1
|
Sonawane R, Patil S, Rahaman J, Mukherjee D. Effect of microgravity on bone Tissue: Mechanisms of osteodegeneration and advanced treatment modalities. Biochem Biophys Res Commun 2025; 771:152055. [PMID: 40409112 DOI: 10.1016/j.bbrc.2025.152055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Preeminent human existence in space raises concerns about bone health due to the effect of microgravity on bone tissue degeneration. Space experiments pose logistical challenges, but ground-based research using microgravity simulation provides information about bone loss mechanisms. This review compiles and evaluates data from astronaut, animal, and cellular experiments, emphasizing microgravity-induced skeletal deconditioning. These findings contribute to creating treatment approaches for preventing bone loss risks in space and potentially on Earth. Astronauts experience notable bone loss, up to 1 %-2 % per month in a gravity-less environment, predominantly influencing weight-bearing bones. These necessitate finding efficient treatment approaches for preventing bone loss risks in space and potentially on Earth. There is a significant need to investigate microgravity's impact on various bone compartments and skeletal recovery processes. The current review explores the stages of bone remodeling and the fundamental causes of bone loss in microgravity, including effects on osteoblasts, osteocytes, osteoclasts, hematopoietic stem cells, and bone marrow stromal cells, as well as the impact on calcium levels. The article also explores various treatment options, including general management, recent therapies, supportive therapies, and emerging therapies such as BP-NELL-PEG, melatonin, calcitonin, and molecular therapies, highlighting their therapeutic potential against microgravity-induced bone loss.
Collapse
Affiliation(s)
- Rushikesh Sonawane
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Shirpur, 425405, Maharashtra, India
| | - Saurav Patil
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Shirpur, 425405, Maharashtra, India
| | - Jiyaur Rahaman
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Shirpur, 425405, Maharashtra, India; Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, V.L. Mehta road, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Dhrubojyoti Mukherjee
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Shirpur, 425405, Maharashtra, India.
| |
Collapse
|
2
|
Taboada P, Trautmann D, Croley G, Lee J, Deng Z, Kim HKW. Morphologic and Histologic Assessment of Porcine Model of Legg-Calvé-Perthes Disease at a Clinically Relevant Time Point. J Orthop Res 2025. [PMID: 40342202 DOI: 10.1002/jor.26093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/20/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025]
Abstract
Previous studies using the porcine model of Legg-Calvé-Perthes Disease (LCPD) focused on treatments initiated 1 week following osteonecrosis (ON) induction, which represents the early avascular necrosis (AVN) stage (Stage Ia). Most patients, however, present to the clinic during later stages, like the late AVN (Stage Ib) or the early fragmentation stage (Stage IIa), when development of deformity and revascularization is initiated. This study's purpose was to determine the histologic and morphologic changes in the porcine model at 3 weeks after the ON induction. We hypothesized that the 3-week time point better represents a clinically relevant stage of LCPD when patients present to clinic. Eighteen piglets underwent ON induction surgery. Femoral heads were assessed using X-ray, histology, and micro-CT at 3 weeks post-ON. X-ray and histologic assessments revealed subchondral fracture, mild femoral head deformity, variable degree of revascularization (33 ± 30%, range 0%-88%), and early necrotic bone remodeling changes with significantly decreased osteoblast (p < 0.0001) and osteoclast numbers (p < 0.0001) compared to the normal side. Taken together, the results of this study provide comprehensive morphologic and histologic evidence of the late AVN stage (Stage 1b) in the porcine model, which is a clinically relevant time point to develop and test new treatment strategies. A wide variability of revascularization was seen at the 3-week time point, which stresses the importance of evaluating femoral head perfusion and individualizing treatment even in the early stage of AVN.
