydrogel mediates biomimetic negative feedback regulation of osteoclastic activity to promote bone regeneration. Bioact Mater 2022; 13:9-22. [PMID: 35224288 PMCID: PMC8844702 DOI: 10.1016/j.bioactmat.2021.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
The intricate dynamic feedback mechanisms involved in bone homeostasis provide valuable inspiration for the design of smart biomaterial scaffolds to enhance in situ bone regeneration. In this work, we assembled a biomimetic hyaluronic acid nanocomposite hydrogel (HA-BP hydrogel) by coordination bonds with bisphosphonates (BPs), which are antiosteoclastic drugs. The HA-BP hydrogel exhibited expedited release of the loaded BP in response to an acidic environment. Our in vitro studies showed that the HA-BP hydrogel inhibits mature osteoclastic differentiation of macrophage-like RAW264.7 cells via the released BP. Furthermore, the HA-BP hydrogel can support the initial differentiation of primary macrophages to preosteoclasts, which are considered essential during bone regeneration, whereas further differentiation to mature osteoclasts is effectively inhibited by the HA-BP hydrogel via the released BP. The in vivo evaluation showed that the HA-BP hydrogel can enhance the in situ regeneration of bone. Our work demonstrates a promising strategy to design biomimetic biomaterial scaffolds capable of regulating bone homeostasis to promote bone regeneration. HA-BP hydrogel can mediate the expedited release of BP in response to the acidic microenvironment created by osteoclasts. HA-BP hydrogel supports preosteoclastic differentiation, but inhibits the further osteoclastic maturation. The implantation of HA-BP hydrogel in critical-sized bone defects significantly promotes in situ bone regeneration in vivo.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Lingchi Kong
- Department of Orthopaedic Surgery, Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, 510150, China
- Corresponding author.
| | - Gang Li
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
- Corresponding author.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Corresponding author. School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
18
|
Liu P, Gao Y, Luo P, Yu H, Guo S, Liu F, Gao J, Xu J, Wang S, Zhang C. Glucocorticoid-induced expansion of classical monocytes contributes to bone loss. Exp Mol Med 2022; 54:765-776. [PMID: 35672449 PMCID: PMC9256622 DOI: 10.1038/s12276-022-00764-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Classical monocytes are commonly involved in the innate inflammatory response and are the progenitors of osteoclasts. Excess endogenous glucocorticoids (GCs) can increase the levels of classical monocytes in blood and bone marrow. The role of this cell population in high-dose exogenous GC-induced osteoporosis (GIOP) remains to be elucidated. In this study, GIOP was established in rats and mice by daily methylprednisolone injection, and monocyte subsets were analyzed by flow cytometry. We demonstrated that classical monocytes accumulate in bone marrow during GIOP. Similarly, the monocyte proportion among bone marrow nucleated cells was also increased in patients with steroid treatment history. We sorted classical monocytes and analyzed their transcriptional profile in response to GCs by RNA sequencing. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that classical monocytes isolated from GC-treated rats exhibited osteoclast differentiation potential. Deletion of classical monocytes by clodronate liposome treatment prevented GIOP via inhibition of osteoclastogenesis and restoration of CD31HiendomucinHi vessels. Regarding the molecular mechanism, classical monocytes express high levels of glucocorticoid receptors. In vitro treatment with GCs increased both the percentage and absolute number of monocytes and promoted their proliferation. In summary, classical monocytes mediated GC-induced bone loss and are a potential target for therapeutic intervention in GIOP treatment.
Collapse
Affiliation(s)
- Pei Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Pengbo Luo
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Hongping Yu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Shang Guo
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Fuyun Liu
- Department of Orthopedic Surgery, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Junjie Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Jianzhong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Shengdian Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, China.
| |
Collapse
|
19
|
He WZ, Yang M, Jiang Y, He C, Sun YC, Liu L, Huang M, Jiao YR, Chen KX, Hou J, Huang M, Xu YL, Feng X, Liu Y, Guo Q, Peng H, Huang Y, Su T, Xiao Y, Li Y, Zeng C, Lei G, Luo XH, Li CJ. miR-188-3p targets skeletal endothelium coupling of angiogenesis and osteogenesis during ageing. Cell Death Dis 2022; 13:494. [PMID: 35610206 PMCID: PMC9130327 DOI: 10.1038/s41419-022-04902-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
A specific bone capillary subtype, namely type H vessels, with high expression of CD31 and endomucin, was shown to couple angiogenesis and osteogenesis recently. The number of type H vessels in bone tissue declines with age, and the underlying mechanism for this reduction is unclear. Here, we report that microRNA-188-3p (miR-188-3p) involves this process. miRNA-188-3p expression is upregulated in skeletal endothelium and negatively regulates the formation of type H vessels during ageing. Mice with depletion of miR-188 showed an alleviated age-related decline in type H vessels. In contrast, endothelial-specific overexpression of miR-188-3p reduced the number of type H vessels, leading to decreased bone mass and delayed bone regeneration. Mechanistically, we found that miR-188 inhibits type H vessel formation by directly targeting integrin β3 in endothelial cells. Our findings indicate that miR-188-3p is a key regulator of type H vessel formation and may be a potential therapeutic target for preventing bone loss and accelerating bone regeneration.
