1
|
Liu G, Wang Z, Li X, Yu P, Ji W, Wu L, Jiang H, Xu S, Liu J. Protective effects of Gumibao recipe on glucocorticoid-included bone microcirculatory endothelial cell injury and the underlying mechanism. Int Immunopharmacol 2024; 142:112989. [PMID: 39217879 DOI: 10.1016/j.intimp.2024.112989] [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: 04/08/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To investigate the protective effects of Gumibao recipe on glucocorticoid-included bone microcirculatory endothelial cell (BMEC) injury, and elucidate the possible underlying mechanism. METHODS BMECs were treated with different concentrations of hydrocortisone at different time points, and the viability as well as migration of BMECs were evaluated; furthermore, the release of LDH, levels of VEGF, PAI-1, t-PA, and the content of NO by BMECs have been evaluated by commercially available kits; moreover, the expressions of eNOS, p-PI3K, p-Akt and p-mTOR in BMECs were examined by WB methods. Next, hydrocortisone treated BMECs were co-treated with Gumibao recipe, and the viability, migration and autophagy of BMECs were evaluated. RESULTS 0.2 mg/ml and 0.3 mg/ml hydrocortisone significantly decreased viability and migration ability of BMECs, and also impeded the endothelial function of BMECs by decreasing the levels of VEGF, t-PA, the content of NO, and increasing the level of PAI-1. Gumibao medicated serum markedly increased the viability and migration of BMECs, and also increased the levels of VEGF, t-PA, the content of NO, meanwhile decreased the level of PAI-1 in 0.3 mg/ml hydrocortisone treated BMECs; moreover, glucocorticoids inhibited the autophagy of BMECs, and Gumibao recipe significantly increased the autophagy of BMECs; meanwhile, autophagy inhibitor 3-MA partially blocked the protective effects of Gumibao recipe. Finally, gumibao recipe partially abrogated the inhibitory effects of hydrocortisone on the activation of PI3K/Akt/mTOR singling, and these effects were further counteracted by PI3K and mTOR inhibitor NVP-BEZ235. CONCLUSIONS We reported for the first time the protective effects of Gumibao recipe on glucocorticoid-included BMECs injury, and the possible underlying mechanism may be regulating the autophagy of BMECs via PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Guanhong Liu
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Zhiqiang Wang
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Xiaochun Li
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Pengfei Yu
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Wanbo Ji
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Liming Wu
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Hong Jiang
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Suliang Xu
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China
| | - Jintao Liu
- Orthopedics and Traumatology Department, Suzhou Hospital of Traditional Chinese Medicine, Suzhou 215000, China.
| |
Collapse
|
2
|
Meng C, Qi B, Luo H, Tang Z, Ren J, Shi H, Li C, Xu Y. Exploring the genetic association between immune cells and susceptibility to osteonecrosis using large-scale population data. Heliyon 2024; 10:e34547. [PMID: 39130408 PMCID: PMC11315082 DOI: 10.1016/j.heliyon.2024.e34547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 08/13/2024] Open
Abstract
Objectives Research shows a close association between aberrant immune reactions in osteonecrotic tissues and immune cell infiltration. However, due to limitations in sample size and dataset comprehensiveness, the causal relationship between them is not fully established. This study aims to determine whether there is a causal relationship using a larger and more diverse dataset. Methods We conducted a comprehensive Mendelian Randomization (MR) analysis to investigate the causal relationship between immune cell characteristics and osteonecrosis. Utilizing publicly available genetic data, we explored the causal relationships between 731 immune cell features and 604 cases from the FinnGen Finnish database, as well as 257 cases from the UK Biobank database with osteonecrosis data. The inverse-variance weighted (IVW) method was used for the primary analysis, and we employed sensitivity analyses to assess the robustness of the main results. In addition, considering data from the two databases used in this study, a meta-analysis was conducted on the significant immune cells associated with osteonecrosis (FDR <0.05). Results our findings suggested that specific immune cell signatures, such as CD20- % lymphocytes, CD62L-monocytes, and CD33br HLA DR+ CD14-cells were associated with increased odds of osteonecrosis. In contrast, EM CD4+ activated cells and DP (CD4+ CD8+) T cells were associated with decreased odds. Notably, osteonecrosis was associated with a potential decrease in CD45 on immature MDSC cell content. Conclusion From a genetic perspective, we demonstrated a close association between immune cells and osteonecrosis. These findings significantly enhance our understanding of the interplay between immune cell infiltration and the risk of osteonecrosis, contributing to the potential design of therapeutic strategies from an immunological standpoint.