Collapse
Affiliation(s)
- Phillip Taboada
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel Trautmann
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Garrett Croley
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joonoh Lee
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zhuo Deng
- Center of Excellence in Hip, Scottish Rite for Children, Dallas, Texas, USA
| | - Harry K W Kim
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center of Excellence in Hip, Scottish Rite for Children, Dallas, Texas, USA
| |
Collapse
|
3
|
Angeli LRADE, Santos GBD, Ferreira JRM, Serafim BLC, Lima TZ, Lima LGCADE, Bueno DF, Guarniero R. LOCAL INJECTION OF HUMAN DENTAL PULP STEM CELLS FOR TREATMENT OF JUVENILE AVASCULAR NECROSIS OF THE FEMORAL HEAD: PRELIMINARY RESULTS IN IMMATURE PIGS. ACTA ORTOPEDICA BRASILEIRA 2025; 33:e283445. [PMID: 40206451 PMCID: PMC11978309 DOI: 10.1590/1413-785220243201e283445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/23/2024] [Indexed: 04/11/2025]
Abstract
Introduction Legg-Calvé-Perthes disease is a major cause of hip joint deformities in children. Currently, experimental research is directed at investigating biological therapies, including the use of human dental pulp stem cells (hDPSC), which have not yet been studied for this purpose in swine models. This study aimed to evaluate whether local injection of hDPSC induces bone mineralization in the proximal femoral epiphysis in an experimental model of avascular necrosis of the femoral head in immature pigs. Methods Ten immature pigs underwent surgery to induce osteonecrosis of the proximal femoral epiphysis on the right side. In the intervention group (IG), hDPSC injections were performed immediately after osteonecrosis induction, and in the control group (CG), no additional procedure was performed. Left hips were used as controls. After 8 weeks, all animals were euthanized, and macroscopic, radiographic, and histological evaluations were performed. Results Bone mineralization was greater in the right hips of the IG compared to the CG (p = 0.0356), with an average mineralization index increase of 77.78% after hDPSC injection. Radiographic evaluation of the epiphyseal index showed a greater collapse in the right IG hips compared to the right CG hips (p < 0.001) and macroscopic evaluation showed a higher chance of the femoral head being flat (p = 0,049). Conclusion The injection of hDPSC into the proximal femoral epiphysis with induced osteonecrosis increases bone mineralization in immature pigs, but these treated hips show more deformity compared to the untreated hips. Level of Evidence IV, Case Series .
Collapse
Affiliation(s)
- Luiz Renato Agrizzi DE Angeli
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | - Gustavo Bispo Dos Santos
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | | | - Bárbara Lívia Corrêa Serafim
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| | | | | | | | - Roberto Guarniero
- Universidade de Sao Paulo, Faculdade de Medicina, Hospital das Clinicas HC-FMUSP, Departamento de Ortopedia e Traumatologia DOT, Sao Paulo, SP, Brazil
| |
Collapse
|
4
|
Liu M, Wu C, Wu C, Zhou Z, Fang R, Liu C, Ning R. Immune cells differentiation in osteoarthritic cartilage damage: friends or foes? Front Immunol 2025; 16:1545284. [PMID: 40201177 PMCID: PMC11975574 DOI: 10.3389/fimmu.2025.1545284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Osteoarthritis (OA) is a chronic disease primarily characterized by degenerative changes in articular cartilage and synovitis, for which there are currently no targeted or curative therapies available in clinical practice. In recent years, the in-depth analysis of OA using single-cell sequencing and immunomics technologies has revealed the presence of multiple immune cell subsets, as well as different differentiation states within the same subset, in OA. Through immune-immune and immune-joint tissue interactions, these cells collectively promote or inhibit the progression of arthritis. This complex immune network, where "friends and foes coexist," has made targeted therapeutic strategies aimed at directly eliminating immune cells challenging, highlighting the urgent need for a detailed review of the composition, distribution, functional heterogeneity, therapeutic potential, and potential risks of immune subsets within the joint. Additionally, the similarities and differences between OA and rheumatoid arthritis (RA) in terms of diagnosis and immunotherapy need to be precisely understood, in order to draw lessons from or reject RA-based immunotherapies. To this end, this review summarizes the major triggers of inflammation in OA, the differentiation characteristics of key immune cell subsets, and compares the similarities and differences between OA and RA in diagnosis and treatment. It also outlines the current immunomodulatory strategies for OA and their limitations. Furthermore, we provide a detailed and focused discussion on immune cells that act as "friends or foes" in arthritis, covering the M1/M2 polarization of macrophages, functional heterogeneity of neutrophils, unique roles of dendritic cells at different maturation states, the balance between pro-inflammatory T cells and regulatory T cells (Tregs), and the diverse functions of B cells, plasma cells, and regulatory B cells (Bregs) in OA. By interpreting the roles of these immune cells, this review clarifies the dynamic changes and interactions of immune cells in OA joints, providing a theoretical foundation for more precise targeted interventions in future clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Chenfeng Liu
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), School of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Rende Ning