Collapse
Affiliation(s)
- Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yangzi Jiang
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yu-Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yi-Li Xu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, Hunan, China.
| |
Collapse
|
20
|
Lin X, Xu F, Zhang KW, Qiu WX, Zhang H, Hao Q, Li M, Deng XN, Tian Y, Chen ZH, Qian AR. Acacetin Prevents Bone Loss by Disrupting Osteoclast Formation and Promoting Type H Vessel Formation in Ovariectomy-Induced Osteoporosis. Front Cell Dev Biol 2022; 10:796227. [PMID: 35517504 PMCID: PMC9062130 DOI: 10.3389/fcell.2022.796227] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Osteoporosis, characterized by the destruction of bone resorption and bone formation, is a serious disease that endangers human health. Osteoporosis prevention and treatment has become one of the important research contents in the field of medicine. Acacetin, a natural flavonoid compound, could promote osteoblast differentiation, and inhibit osteoclast formation in vitro. However, the mechanisms of acacetin on osteoclast differentiation and type H vessel formation, as well as the effect of preventing bone loss, remain unclear. Here, we firstly used primary bone marrow derived macrophages (BMMs), endothelial progenitor cells (EPCs), and ovariectomized (OVX) mice to explore the function of acacetin on bone remodeling and H type vessel formation. In this study, we found that acacetin inhibits osteoclast formation and bone resorption of BMMs induced by the macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) in a concentration of 20 μM without exerting cytotoxic effects. It was accompanied by downregulation of osteoclast differentiation marker genes (Ctsk, Acp5, and Mmp9) and cell fusion genes (CD9, CD47, Atp6v0d2, Dc-stamp, and Oc-stamp). Moreover, acacetin disrupted actin ring formation and extracellular acidification in osteoclasts. Mechanistic analysis revealed that acacetin not only inhibits the expression of the major transcription factor NFATc1 and NF-κB during RANKL-induced osteoclast formation, but also suppresses RANKL-induced the phosphorylation of Akt, GSK3β, IκBα, and p65. Additionally, acacetin enhanced the ability of M-CSF and RANKL-stimulated BMMs to promote angiogenesis and migration of EPCs. We further established that, in vivo, acacetin increased trabecular bone mass, decreased the number of osteoclasts, and showed more type H vessels in OVX mice. These data demonstrate that acacetin prevents OVX-induced bone loss in mice through inhibition of osteoclast function and promotion of type H vessel formation via Akt/GSK3β and NF-κB signalling pathway, suggesting that acacetin may be a novel therapeutic agent for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Xiao Lin
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Fang Xu
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Ke-Wen Zhang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Wu-Xia Qiu
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Hui Zhang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Xiao-Ni Deng
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| | - Zhi-Hao Chen
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| | - Ai-Rong Qian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
- *Correspondence: Ye Tian, ; Zhi-Hao Chen, ; Ai-Rong Qian,
| |
Collapse
|
21
|
Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol 2022; 123:36-47. [PMID: 34281770 DOI: 10.1016/j.semcdb.2021.06.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis, hematopoiesis and osteogenesis are fundamental processes mediating complex and essential biological functions. In the bone marrow, endothelial cells (ECs) are a principal mediator of regulatory signals that govern hematopoietic and mesenchymal stem cells. EC and osteoblast interactions and niche functions of ECs are fundamental in maintaining bone health and coordinating repair and regeneration following injury. These cellular interactions are subject to dysregulation and deterioration under stress, aging, chronic disease states and malignancy. Thus, the prospect of manipulating the bone vasculature has tremendous potential to advance therapeutic interventions for the management of bone diseases. This review discusses the current state of vascular-skeletal tissue interactions focusing on osteoblast and hematopoietic stem cells interaction with ECs.
Collapse
Affiliation(s)
- Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali Kusumbe
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
22
|
Gado M, Baschant U, Hofbauer LC, Henneicke H. Bad to the Bone: The Effects of Therapeutic Glucocorticoids on Osteoblasts and Osteocytes. Front Endocrinol (Lausanne) 2022; 13:835720. [PMID: 35432217 PMCID: PMC9008133 DOI: 10.3389/fendo.2022.835720] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the continued development of specialized immunosuppressive therapies in the form of monoclonal antibodies, glucocorticoids remain a mainstay in the treatment of rheumatological and auto-inflammatory disorders. Therapeutic glucocorticoids are unmatched in the breadth of their immunosuppressive properties and deliver their anti-inflammatory effects at unparalleled speed. However, long-term exposure to therapeutic doses of glucocorticoids decreases bone mass and increases the risk of fractures - particularly in the spine - thus limiting their clinical use. Due to the abundant expression of glucocorticoid receptors across all skeletal cell populations and their respective progenitors, therapeutic glucocorticoids affect skeletal quality through a plethora of cellular targets and molecular mechanisms. However, recent evidence from rodent studies, supported by clinical data, highlights the considerable role of cells of the osteoblast lineage in the pathogenesis of glucocorticoid-induced osteoporosis: it is now appreciated that cells of the osteoblast lineage are key targets of therapeutic glucocorticoids and have an outsized role in mediating their undesirable skeletal effects. As part of this article, we review the molecular mechanisms underpinning the detrimental effects of supraphysiological levels of glucocorticoids on cells of the osteoblast lineage including osteocytes and highlight the clinical implications of recent discoveries in the field.