Collapse
Affiliation(s)
- Chen Meng
- Graduate School of Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Baochuang Qi
- Graduate School of Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Huan Luo
- Graduate School of Kunming Medical University, Kunming, Yunnan, China
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Zhifang Tang
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Junxiao Ren
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Hongxin Shi
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Chuan Li
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yongqing Xu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| |
Collapse
|
3
|
Fang L, Zhang G, Wu Y, Li H, Li Z, Yu B, Wang B, Zhou L. Fibroblast growth factor 23 inhibition attenuates steroid-induced osteonecrosis of the femoral head through pyroptosis. Sci Rep 2024; 14:16270. [PMID: 39009650 PMCID: PMC11251279 DOI: 10.1038/s41598-024-66799-z] [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: 01/25/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is the predominant cause of non-traumatic osteonecrosis of the femoral head (ONFH). Impaired blood supply and reduced osteogenic activity of the femoral head are the key pathogenic mechanisms of SONFH. Fibroblast growth factor 23 (FGF23) levels are not only a biomarker for early vascular lesions caused by abnormal mineral metabolism, but can also act directly on the peripheral vascular system, leading to vascular pathology. The aim of this study was to observe the role of FGF23 on bone microarchitecture and vascular endothelium, and to investigate activation of pyroptosis in SONFH. Lipopolysaccharide (LPS) combined with methylprednisolone (MPS) was applied for SONFH mouse models, and adenovirus was used to increase or decrease the level of FGF23. Micro-CT and histopathological staining were used to observe the structure of the femoral head, and immunohistochemical staining was used to observe the vascular density. The cells were further cultured in vitro and placed in a hypoxic environment for 12 h to simulate the microenvironment of vascular injury during SONFH. The effect of FGF23 on osteogenic differentiation was evaluated using alkaline phosphatase staining, alizarin red S staining and expression of bone formation-related proteins. Matrigel tube formation assay in vitro and immunofluorescence were used to detect the ability of FGF23 to affect endothelial cell angiogenesis. Steroids activated the pyroptosis signaling pathway, promoted the secretion of inflammatory factors in SONFH models, led to vascular endothelial dysfunction and damaged the femoral head structure. In addition, FGF23 inhibited the HUVECs angiogenesis and BMSCs osteogenic differentiation. FGF23 silencing attenuated steroid-induced osteonecrosis of the femoral head by inhibiting the pyroptosis signaling pathway, and promoting osteogenic differentiation of BMSCs and angiogenesis of HUVECs in vitro.
Collapse
Affiliation(s)
- Lun Fang
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
- Medical School of Nanjing University, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Gang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, Shandong, People's Republic of China
| | - Yadi Wu
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Hao Li
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Zhongzhe Li
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Beilei Yu
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Bin Wang
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Lu Zhou
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China.
| |
Collapse
|
4
|
Zhang L, Xiang Y, Cao C, Tan J, Li F, Yang X. Ciliary neurotrophic factor promotes the development of homocysteine-induced vascular endothelial injury through inflammation mediated by the JAK2/STAT3 signaling pathway. Exp Cell Res 2024; 440:114103. [PMID: 38848951 DOI: 10.1016/j.yexcr.2024.114103] [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: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024]
Abstract
Elevated homocysteine (Hcy) levels have been recognized as significant risk factor for cardiovascular and cerebrovascular diseases, closely related to endothelial injury. While expression of Ciliary Neurotrophic Factor (CNTF) significantly increases during Hcy-induced vascular endothelial cell injury, the precise molecular pathways through which CNTF operates remain to be clarified. To induce vascular endothelial cell injury, human umbilical vein endothelial cells (HUVECs) were treated with Hcy. Cell viability and apoptosis in HUVECs were assessed using the CCK-8 assay and flow cytometry. Western blot analysis determined the expression levels of the JAK2-STAT3 pathway, inflammation-related factors (IL-1β, NLRP3, ICAM-1, VCAM-1), and apoptosis-related factors (cleaved Caspase-3 and Bax). Immunofluorescence staining and western blotting were employed to examine CD31 and α-SMA expression. Knockdown of CNTF was achieved using lentiviral interference, and its effects on inflammation and cell injury were evaluated. Chromatin immunoprecipitation (ChIP) and dual luciferase reporter analysis were conducted to investigate the interaction between the MAFK and CNTF promoters. Our results indicated that Hcy induced high expression of CNTF and activated the JAK2-STAT3 signaling pathway, thereby upregulating factors associated with inflammation and cell apoptosis. Inhibiting CNTF alleviated Hcy-induced inflammation and cell injury. MAFK was identified as a transcription factor promoting CNTF transcription, and its overexpression exacerbated inflammation and cell injury in Hcy-treated HUVECs through the CNTF-JAK2-STAT3 axis, which could be reversed by knocking down CNTF. Activation of MAFK leads to CNTF upregulation, which activates the JAK2-STAT3 signaling pathway, regulating inflammation and inducing injury in Hcy-exposed vascular endothelial cells. Targeting CNTF or its upstream regulator MAFK may represent potential therapeutic strategies for mitigating endothelial dysfunction associated with hyperhomocysteinemia and cardiovascular diseases.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Yan Xiang
- Department of Endocrinology, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, China; Ouyang Road Community Health Service Center, Hongkou District, Shanghai, China
| | - Chengxiu Cao
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Jiaorong Tan
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Fei Li
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China
| | - Xin Yang
- Department of Endocrinology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, 200060, China.