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University (The First People’s Hospital of Hefei), School of Life Science, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
5
|
Longfei H, Weihua F, Mingli H, Zhikun Z, Mincong H, Qiushi W. Fibroblast IRF7-mediated chondrocyte apoptosis affects the progression of collapse in steroid-induced osteonecrosis of the femoral head. J Orthop Surg Res 2025; 20:292. [PMID: 40102965 PMCID: PMC11921700 DOI: 10.1186/s13018-025-05557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
PURPOSE The objective of this study was to identify potential genes implicated in the "peri-collapse" synovium of osteonecrosis of the femoral head through coding gene sequencing and to further clarify their specific mechanisms via in vitro experiments. METHODS Steroid-induced osteonecrosis of the femoral head (SIONFH) (n = 3), femoral neck fracture (FNF) (n = 3), and hip osteoarthritis (HOA) (n = 3) Synovial tissue of the hip joint was collected in total hip arthroplasty. A cellular model of SIONFH constructed from rat synovial fibroblasts by lipopolysaccharide intervention. Lentiviral technology was used to construct a model for fibroblast knockout of the Irf7 gene. HE was used to compare the characteristics of synovial tissue damage, and immunofluorescence and immunohistochemistry were used to compare the expression levels of VIM, IRF7, and IFNα. PCR, WB, and IF were used to examine Irf7 knockdown efficiency, chondrocyte proliferation (Col2a1, Aggrecan, Sox9), cartilage matrix degradation (Mmp13), and apoptosis (Bcl2, Bax, and Caspase3) expression under co-culture conditions. Crystalline violet staining was used to observe the migration rate of fibroblasts, and flow cytometry was used to detect the apoptosis level of chondrocytes under co-culture conditions. RESULTS Transcriptome sequencing of synovial tissue and fibroblasts ultimately screened for six differential genes, HOOK1, RNPC3, KCNA3, CD48, IRF7, SAMD9. Compared to FNF and HOA, synovial inflammatory cell recruitment and synovial hyperplasia were more pronounced in SIONFH. IF and IHC confirmed high expression of IRF7 and IFNα in the synovium of SIONFH. PCR and WB results suggested that fibroblasts highly expressed Irf7, Hook1, Rnpc3, Kcna3, Cd48, Samd9, Il-6, and Tnfα after lipopolysaccharide intervention, and the expression levels of Il-6 and Tnfα were significantly reduced after knockdown of Irf7 (P < 0.001). In the co-culture system, fibroblasts intervened with lipopolysaccharide significantly promoted chondrocyte apoptosis, the rate of cartilage matrix degradation, while inhibiting the level of chondrocyte proliferation, and this result was significantly reversed in Irf7 knockout fibroblasts. This was supported by flow cytometry results. CONCLUSIONS IRF7, HOOK1, RNPC3, KCNA3, CD48, and SAMD9 as potential genes affecting the progression of SIONFH collapse. Irf7 mediates the fibroblast inflammatory response and affects the collapse process of SIONFH by influencing chondrocyte apoptosis. Thus, intervention in IRF7 holds promise as one of the key targets for reversing the collapse process of SIONFH.
Collapse
Affiliation(s)
- Han Longfei
- Guangzhou University of Chinese Medicine, Guangdong, 510405, Guangzhou, China
| | - Fang Weihua
- Guangzhou University of Chinese Medicine, Guangdong, 510405, Guangzhou, China
| | - Han Mingli
- Guangzhou University of Chinese Medicine, Guangdong, 510405, Guangzhou, China
| | - Zhuang Zhikun
- Department of Orthopaedic Surgery, Quanzhou Orthopedic-traumatological Hospital, Quanzhou, 362000, China.
| | - He Mincong
- Guangdong Academy of Traditional Chinese Medicine Orthopedics and Traumatology, Guangdong, 510378, Guangzhou, China.
- Joint Center of the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510378, Guangzhou, China.
| | - Wei Qiushi
- Guangdong Academy of Traditional Chinese Medicine Orthopedics and Traumatology, Guangdong, 510378, Guangzhou, China.
- Joint Center of the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong, 510378, Guangzhou, China.
| |
Collapse
|
6
|
Li L, Zhao S, Leng Z, Chen S, Shi Y, Shi L, Li J, Mao K, Tang H, Meng B, Wang Y, Shang G, Liu H. Pathological mechanisms and related markers of steroid-induced osteonecrosis of the femoral head. Ann Med 2024; 56:2416070. [PMID: 39529511 PMCID: PMC11559024 DOI: 10.1080/07853890.2024.2416070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Osteonecrosis of the femoral head (ONFH) is a refractory orthopedic disease with a high disability rate. Long-term administration of steroids is the most common pathogenic factor for non-traumatic ONFH. Early diagnosis of steroid-induced osteonecrosis of the femoral head (SONFH) is difficult and mainly depends on imaging. OBJECTIVES The objectives of this review were to examine the pathological mechanisms of SONFH, summarize related markers of SONFH, and identify areas for future studies. METHODS We reviewed studies on pathological mechanisms and related markers of SONFH and discussed the relationship between them, as well as clinical applications and the outlook of potential markers. RESULTS The pathological mechanisms of SONFH included decreased osteogenesis, lipid accumulation, increased intraosseous pressure, and microcirculation disruption. Differential proteomics and genomics play crucial roles in the occurrence, progression, and outcome of SONFH, providing novel insights into SONFH. Additionally, the biological functions of mesenchymal stem cells (MSCs) and exosomes (Exos) in SONFH have attracted increasing attention. CONCLUSIONS The pathological mechanisms of SONFH are complex. The related markers mentioned in the current review can predict the occurrence and progression of SONFH, which will help provide effective early clinical prevention and treatment strategies for SONFH.