Collapse
Affiliation(s)
- Manuel Gado
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C. Hofbauer
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Holger Henneicke
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Holger Henneicke,
| |
Collapse
|
23
|
Wang Q, Zhou J, Wang X, Xu Y, Liang Z, Gu X, He C. Coupling induction of osteogenesis and type H vessels by pulsed electromagnetic fields in ovariectomy-induced osteoporosis in mice. Bone 2022; 154:116211. [PMID: 34560308 DOI: 10.1016/j.bone.2021.116211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
The growth of blood vessels and osteogenesis are coupled in bone tissue. A specialized subset of CD31hiEndomucinhi (CD31hiEmcnhi) vascular endothelium in bone has been identified to positively regulate bone formation. Pulsed electromagnetic field (PEMF) can promote the facture healing and reverse the loss of bone mass. However, the underlying mechanisms mediating in the positive effects of PEMF on bone mass accrual remain unclear. In the ovariectomized (OVX) osteoporotic mouse model, PEMF with specific parameters was administrated after 12 weeks of surgery and continued for 8 weeks. μCT analysis, quantitative PCR and Elisa assays were used to assess the PEMF-induced the osteogenesis, while immunostaining and flow cytometry were used to evaluate the abundance of CD31hiEmcnhi endothelium in the metaphysis near the growth plate. Administration of PEMF substantially countered OVX-induced bone loss as shown by greater trabecular bone, higher expression of Osterix, PDGFB and Col-1a1 transcripts, and modulation of bone anabolic and catabolic activity. The PEMF-induced osteogenesis was coupled by the expansion of CD31hiEmcnhi endothelium as demonstrated by CD31 and Endomucin double-positive immunostaining and flow cytometry. Concurrently, the higher level of HIF-α was found in PEMF-treated mice than in vehicle controls. Notably, inhibition of HIF-1α considerably reduced PEMF-induced osteogenesis, and led to a remarkable decrease of CD31hiEmcnhi vessels in the PEMF-treated OVX mice. The present study demonstrated the PEMF-induced coupling promotion of osteogenesis and CD31hiEmcnhi endothelial cells in a mouse model of postmenopausal osteoporosis. This coupling effect might be mediated in HIF-1α signaling in CD31hiEmcnhi endothelium. These findings open up new directions of al that might enable therapeutic improvement of osteogenesis in patients with osteoporosis.
Collapse
Affiliation(s)
- Qian Wang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Jun Zhou
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiangxiu Wang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Yang Xu
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Zhejun Liang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Xintong Gu
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Chengqi He
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
24
|
The endothelium-bone axis in development, homeostasis and bone and joint disease. Nat Rev Rheumatol 2021; 17:608-620. [PMID: 34480164 DOI: 10.1038/s41584-021-00682-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 01/20/2023]
Abstract
Blood vessels form a versatile transport network that is best known for its critical roles in processes such as tissue oxygenation, metabolism and immune surveillance. The vasculature also provides local, often organ-specific, molecular signals that control the behaviour of other cell types in their vicinity during development, homeostasis and regeneration, and also in disease processes. In the skeletal system, the local vasculature is actively involved in both bone formation and resorption. In addition, blood vessels participate in inflammatory processes and contribute to the pathogenesis of diseases that affect the joints, such as rheumatoid arthritis and osteoarthritis. This Review summarizes the current understanding of the architecture, angiogenic growth and functional properties of the bone vasculature. The effects of ageing and pathological conditions, including arthritis and osteoporosis, are also discussed.
Collapse
|
25
|
Duan L, Li Y, Hu J, Ma Q, Yu T, Zhang C, Luo F, Xu J, Dou C. Light rare earth elements hinder bone development via inhibiting type H vessels formation in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 218:112275. [PMID: 33962277 DOI: 10.1016/j.ecoenv.2021.112275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/14/2021] [Accepted: 04/18/2021] [Indexed: 06/12/2023]
Abstract
Light rare earth elements (LREEs) are widely used in medical, industrial, and agricultural fields. Wide application of light rare earth and exposure to these elements in human society leads to increasing accumulation of LREE in human skeletal system. However, the effects of LREEs on human bone health is not clear. In this study, we found that LREE reduced CD31highEmcnhigh endothelial cell mediated type H vessels formation at the metaphyseal sites, resulting in reduced bone mass and low bone quality in mouse bone development. To explore the underlying mechanism, we induced bone marrow macrophages (BMMs) to preosteoclasts (pOCs) with exposure of LREE (Pr3+, Nd3+, Sm3+). The cytotoxicity of LREE was evaluated by CCK-8. Platelet-derived growth factor (PDGF-BB) is the cytokine secreted by pOCs that most responsible for inducing Type H vessel formation. We used ELISA kit to determine the PDGF-BB level in pOC supernatant, and mouse serum finding that the PDGF-BB level was reduced by LREEs treatment. Then we tested the ability of migration and tube formation of HUVECs using condition medium from pOCs. The migration and tube formation ability of HUVECs were both suppressed with LREEs pretreatment. We concluded that LREEs hinder mouse bone development by suppressing type H vessels associated bone formation. DATA AND MATERIALS AVAILABILITY: All data generated or analyzed during this study are included in this article. Please contact the corresponding author for unique material requests. Some material used in the reported research may require requests to collaborators and agreements with both commercial and non-profit institutions, as specified in the paper. Requests are reviewed by Third Military Medical University to verify whether the request is subject to any intellectual property or confidentiality obligations. Any material that can be shared will be released via a Material Transfer Agreement.
Collapse
Affiliation(s)
- Lianli Duan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Yang Li
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Junxian Hu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Qinyu Ma
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Tao Yu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Chengmin Zhang
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China.