| |
Collapse
|
5
|
Zhang F, Wei L, Wang L, Wang T, Xie Z, Luo H, Li F, Zhang J, Dong W, Liu G, Kang Q, Zhu X, Peng W. Author Correction: FAR591 promotes the pathogenesis and progression of SONFH by regulating Fos expression to mediate the apoptosis of bone microvascular endothelial cells. Bone Res 2024; 12:30. [PMID: 38769361 PMCID: PMC11106272 DOI: 10.1038/s41413-024-00339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Affiliation(s)
- Fei Zhang
- Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Lei Wei
- Department of Orthopedics, Rhode Island Hospital, Brown University, Providence, RI, 02903, USA
| | - Lei Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Tao Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Zhihong Xie
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Hong Luo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Fanchao Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Zhang
- Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Wentao Dong
- Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Gang Liu
- Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Qinglin Kang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xuesong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Wuxun Peng
- Department of Emergency Orthopedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
6
|
Yu Y, Jiang Y, Ge H, Fan X, Gao H, Zhou Z. HIF-1α in cartilage homeostasis, apoptosis, and glycolysis in mice with steroid-induced osteonecrosis of the femoral head. J Cell Physiol 2024; 239:e31224. [PMID: 38481029 DOI: 10.1002/jcp.31224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 05/16/2024]
Abstract
With the prevalence of coronavirus disease 2019, the administration of glucocorticoids (GCs) has become more widespread. Treatment with high-dose GCs leads to a variety of problems, of which steroid-induced osteonecrosis of the femoral head (SONFH) is the most concerning. Since hypoxia-inducible factor 1α (HIF-1α) is a key factor in cartilage development and homeostasis, it may play an important role in the development of SONFH. In this study, SONFH models were established using methylprednisolone (MPS) in mouse and its proliferating chondrocytes to investigate the role of HIF-1α in cartilage differentiation, extracellular matrix (ECM) homeostasis, apoptosis and glycolysis in SONFH mice. The results showed that MPS successfully induced SONFH in vivo and vitro, and MPS-treated cartilage and chondrocytes demonstrated disturbed ECM homeostasis, significantly increased chondrocyte apoptosis rate and glycolysis level. However, compared with normal mice, not only the expression of genes related to collagens and glycolysis, but also chondrocyte apoptosis did not demonstrate significant differences in mice co-treated with MPS and HIF-1α inhibitor. And the effects observed in HIF-1α activator-treated chondrocytes were similar to those induced by MPS. And HIF-1α degraded collagens in cartilage by upregulating its downstream target genes matrix metalloproteinases. The results of activator/inhibitor of endoplasmic reticulum stress (ERS) pathway revealed that the high apoptosis rate induced by MPS was related to the ERS pathway, which was also affected by HIF-1α. Furthermore, HIF-1α affected glucose metabolism in cartilage by increasing the expression of glycolysis-related genes. In conclusion, HIF-1α plays a vital role in the pathogenesis of SONFH by regulating ECM homeostasis, chondrocyte apoptosis, and glycolysis.
Collapse
Affiliation(s)
- Yaling Yu
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yixin Jiang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongfan Ge
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoli Fan
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hang Gao
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhenlei Zhou
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
7
|
Huang C, Qing L, Xiao Y, Tang J, Wu P. Insight into Steroid-Induced ONFH: The Molecular Mechanism and Function of Epigenetic Modification in Mesenchymal Stem Cells. Biomolecules 2023; 14:4. [PMID: 38275745 PMCID: PMC10813482 DOI: 10.3390/biom14010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common refractory orthopedic disease, which is one of the common causes of hip pain and dysfunction. ONFH has a very high disability rate, which is associated with a heavy burden to patients, families, and society. The pathogenesis of ONFH is not completely clear. At present, it is believed that it mainly includes coagulation dysfunction, abnormal lipid metabolism, an imbalance of osteogenic/adipogenic differentiation, and poor vascularization repair. The prevention and treatment of ONFH has always been a great challenge for clinical orthopedic surgeons. However, recent studies have emphasized that the use of mesenchymal stem cells (MSCs) to treat steroid-induced ONFH (SONFH) is a promising therapy. This review focuses on the role and molecular mechanism of epigenetic regulation in the progress of MSCs in the treatment of SONFH, and discusses the significance of the latest research in the treatment of SONFH from the perspective of epigenetics.
Collapse
Affiliation(s)
| | | | | | - Juyu Tang
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China; (C.H.); (L.Q.); (Y.X.)
| | - Panfeng Wu
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China; (C.H.); (L.Q.); (Y.X.)
| |
Collapse
|