Collapse
Affiliation(s)
- Longyu Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shangkun Zhao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zikuan Leng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yifang Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijun Shi
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinfeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Keya Mao
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, China
| | - Hai Tang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bin Meng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yisheng Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Wang X, Wu L, Luo D, He L, Wang H, Peng B. Mechanism of action of Salvia miltiorrhiza on avascular necrosis of the femoral head determined by integrated network pharmacology and molecular dynamics simulation. Sci Rep 2024; 14:28479. [PMID: 39558045 PMCID: PMC11574184 DOI: 10.1038/s41598-024-79532-7] [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: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Avascular necrosis of the femoral head (ANFH) is a progressive, multifactorial, and challenging clinical condition that often leads to hip dysfunction and deterioration. The pathogenesis of ANFH is complex, and there is no foolproof treatment strategy. Although some pharmacologic and surgical treatments have been shown to improve ANFH, the associated side effects and poor prognosis are of concern. Therefore, there is an urgent need to explore therapeutic interventions with superior efficacy and safety to improve the quality of life of patients with ANFH. Salvia miltiorrhiza (SM), a traditional Chinese medicine with a long history, is widely used for the treatment of cardiovascular and musculoskeletal diseases due to its multiple pharmacological activities. However, the molecular mechanism of SM for the treatment of ANFH is still unclear. Therefore, this study aimed to explore the potential targets and mechanisms of SM for the treatment of ANFH using network pharmacology and molecular modeling techniques. By searching multiple databases, we screened 52 compounds and 42 common targets involved in ANFH therapy and identified dan-shexinkum d, cryptotanshinone, tanshinone iia, and dihydrotanshinlactone as key compounds. Based on the protein-protein interaction (PPI) network, TP53, AKT1, EGFR, STAT3, BCL2, IL6, and TNF were identified as core targets. Subsequent enrichment analysis revealed that these targets were mainly enriched in the AGE-RAGE, IL-17, and TNF pathways, which were mainly associated with inflammatory responses, apoptosis, and oxidative stress. In addition, molecular docking and 100 nanoseconds molecular dynamics (MD) simulations showed that the bioactive compounds of SM had excellent affinity and binding strength to the core targets. Among them, dan-shexinkum d possessed the lowest binding free energy (-215.874 kcal/mol and - 140.277 kcal/mol, respectively) for AKT1 and EGFR. These results demonstrated the multi-component, multi-target, and multi-pathway intervention mechanism of SM in the treatment of ANFH, which provided theoretical basis and clues for further experimental validation and development of anti-ANFH drugs.
Collapse
Affiliation(s)
- Xiangjin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Lijiao Wu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Dan Luo
- Basic Medical College of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Langyu He
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Hao Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, 610000, China
| | - Bo Peng
- Department of Respiratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China.
| |
Collapse
|
8
|
Zhu Y, Wang X, Liu R. Bioinformatics proved the existence of potential hub genes activating autophagy to participate in cartilage degeneration in osteonecrosis of the femoral head. J Mol Histol 2024; 55:539-554. [PMID: 38758521 DOI: 10.1007/s10735-024-10200-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 05/01/2024] [Indexed: 05/18/2024]
Abstract
The obvious degeneration of articular cartilage occurs in the late stage of osteonecrosis of the femoral head (ONFH), which aggravates the condition of ONFH. This study aimed to demonstrate aberrant activation of autophagy processes in ONFH chondrocytes through bioinformatics and to predict and identify relevant hub genes and pathways. Differentially expressed genes (DEGs) were identified using R software in the GSE74089 dataset from the GEO database. DEGs were crossed with the Human Autophagy Database (HADb) autophagy genes to screen out autophagy-related differential genes (AT-DEGs). GSEA, GSVA, GO, and KEGG pathway enrichment analyses of AT-DEGs were performed. The STRING database was used to analyze the protein-protein interaction (PPI) of the AT-DEGs network, and the MCODE and CytoHubba plugin in the Cytoscape software was used to analyze the key gene cluster module and screen the hub genes. The PPI network of hub genes was constructed using the GeneMANIA database, and functional enrichment and gene connectivity categories were analyzed. The expression levels of hub genes of related genes in the ONFH patients were verified in the dataset GSE123568, and the protein expression was verified by immunohistochemistry in tissues. The analysis of DEGs revealed abnormal autophagy in ONFH cartilage. AT-DEGs in ONFH have special enrichment in macroautophagy, autophagosome membrane, and phosphatidylinositol-3-phosphate binding. In the GSE123568 dataset, it was also found that ATG2B, ATG4B, and UVRAG were all significantly upregulated in ONFH patients. By immunohistochemistry, it was verified that ATG2B, ATG4B, and UVRAG were significantly overexpressed. These three genes regulate the occurrence and extension of autophagosomes through the PI3KC3C pathway. Finally, we determined that chondrocytes in ONFH undergo positive regulation of autophagy through the corresponding pathways involved in three genes: ATG2B, ATG4B, and UVRAG.