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
26
|
Liu X, Chai Y, Liu G, Su W, Guo Q, Lv X, Gao P, Yu B, Ferbeyre G, Cao X, Wan M. Osteoclasts protect bone blood vessels against senescence through the angiogenin/plexin-B2 axis. Nat Commun 2021; 12:1832. [PMID: 33758201 PMCID: PMC7987975 DOI: 10.1038/s41467-021-22131-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/27/2021] [Indexed: 01/31/2023] Open
Abstract
Synthetic glucocorticoids (GCs), one of the most effective treatments for chronic inflammatory and autoimmune conditions in children, have adverse effects on the growing skeleton. GCs inhibit angiogenesis in growing bone, but the underlying mechanisms remain unclear. Here, we show that GC treatment in young mice induces vascular endothelial cell senescence in metaphysis of long bone, and that inhibition of endothelial cell senescence improves GC-impaired bone angiogenesis with coupled osteogenesis. We identify angiogenin (ANG), a ribonuclease with pro-angiogenic activity, secreted by osteoclasts as a key factor for protecting the neighboring vascular cells against senescence. ANG maintains the proliferative activity of endothelial cells through plexin-B2 (PLXNB2)-mediated transcription of ribosomal RNA (rRNA). GC treatment inhibits ANG production by suppressing osteoclast formation in metaphysis, resulting in impaired endothelial cell rRNA transcription and subsequent cellular senescence. These findings reveal the role of metaphyseal blood vessel senescence in mediating the action of GCs on growing skeleton and establish the ANG/PLXNB2 axis as a molecular basis for the osteoclast-vascular interplay in skeletal angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Bone Development/drug effects
- Cell Proliferation/drug effects
- Cellular Senescence/drug effects
- Cellular Senescence/genetics
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Glucocorticoids/pharmacology
- Human Umbilical Vein Endothelial Cells
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Methylprednisolone/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neovascularization, Pathologic
- Neovascularization, Physiologic/drug effects
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Osteoclasts/drug effects
- Osteoclasts/enzymology
- Osteoclasts/metabolism
- Osteogenesis/drug effects
- RNA, Ribosomal/biosynthesis
- RNA, Small Interfering
- Recombinant Proteins
- Ribonuclease, Pancreatic/genetics
- Ribonuclease, Pancreatic/metabolism
- Ribonuclease, Pancreatic/pharmacology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Tomography Scanners, X-Ray Computed
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Chai
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guanqiao Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weiping Su
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiao Lv
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peisong Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
28
|
Stucker S, De Angelis J, Kusumbe AP. Heterogeneity and Dynamics of Vasculature in the Endocrine System During Aging and Disease. Front Physiol 2021; 12:624928. [PMID: 33767633 PMCID: PMC7987104 DOI: 10.3389/fphys.2021.624928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
The endocrine system consists of several highly vascularized glands that produce and secrete hormones to maintain body homeostasis and regulate a range of bodily functions and processes, including growth, metabolism and development. The dense and highly vascularized capillary network functions as the main transport system for hormones and regulatory factors to enable efficient endocrine function. The specialized capillary types provide the microenvironments to support stem and progenitor cells, by regulating their survival, maintenance and differentiation. Moreover, the vasculature interacts with endocrine cells supporting their endocrine function. However, the structure and niche function of vasculature in endocrine tissues remain poorly understood. Aging and endocrine disorders are associated with vascular perturbations. Understanding the cellular and molecular cues driving the disease, and age-related vascular perturbations hold potential to manage or even treat endocrine disorders and comorbidities associated with aging. This review aims to describe the structure and niche functions of the vasculature in various endocrine glands and define the vascular changes in aging and endocrine disorders.
Collapse
Affiliation(s)
| | | | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Shangguan Y, Wu Z, Xie X, Zhou S, He H, Xiao H, Liu L, Zhu J, Chen H, Han H, Wang H, Chen L. Low-activity programming of the PDGFRβ/FAK pathway mediates H-type vessel dysplasia and high susceptibility to osteoporosis in female offspring rats after prenatal dexamethasone exposure. Biochem Pharmacol 2021; 185:114414. [PMID: 33434537 DOI: 10.1016/j.bcp.2021.114414] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/13/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022]
Abstract
Dexamethasone is a common synthetic glucocorticoid drug that can promote foetal lung maturity. An increasing number of studies have shown that prenatal dexamethasone exposure (PDE) can cause a variety of short-term and long-term hazards to offspring, including bone development toxicity. H-type vessels are a newly discovered subtype of blood vessels associated with promoted bone formation and maintenance of bone mass. In this study, we aimed to explore whether H-type blood vessels are involved in PDE-induced long bone development toxicity in offspring and its mechanism. In vivo, we injected dexamethasone (0.2 mg/kg.d) subcutaneously at gestational days 9-20 and observed the H-type vessel abundance and bone mass at different time points in the offspring rats. In vitro, we investigated the effect of dexamethasone (0, 20, 100, and 500 nM) on the tube formation function of rat bone marrow-derived endothelial progenitor cells (EPCs) and explored its mechanism. Our results showed that the adult PDE female offspring rats were susceptible to osteoporosis. In addition, PDE inhibited bone mass, H-type vessel formation and the expression of bone platelet-derived growth factor receptor β (PDGFRβ)/focal adhesion kinase (FAK) pathway-related genes in antenatal and postnatal female offspring. Moreover, PDE promoted the expression of bone glucocorticoid receptor (GR), CCAAT and enhancer binding protein α (C/EBPα) and miR-34c in female foetuses. Dexamethasone suppressed the tube formation of rat bone marrow-derived EPCs and the activity of the PDGFRβ/FAK pathway, which was mediated by GR/C/EBPα/miR-34c signalling activation. In summary, PDE can cause H-type vessel dysplasia and high susceptibility to osteoporosis in female offspring, and its mechanism is related to the low-activity programming of the PDGFRβ/FAK pathway induced by GR/C/EBPα/miR-34c signalling activation. This study enhances the understanding of the molecular mechanism of dexamethasone-induced bone development toxicity and provides new insights for exploring the early intervention and therapeutic targets of foetal-derived osteoporosis.