Collapse
Affiliation(s)
- Yingkang Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China
| | - Xianxuan Wang
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruiyu Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xi' an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
9
|
Yuan N, Zhang W, Yang W, Ji W, Li J. Exosomes derived from M2 macrophages prevent steroid-induced osteonecrosis of the femoral head by modulating inflammation, promoting bone formation and inhibiting bone resorption. J Orthop Surg Res 2024; 19:243. [PMID: 38622659 PMCID: PMC11020342 DOI: 10.1186/s13018-024-04711-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Inflammatory reactions are involved in the development of steroid-induced osteonecrosis of the femoral head(ONFH). Studies have explored the therapeutic efficacy of inhibiting inflammatory reactions in steroid-induced ONFH and revealed that inhibiting inflammation may be a new strategy for preventing the development of steroid-induced ONFH. Exosomes derived from M2 macrophages(M2-Exos) display anti-inflammatory properties. This study aimed to examine the preventive effect of M2-Exos on early-stage steroid-induced ONFH and explore the underlying mechanisms involved. In vitro, we explored the effect of M2-Exos on the proliferation and osteogenic differentiation of bone marrow-derived mesenchymal stem cells(BMMSCs). In vivo, we investigated the role of M2-Exos on inflammation, osteoclastogenesis, osteogenesis and angiogenesis in an early-stage rat model of steroid-induced ONFH. We found that M2-Exos promoted the proliferation and osteogenic differentiation of BMMSCs. Additionally, M2-Exos effectively attenuated the osteonecrotic changes, inhibited the expression of proinflammatory mediators, promoted osteogenesis and angiogenesis, reduced osteoclastogenesis, and regulated the polarization of M1/M2 macrophages in steroid-induced ONFH. Taken together, our data suggest that M2-Exos are effective at preventing steroid-induced ONFH. These findings may be helpful for providing a potential strategy to prevent the development of steroid-induced ONFH.
Collapse
Affiliation(s)
- Na Yuan
- Department of Ultrasonography, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Weiying Zhang
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
- Xizang Minzu University, XianYang, Shaanxi Province, 712082, China
| | - Weizhou Yang
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Wenchen Ji
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China
| | - Jia Li
- Department of Orthopedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, 710061, China.
| |
Collapse
|
10
|
Ozawa Y, Takegami Y, Osawa Y, Asamoto T, Tanaka S, Imagama S. Anti-sclerostin antibody therapy prevents post-ischemic osteonecrosis bone collapse via interleukin-6 association. Bone 2024; 181:117030. [PMID: 38309414 DOI: 10.1016/j.bone.2024.117030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating condition characterized by subchondral bone necrosis, which frequently culminates in joint destruction. Although total hip arthroplasty is conventionally practiced to remediate ONFH, for patients under the age of 60, the outcomes can be suboptimal. Chronic inflammation, particularly that mediated by interleukin-6 (IL-6), has been conjectured to be a potential mechanism underlying the etiology of ONFH. This study aimed at exploring the interplay between IL-6, the canonical Wnt signaling pathway, and ONFH to provide insights for potential therapeutic interventions. Human ONFH specimens depicted an elevation in β-catenin expression in the transitional layer, while IL-6 levels were pronounced in the same region. Subsequently, mouse models of ischemic osteonecrosis were treated with an anti-sclerostin antibody to assess its effects on bone metabolism and cellular processes. Histological analysis revealed that the administration of anti-sclerostin antibodies effectuated early recovery from bone necrosis, reduced empty lacunae, and suppressed IL-6 expression. The treatment evidently initiated the activation of the Wnt/β-catenin signaling pathway, presenting a potential mechanism associated with IL-6-mediated inflammation. Furthermore, the antibody upregulated osteoblast formation, downregulated osteoclast formation, and increased bone volume. Micro-CT imaging demonstrated increased bone volume, prevented epiphyseal deformity, and improved compression strength. Therefore, this study yields significant findings, indicating the potency of anti-sclerostin antibodies in effectively modulating the Wnt/β-catenin pathway, associating with IL-6 expression, and preventing post-ONFH bone collapse. Additionally, this preclinical investigation in mouse models offers an avenue for prospective research on potential therapeutic interventions against human ONFH.
Collapse
Affiliation(s)
- Yuto Ozawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Yasuhiko Takegami
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan.
| | - Yusuke Osawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Takamune Asamoto
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shinya Tanaka
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| |
Collapse
|
11
|
Deng Z, Aguirre-Flores M, Kim HKW, Ren Y. Obesity impairs revascularization and bone healing in a mouse model of osteonecrosis. J Orthop Res 2024; 42:811-820. [PMID: 37975620 DOI: 10.1002/jor.25728] [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: 04/03/2023] [Revised: 07/21/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating bone disease that is caused by a disruption of blood supply leading to necrotic cell death. Clinically, it was found that obesity has a high prevalence with ONFH. However, it remains unclear how obesity may directly affect tissue regeneration and bone healing in osteonecrosis (ON). The purpose of this study is to investigate the effects of obesity and weight loss (WL) on ON healing. In this study, we induced obesity and WL in an established surgery-induced ON mouse model via feeding a high-fat diet (HFD) and altering the diet respectively. All mice received a surgical induction of ON of distal femoral epiphysis at the age of 12 weeks. HFD was switched to normal diet (ND) after ON surgery to induce WL. Mouse body weight was recorded weekly. Mouse body composition was scanned by DEXA (Dual-energy X-ray absorptiometry) right after sacrifice at the age of 16 weeks. The distal femoral bone samples were fixed and embedded for histology such as H&E, immunohistochemistry, and TRAP staining. In this study, we found that HFD-induced obesity impaired revascularization and bone remodeling showing decreased vessel areas and reduced osteoblast and osteoclast numbers. WL could rescue obesity-induced bone healing defects. Our study is the first to test the direct effects of obesity and WL on ON bone healing. We believe our work may provide new concepts for osteonecrosis treatment in obese patients.
Collapse
Affiliation(s)
- Zhuo Deng
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, Texas, USA
| | | | - Harry K W Kim
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, Texas, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yinshi Ren
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, Texas, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
12
|
Meng Q, Wang Y, Yuan T, Su Y, Li Z, Sun S. Osteoclast: The novel whistleblower in osteonecrosis of the femoral head. GENE REPORTS 2023; 33:101833. [DOI: 10.1016/j.genrep.2023.101833] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Kuroyanagi G, Kamiya N, Yamaguchi R, Kim HK. Interleukin-6 receptor blockade improves bone healing following ischemic osteonecrosis in adolescent mice. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100386. [PMID: 37600923 PMCID: PMC10432805 DOI: 10.1016/j.ocarto.2023.100386] [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: 10/19/2022] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
Objective Juvenile ischemic osteonecrosis (JIO) of the femoral head is one of the most serious hip disorders causing a permanent deformity of the femoral head in childhood. We recently reported that interleukin 6 (IL-6) is significantly increased in the hip synovial fluid of patients with JIO and that articular chondrocytes are primary source of IL-6. Adolescent JIO is particularly challenging to treat and has poor outcome. This study determined if IL-6 receptor blockade prevents bone loss and improves the bone healing in adolescent JIO. Method Adolescent mice (12-week-old) surgically induced with JIO were treated with either saline or MR16-1, an IL-6 receptor blocker. Results Micro-CT assessment showed significantly increased bone volume (p < 0.001, Cohen's d = 2.0) and trabecular bone thickness (p < 0.001, d = 2.3) after the MR16-1 treatment. Histomorphometric assessment showed significantly increased osteoblast number (p < 0.01, d = 2.3), bone formation rate (p < 0.01, d = 4.3), and mineral apposition rate (p < 0.01, d = 4.1) after the MR16-1 treatment. The number of osteoclasts was unchanged. Histologic assessment showed significantly increased revascularization (p < 0.01) and restoration of the necrotic marrow with new hematopoietic bone marrow (p < 0.01). Vascular endothelial growth factor (VEGF) expression was increased in the revascularized area and the articular cartilage, and in the cultured chondrocytes treated with IL-6 receptor inhibitor. Conclusion IL-6 blockade in adolescent mice with JIO enhanced bone formation and revascularization. The findings suggest IL-6 receptor blocker as a potential medical therapy for adolescent JIO.
Collapse
Affiliation(s)
- Gen Kuroyanagi
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, TX 75219, USA
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Nobuhiro Kamiya
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, TX 75219, USA
- Faculty of Budo and Sport Studies, Tenri University, Nara 6320071, Japan
| | - Ryosuke Yamaguchi
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, TX 75219, USA
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Harry K.W. Kim
- Center for Excellence in Hip, Scottish Rite for Children, Dallas, TX 75219, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8883, USA
| |
Collapse
|
14
|
Wang Y, Zhan Y, Ji C, Shi C, Han J. Houttuynia cordata Thunb repairs steroid-induced avascular necrosis of the femoral head through regulating NF-κB signaling pathway. Toxicon 2023; 233:107270. [PMID: 37652100 DOI: 10.1016/j.toxicon.2023.107270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
To investigate the influences of Houttuynia cordata Thunb (HCT) in steroid-induced avascular necrosis of the femoral head (SANFH), we conducted a comprehensive study evaluating the effects of HCT on various aspects. Cell Counting Kit-8 assay was used to examine bone marrow stem cells (BMSCs) cell viability. Flow cytometry and lactate dehydrogenase detection assay were conducted to determine cell apoptosis. The levels of apoptosis-related proteins, osteogenic-related markers, inflammatory factors, and nuclear factor kappa B (NF-κB) pathway-associated proteins were determined via western blotting. Hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling assays were utilized to verify the effects of HCT in SANFH rats. Our findings revealed that HCT could enhanced cell viability and arrested cell apoptosis in dexamethasone (Dex)-treated BMSCs. Dex increased the levels of cleaved caspase-3, Bcl2-associated X, interleukin (IL)-1β, IL-18, IL-6, p65, and inhibitor of NF-κB kinase β (IKKβ), while this promoting trend was weakened by HCT. Moreover, pyrrolidine dithiocarbamate (PDTC, an inhibitor of NF-κB signaling pathway) further increased the inhibitory role of apoptosis and the levels of IL-1β, IL-18, and IL-6 and the promotional effect of the levels of RUNX2 and ALP in Dex-treated BMSCs. The in-vivo assays showed that HCT decreased the percentage of empty lacunae, apoptosis, and the levels of IL-1β, IL-18, IL-6, p65, and IKKβ in SANFH rats. In conclusion, our study demonstrated that HCT relieved SANFH, which might be possibly achieved by NF-κB pathway.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Medical Insurance Office, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital, Qingdao, Shandong, 266000, PR China
| | - Yaqi Zhan
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Chunxiao Ji
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Chunlei Shi
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China
| | - Jie Han
- Hematology Department, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, Shandong, 266000, PR China.