Collapse
Affiliation(s)
- Yangfan Shangguan
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Zhixin Wu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Xingkui Xie
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Siqi Zhou
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hangyuan He
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hao Xiao
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Liang Liu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Jiayong Zhu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Haitao Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Han
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
30
|
Martin CS, Cooper MS, Hardy RS. Endogenous Glucocorticoid Metabolism in Bone: Friend or Foe. Front Endocrinol (Lausanne) 2021; 12:733611. [PMID: 34512556 PMCID: PMC8429897 DOI: 10.3389/fendo.2021.733611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023] Open
Abstract
The role of tissue specific metabolism of endogenous glucocorticoids (GCs) in the pathogenesis of human disease has been a field of intense interest over the last 20 years, fuelling clinical trials of metabolism inhibitors in the treatment of an array of metabolic diseases. Localised pre-receptor metabolism of endogenous and therapeutic GCs by the 11β-hydroxysteroid dehydrogenase (11β-HSD) enzymes (which interconvert endogenous GCs between their inactive and active forms) are increasingly recognised as being critical in mediating both their positive and negative actions on bone homeostasis. In this review we explore the roles of endogenous and therapeutic GC metabolism by the 11β-HSD enzymes in the context of bone metabolism and bone cell function, and consider future strategies aimed at modulating this system in order to manage and treat various bone diseases.
Collapse
Affiliation(s)
- Claire S. Martin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Mark S. Cooper
- Australian and New Zealand Army Corps (ANZAC) Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Rowan S. Hardy
- Arthritis Research United Kingdom (UK) Career Development Fellow, University of Birmingham, Birmingham, United Kingdom
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Rowan S. Hardy,
| |
Collapse
|
31
|
Chai Y, Su J, Hong W, Zhu R, Cheng C, Wang L, Zhang X, Yu B. Antenatal Corticosteroid Therapy Attenuates Angiogenesis Through Inhibiting Osteoclastogenesis in Young Mice. Front Cell Dev Biol 2020; 8:601188. [PMID: 33384997 PMCID: PMC7769874 DOI: 10.3389/fcell.2020.601188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/09/2020] [Indexed: 11/13/2022] Open
Abstract
Antenatal corticosteroid therapy (ACT) has been shown to reduce morbidity and mortality rates in preterm delivery, but the fetus is more likely to face the risk of low bone mineralization and low fetal linear growth. However, the mechanism of ACT inducing low bone mineralization remains largely unknown. Pre-osteoclasts, which play an important role in angiogenesis and osteogenesis, are specifically regulating type H vessels (CD31hiEmcnhi) and vessel formation by secreting platelet-derived growth factor-BB (PDGF-BB). We find that the number of pre-osteoclasts and POC-secreted PDGF-BB is dramatically decreased in ACT mice, contributing to the reduction in type H vessels and bone mineralization during the mouse offspring. Quantitative analyses of micro-computed tomography show that the ACT mice have a significant reduction in the mass of trabecular bone relative to the control group. Mononuclear pre-osteoclasts in trabecular bone decreased in ACT mice, which leads to the amount of PDGF-BB reduced and attenuates type H vessel formation. After sorting the Rank+ osteoclast precursors using flow cytometry, we show that the enhancer of zeste homolog 2 (Ezh2) expression is decreased in Rank+ osteoclast precursors in ACT mice. Consistent with the flow data, by using small molecule Ezh2 inhibitor GSK126, we prove that Ezh2 is required for osteoclast differentiation. Downregulating the expression of Ezh2 in osteoclast precursors would reduce PDGF-BB production. Conditioned medium from osteoclast precursor cultures treated with GSK126 inhibited endothelial tube formation, whereas conditioned medium from vehicle group stimulated endothelial tube formation. These results indicate Ezh2 expression of osteoclast precursors is suppressed after ACT, which reduced the pre-osteoclast number and PDGF-BB secretion, thus inhibiting type H vessel formation and ACT-associated low bone mineralization.
Collapse
Affiliation(s)
- Yu Chai
- Division of Orthopaedic Surgery, 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
| | - Jianwen Su
- Division of Orthopaedic Surgery, 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
| | - Weisheng Hong
- Division of Orthopaedic Surgery, 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
| | - Runjiu Zhu
- Division of Orthopaedic Surgery, 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
| | - Caiyu Cheng
- Division of Orthopaedic Surgery, 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
| | - Lei Wang
- Division of Orthopaedic Surgery, 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
| | - Xianrong Zhang
- Division of Orthopaedic Surgery, 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
- Division of Orthopaedic Surgery, 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
|
32
|
Stucker S, Chen J, Watt FE, Kusumbe AP. Bone Angiogenesis and Vascular Niche Remodeling in Stress, Aging, and Diseases. Front Cell Dev Biol 2020; 8:602269. [PMID: 33324652 PMCID: PMC7726257 DOI: 10.3389/fcell.2020.602269] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/05/2020] [Indexed: 02/05/2023] Open
Abstract
The bone marrow (BM) vascular niche microenvironments harbor stem and progenitor cells of various lineages. Bone angiogenesis is distinct and involves tissue-specific signals. The nurturing vascular niches in the BM are complex and heterogenous consisting of distinct vascular and perivascular cell types that provide crucial signals for the maintenance of stem and progenitor cells. Growing evidence suggests that the BM niche is highly sensitive to stress. Aging, inflammation and other stress factors induce changes in BM niche cells and their crosstalk with tissue cells leading to perturbed hematopoiesis, bone angiogenesis and bone formation. Defining vascular niche remodeling under stress conditions will improve our understanding of the BM vascular niche and its role in homeostasis and disease. Therefore, this review provides an overview of the current understanding of the BM vascular niches for hematopoietic stem cells and their malfunction during aging, bone loss diseases, arthritis and metastasis.
Collapse
Affiliation(s)
- Sina Stucker
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Junyu Chen
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fiona E. Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Anjali P. Kusumbe
- Tissue and Tumor Microenvironments Group, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
33
|
Li X, Jin L, Tan Y. Different roles of matrix metalloproteinase 2 in osteolysis of skeletal dysplasia and bone metastasis (Review). Mol Med Rep 2020; 23:70. [PMID: 33236155 PMCID: PMC7716421 DOI: 10.3892/mmr.2020.11708] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/02/2020] [Indexed: 01/18/2023] Open
Abstract
Matrix metalloproteinase 2 (MMP2) is a well-characterized protein that is indispensable for extracellular matrix remodeling and other pathological processes, such as tumor progression and skeletal dysplasia. Excessive activation of MMP2 promotes osteolytic metastasis and bone destruction in late-stage cancers, while its loss-of-function mutations result in the decreased bone mineralization and generalized osteolysis occurring progressively in skeletal developmental disorders, particularly in multicentric osteolysis, nodulosis and arthropathy (MONA). Either upregulation or downregulation of MMP2 activity can result in the same osteolytic effects. Thus, different functions of MMP2 have been recently identified that could explain this observation. While MMP2 can degrade bone matrix, facilitate osteoclastogenesis and amplify various signaling pathways that enhance osteolysis in bone metastasis, its role in maintaining the number of bone cells, supporting osteocytic canalicular network formation and suppressing leptin-mediated inhibition of bone formation has been implicated in osteolytic disorders caused by MMP2 deficiency. Furthermore, the proangiogenic activity of MMP2 is one of the potential mechanisms that are associated with both pathological situations. In the present article, the latest research on MMP2 in bone homeostasis is reviewed and the mechanisms underlying the role of this protein in skeletal metastasis and developmental osteolysis are discussed.
Collapse
Affiliation(s)
- Xiumao Li
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Libin Jin
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yanbin Tan
- Department of Orthopedics, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
34
|
Chen J, Hendriks M, Chatzis A, Ramasamy SK, Kusumbe AP. Bone Vasculature and Bone Marrow Vascular Niches in Health and Disease. J Bone Miner Res 2020; 35:2103-2120. [PMID: 32845550 DOI: 10.1002/jbmr.4171] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Junyu Chen
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexandros Chatzis
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
35
|
Liu W, Guo S, Tang Z, Wei X, Gao P, Wang N, Li X, Guo Z. Magnesium promotes bone formation and angiogenesis by enhancing MC3T3-E1 secretion of PDGF-BB. Biochem Biophys Res Commun 2020; 528:664-670. [DOI: 10.1016/j.bbrc.2020.05.113] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/16/2020] [Indexed: 01/24/2023]
|
36
|
Peng Y, Lv S, Li Y, Zhu J, Chen S, Zhen G, Cao X, Wu S, Crane JL. Glucocorticoids Disrupt Skeletal Angiogenesis Through Transrepression of NF-κB-Mediated Preosteoclast Pdgfb Transcription in Young Mice. J Bone Miner Res 2020; 35:1188-1202. [PMID: 32078184 PMCID: PMC8554682 DOI: 10.1002/jbmr.3987] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022]
Abstract
In the growing skeleton, angiogenesis is intimately coupled with osteogenesis. Chronic, high doses of glucocorticoids (GCs) are associated with decreased bone vasculature and induce osteoporosis and growth failure. The mechanism of GC-suppression of angiogenesis and relationship to osteoporosis and growth retardation remains largely unknown. Type H vessels, which are regulated by preosteoclast (POC) platelet-derived growth factor-BB (PDGF-BB), are specifically coupled with bone formation and development. We determined the effect of GCs on POC synthesis of PDGF-BB in relation to type H vessel formation, bone mass, and bone growth in the distal femur of 2-week-old young mice receiving prednisolone or vehicle for 2, 4, or 6 weeks. After 2 weeks of prednisolone, the number of POCs were unchanged while POC synthesis of PDGF-BB was reduced. Longer treatment with prednisolone reduced POCs numbers and PDGF-BB. These changes were associated with a reduction in type H vessels, bone formation rate, bone mass, and bone length at each time point. In vitro, excessive concentrations of prednisolone (10-6 M) resulted in decreased PDGF-BB concentration and POC numbers. Conditioned medium from POC cultures treated with control concentration of prednisolone (10-7 M) or recombinant PDGF-BB stimulated endothelial tube formation, whereas conditioned medium from control concentration of prednisolone-treated POC cultures neutralized by PDGF-BB antibody or excessive prednisolone inhibited endothelial tube formation. Administration of excessive prednisolone attenuated the P65 subunit of nuclear factor kappa B (NF-κB) binding to the Pdgfb promoter, resulting in lower Pdgfb transcription. Co-treatment with excessive prednisolone and the glucocorticoid receptor (GR) antagonist (RU486), GR siRNA, or TNFα rescued NF-κB binding to the Pdgfb promoter and endothelial tube formation. These results indicate that PDGF-BB synthesis in POCs is suppressed by GCs through transrepression of GR/NF-κB, thus inhibiting type H vessel formation and associated osteoporosis and growth failure. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yi Peng
- Department of Orthopedic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China.,Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shan Lv
- Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yusheng Li
- Department of Orthopedic Surgery, Xiangya Hospital of Central South University, Changsha, China
| | - Jianxi Zhu
- Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Shijie Chen
- Department of Orthopedic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xu Cao
- Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Song Wu