| |
Collapse
|
15
|
Zhang T, Hu X, Yu S, Wei C. Construction of ceRNA network based on RNA-seq for identifying prognostic lncRNA biomarkers in Perthes disease. Front Genet 2023; 14:1105893. [PMID: 37303951 PMCID: PMC10252144 DOI: 10.3389/fgene.2023.1105893] [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: 11/23/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction: Legg-Calvé-Perthes disease or Perthes disease is a condition that occurs in children aged 2 to 15 years, and is characterized by osteonecrosis of the femoral head, which results in physical limitations. Despite ongoing research, the pathogenesis and molecular mechanisms underlying the development of Perthes disease remain unclear. In order to obtain further insights, the expression patterns of long non-coding RNAs (lncRNAs), miRNAs, and mRNAs in a rabbit model of Perthes disease were analyzed in this study by transcriptome sequencing. Methods and results: The results of RNA-seq analyses revealed that 77 lncRNAs, 239 miRNAs, and 1027 mRNAs were differentially expressed in the rabbit model. This finding suggested that multiple genetic pathways are involved in the development of Perthes disease. A weighted gene co-expression network analysis (WGCNA) network was subsequently constructed using the differentially expressed mRNAs (DEmRNAs), and network analysis revealed that the genes associated with angiogenesis and platelet activation were downregulated, which was consistent with the findings of Perthes disease. A competing endogenous RNA (ceRNA) network was additionally constructed using 29 differentially expressed lncRNAs (including HIF3A and LOC103350994), 28 differentially expressed miRNAs (including ocu-miR-574-5p and ocu-miR-324-3p), and 76 DEmRNAs (including ALOX12 and PTGER2). Disscusion: The results obtained herein provide novel perspectives regarding the pathogenesis and molecular mechanisms underlying the development of Perthes disease. The findings of this study can pave the way for the development of effective therapeutic strategies for Perthes disease in future.
Collapse
Affiliation(s)
- Tianjiu Zhang
- Guizhou Children’s Hospital, Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaolin Hu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Yu
- Guizhou Children’s Hospital, Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chunyan Wei
- Department of Gynecoloay, Obstetrics and Gynecoloay Hospital of Fudan University, Shanchai, China
| |
Collapse
|
16
|
Molecular Biomarkers in Perthes Disease: A Review. Diagnostics (Basel) 2023; 13:diagnostics13030471. [PMID: 36766577 PMCID: PMC9914190 DOI: 10.3390/diagnostics13030471] [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: 12/18/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Perthes disease is a juvenile form of osteonecrosis of the femoral head that affects children under the age of 15. One hundred years after its discovery, some light has been shed on its etiology and the biological factors relevant to its etiology and disease severity. METHODS The aim of this study was to summarize the literature findings on the biological factors relevant to the pathogenesis of Perthes disease, their diagnostic and clinical significance, and their therapeutic potential. A special focus on candidate genes as susceptibility factors and factors relevant to clinical severity was made, where studies reporting clinical or preclinical results were considered as the inclusion criteria. PubMed databases were searched by two independent researchers. Sixty-eight articles were included in this review. Results on the factors relevant to vascular involvement and inflammatory molecules indicated as factors that contribute to impaired bone remodeling have been summarized. Moreover, several candidate genes relevant to an active phase of the disease have been suggested as possible biological therapeutic targets. CONCLUSIONS Delineation of molecular biomarkers that underlie the pathophysiological process of Perthes disease can allow for the provision of earlier and more accurate diagnoses of the disease and more precise follow-ups and treatment in the early phases of the disease.