- Department of Orthopedic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Janet L Crane
- Department of Orthopedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Sun X, Li X, Qi H, Hou X, Zhao J, Yuan X, Ma X. MiR-21 nanocapsules promote early bone repair of osteoporotic fractures by stimulating the osteogenic differentiation of bone marrow mesenchymal stem cells. J Orthop Translat 2020; 24:76-87. [PMID: 32695607 PMCID: PMC7349941 DOI: 10.1016/j.jot.2020.04.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/10/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Objective The healing of osteoporotic fractures in the elderly patients is a difficult clinical problem. Currently, based on the internal fixation of fractures, the available drug treatments mainly focus on either inhibiting osteoclast function, such as bisphosphonate, calcitonin, oestrogen or promoting osteogenesis, such as parathyroid hormones. However, the availability of current antiosteoporotic drugs in promoting osteoporotic fracture healing is limited. The objective of the present study was to investigate the ability of the MiR-21/nanocapsule to enhance the early bone repair of osteoporotic fractures. Methods Based on the presence of matrix metalloproteinases that are overexpressed at the fracture site, we designed the matrix metalloproteinase–sensitive nanocapsules which were formed by in situ free radical polymerisation on the surface of MiR-21 with 2-(methacryloyloxy) ethyl phosphorylcholine and the bisacryloylated VPLGVRTK peptide. The MiR-21/nanocapsule [n (miR-21)] and O-carboxymethyl chitosan (CMCS) were mixed until they formed a gel-like material [CMCS/n (miR-21)] with good fluidity and injectability. Thirty elderly Sprague Dawley (SD) rats (female, 14-month-old, 380 ± 10 g) were subjected to bilateral removal of the ovaries (ovariectomised). All rats were subjected to bilateral bone defects (2 mm diameter) of the proximal tibia and randomly divided into three groups (groups A, B, and C): separately injected with CMCS/n (miR-21), CMCS/n (NC-miR), and saline. Micro-computed tomography (CT) imaging was performed to evaluate newly formed bone volume and connectivity. Nondecalcified histology and toluidine blue staining were performed to measure the effects of CMCS/n (miR-21) on bone repair. In vitro, the effect of n (miR-21) on osteogenic differentiation to bone marrow mesenchymal stem cells (BMSCs) which derived from the ovariectomised rat model was observed. Results The morphology of n (miR-21) was a regular spherical nanocapsule with a uniform small size (25–35 nm). The results confirmed that n (miR-21) could be efficiently phagocytosed by BMSCs and released in the cytoplasm to promote osteogenesis. The expression level of alkaline phosphatase and Runt-related transcription factor 2 mRNA in the n (miR-21) group was higher than that in the n (NC-miR) group. Animal experiments proved that CMCS/n (miR-21) produced better bone repair compared with the CMCS/n (NC-miR) group in the early stages of fracture healing at 4 weeks. In the late stage of fracture healing (8 weeks), micro-CT quantitative analysis showed that the new bone trabeculae in the CMCS/n (miR-21) group has decreased compared with the CMCS/n (NC-miR) group. In the CMCS/n (miR-21) group, the new cancellous bone had been absorbed, and the process of bone healing was almost completed. In contrast, the new bone in the CMCS/n (NC-miR) and the control groups was still in the healing process. Conclusion The cytological tests confirmed that n (miR-21) can promote osteogenic differentiation of BMSCs derived from the osteoporosis rat model. Furthermore, the results of animal tests demonstrated that local injection of CMCS/n (miR-21) promoted the early healing of osteoporotic bone defects. Consequently CMCS/n (miR-21) promoted the bone repair process to enter the moulding phase earlier. The translational potential of this article CMCS/n (miR-21) can be widely applied to elderly patients with osteoporotic fractures. This method can help patients with osteoporotic fractures recover earlier and avoid serious complications. It provides a potential approach for the clinical treatment of osteoporotic fractures in the elderly.
Collapse
Affiliation(s)
- Xiaolei Sun
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China.,Department of Orthopaedics, Tianjin Hospital, Tianjin, 300211, China
| | - Xueping Li
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Hongzhao Qi
- Institute for Translational Medicine, Qingdao University, Qingdao, 266021, China
| | - Xin Hou
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Jin Zhao
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Xubo Yuan
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Xinlong Ma
- Department of Orthopaedics, Tianjin Hospital, Tianjin, 300211, China
| |
Collapse
|
38
|
Zhao Y, Xie L. Unique bone marrow blood vessels couple angiogenesis and osteogenesis in bone homeostasis and diseases. Ann N Y Acad Sci 2020; 1474:5-14. [PMID: 32242943 DOI: 10.1111/nyas.14348] [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: 01/10/2020] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 02/05/2023]
Abstract
Blood vessels serve as a versatile transport system and play crucial roles in organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular stem cells or osteoprogenitor cells and thereby regulate bone formation. Recent studies reported that a specialized capillary subtype, termed type H vessels, is shown to couple angiogenesis and osteogenesis in rodents and humans. They can be distinguished by specific cell surface markers, as the endothelial cells in the metaphysis and endosteum highly express the junctional protein CD31 and the sialoglycoprotein endomucin. Here, we provide an overview of the role of type H vessels in bone homeostasis and summarize their linkage with various cytokines to control bone cell behavior and bone formation. We also discuss the potential clinical application for bone disorders by targeting these specific vessels according to their physiological and pathobiological settings.
Collapse
Affiliation(s)
- Yifan Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Peng Y, Wu S, Li Y, Crane JL. Type H blood vessels in bone modeling and remodeling. Theranostics 2020; 10:426-436. [PMID: 31903130 PMCID: PMC6929606 DOI: 10.7150/thno.34126] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/12/2019] [Indexed: 12/29/2022] Open
Abstract
In the mammalian skeletal system, osteogenesis and angiogenesis are intimately linked during bone growth and regeneration in bone modeling and during bone homeostasis in bone remodeling. Recent studies have expanded our knowledge about the molecular and cellular mechanisms responsible for coupling angiogenesis and bone formation. Type H vessels, termed such because of high expression of Endomucin (Emcn) and CD31, have recently been identified and have the ability to induce bone formation. Factors including platelet-derived growth factor type BB (PDGF-BB), slit guidance ligand 3 (SLIT3), hypoxia-inducible factor 1-alpha (HIF-1α), Notch, and vascular endothelial growth factor (VEGF) are involved in the coupling of angiogenesis and osteogenesis. This review summarizes the current understanding of signaling pathways that regulate type H vessels and how type H vessels modulate osteogenesis. Further studies dissecting the regulation and function of type H vessels will provide new insights into the role of bone vasculature in the metabolism of the skeleton. We also discuss considerations for therapeutic approaches targeting type H vessels to promote fracture healing, prevent pathological bone loss, osteonecrosis, osteoarthritis, and bone metastases.
Collapse
Affiliation(s)
- Yi Peng
- Department of Orthopedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Song Wu
- Department of Orthopedic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Yusheng Li
- Department of Orthopedic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 41000, China
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Ahmad M, Hachemi Y, Paxian K, Mengele F, Koenen M, Tuckermann J. A Jack of All Trades: Impact of Glucocorticoids on Cellular Cross-Talk in Osteoimmunology. Front Immunol 2019; 10:2460. [PMID: 31681333 PMCID: PMC6811614 DOI: 10.3389/fimmu.2019.02460] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids (GCs) are known to have a strong impact on the immune system, metabolism, and bone homeostasis. While these functions have been long investigated separately in immunology, metabolism, or bone biology, the understanding of how GCs regulate the cellular cross-talk between innate immune cells, mesenchymal cells, and other stromal cells has been garnering attention rather recently. Here we review the recent findings of GC action in osteoporosis, inflammatory bone diseases (rheumatoid and osteoarthritis), and bone regeneration during fracture healing. We focus on studies of pre-clinical animal models that enable dissecting the role of GC actions in innate immune cells, stromal cells, and bone cells using conditional and function-selective mutant mice of the GC receptor (GR), or mice with impaired GC signaling. Importantly, GCs do not only directly affect cellular functions, but also influence the cross-talk between mesenchymal and immune cells, contributing to both beneficial and adverse effects of GCs. Given the importance of endogenous GCs as stress hormones and the wide prescription of pharmaceutical GCs, an improved understanding of GC action is decisive for tackling inflammatory bone diseases, osteoporosis, and aging.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Yasmine Hachemi
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Kevin Paxian
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Florian Mengele
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Praxisklinik für Orthopädie, Unfall- und Neurochirurgie Prof. Bischoff/ Dr. Spies/ Dr. Mengele, Neu-Ulm, Germany
| | - Mascha Koenen
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| |
Collapse
|
41
|
Lin HT, Chen SK, Guo JW, Su IC, Huang CJ, Chien CC, Chang CJ. Dynamic expression of SMAD3 is critical in osteoblast differentiation of PDMCs. Int J Mol Med 2018; 43:1085-1093. [PMID: 30483761 DOI: 10.3892/ijmm.2018.4001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/19/2018] [Indexed: 11/06/2022] Open
Abstract
Human pluripotent stem cells have the potential assist in the identification of genes involved in mammalian development. The human placenta is considered a repository of stem cells, termed placenta‑derived multipotent cells (PDMCs), which are able to differentiate into cells with an osteoblastic phenotype. This plasticity of PDMCs maybe applied clinically to the understanding of osteogenesis and osteoporosis. In the presentstudy, osteoblasts were generated by culturing PDMCs in osteogenic medium. Reverse transcription quantitative polymerase chain reactionand the degree of osteoblast calcification were used to evaluate the efficacy of osteogenesis. The results suggestedthat the expression of mothers against decapentaplegic homolog 3 (SMAD3) increased in the initial stages of osteogenic differentiation but decreased in the later stages. However, osteogenesis was inhibitedwhen the PDMCs overexpressed SMAD3 throughout the differentiation period. In addition, the rate of osteogenic differentiation was decreased when SMAD3 signaling was impaired. In conclusion, SMAD3 serves an important role in osteoblast differentiation and bone formation in a time‑dependent manner. The data from the present study indicate that arapid increase in SMAD3 expression is crucial for osteogenesis and suggest a role for PDMCs in the treatment of patients with osteoporosis.
Collapse
Affiliation(s)
- Hsi-Ting Lin
- Department of Orthopedics, Cathay General Hospital, Taipei 10630, Taiwan, R.O.C
| | - Shao-Kuan Chen
- Department of Urology, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Jiun-Wen Guo
- Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - I-Chang Su
- Department of Neurosurgery, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Chi-Jung Huang
- Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Chih-Cheng Chien
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| | - Chih-Ju Chang
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan, R.O.C
| |
Collapse
|