Collapse
|
17
|
A GP130-Targeting Small Molecule, LMT-28, Reduces LPS-Induced Bone Resorption around Implants in Diabetic Models by Inhibiting IL-6/GP130/JAK2/STAT3 Signaling. Mediators Inflamm 2023; 2023:9330439. [PMID: 36643585 PMCID: PMC9839413 DOI: 10.1155/2023/9330439] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 12/10/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
In this study, we examined the effect of the GP130-targeting molecule, LMT-28, on lipopolysaccharide- (LPS-) induced bone resorption around implants in diabetic models using in vitro and rat animal experiments. First, LMT-28 was added to osteoblasts stimulated by LPS and advanced glycation end products (AGEs), and nuclear factor-κB receptor-activating factor ligand (RANKL) and associated pathways were evaluated. Then, LMT-28 was administered by gavage at 0.23 mg/kg once every 5 days for 2 weeks to type 2 diabetic rats with peri-implantitis induced by LPS injection and silk ligature. The expression of IL-6 and RANKL was evaluated by immunohistochemistry, and the bone resorption around implants was evaluated by microcomputed tomography. The results showed that LMT-28 downregulated the expression of RANKL through the JAK2/STAT3 signaling pathway in osteoblasts stimulated by LPS and AGEs, reduced bone resorption around implants with peri-implantitis, decreased the expression of IL-6 and RANKL, and decreased osteoclast activity in type 2 diabetic rats. This study confirmed the ability of LMT-28 to reduce LPS-induced bone resorption around implants in diabetic rats.
Collapse
|
18
|
Ma J, Sun Y, Zhou H, Li X, Bai Y, Liang C, Jia X, Zhang P, Yang L. Animal Models of Femur Head Necrosis for Tissue Engineering and Biomaterials Research. Tissue Eng Part C Methods 2022; 28:214-227. [PMID: 35442092 DOI: 10.1089/ten.tec.2022.0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Femur head necrosis, also known as osteonecrosis of the femoral head (ONFH), is a widespread disabling pathology mostly affecting young and middle-aged population and one of the major causes of total hip arthroplasty in the elderly. Currently, there are limited number of different clinical or medication options for the treatment or the reversal of progressive ONFH, but their clinical outcomes are neither satisfactory nor consistent. In pursuit of more reliable therapeutic strategies for ONFH, including recently emerged tissue engineering and biomaterials approaches, in vivo animal models are extremely important for therapeutic efficacy evaluation and mechanistic exploration. Based on the better understanding of pathogenesis of ONFH, animal modeling method has evolved into three major routes, including steroid-, alcohol-, and injury/trauma-induced osteonecrosis, respectively. There is no consensus yet on a standardized ONFH animal model for tissue engineering and biomaterial research; therefore, appropriate animal modeling method should be carefully selected depending on research purposes and scientific hypotheses. In this work, mainstream types of ONFH animal model and their modeling techniques are summarized, showing both merits and demerits for each. In addition, current studies and experimental techniques of evaluating therapeutic efficacy on the treatment of ONFH using animal models are also summarized, along with discussions on future directions related to tissue engineering and biomaterial research. Impact statement Exploration of tissue engineering and biomaterial-based therapeutic strategy for the treatment of femur head necrosis is important since there are limited options available with satisfactory clinical outcomes. To promote the translation of these technologies from benchwork to bedside, animal model should be carefully selected to provide reliable results and clinical outcome prediction. Therefore, osteonecrosis of the femoral head animal modeling methods as well as associated tissue engineering and biomaterial research are overviewed and discussed in this work, as an attempt to provide guidance for model selection and optimization in tissue engineering and biomaterial translational studies.
Collapse
Affiliation(s)
- Jiali Ma
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Yuting Sun
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Huan Zhou
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Center for Health Sciences and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yanjie Bai
- School of Chemical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Chunyong Liang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Changzhou Blon Minimally Invasive Medical Device Technology Co. Ltd., Jiangsu, People's Republic of China
| | - Xiaowei Jia
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Lei Yang
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, People's Republic of China.,Center for Health Sciences and Engineering, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, People's Republic of China
| |
Collapse
|
19
|
Tan Z, Wang Y, Chen Y, Liu Y, Ma M, Ma Z, Wang C, Zeng H, Xue L, Yue C, Wang D. The Dynamic Feature of Macrophage M1/M2 Imbalance Facilitates the Progression of Non-Traumatic Osteonecrosis of the Femoral Head. Front Bioeng Biotechnol 2022; 10:912133. [PMID: 35573242 PMCID: PMC9094367 DOI: 10.3389/fbioe.2022.912133] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Non-traumatic osteonecrosis of the femoral head (NONFH) remains a common refractory disease with poorly understood pathogenesis. Macrophage M1/M2 imbalance and chronic inflammatory microenvironment have been suggested to be closely related to osteonecrosis. Here we describe direct visual evidence for the involvement of dynamic changes in macrophages and the chronic inflammatory microenvironment in human NONFH. Osteonecrosis induces inflammatory responses and macrophage enrichment in the reparative area, and the number of inflammatory cells and macrophages falls during progressive-to end-stage NONFH. Multiplex immunohistochemistry demonstrated that macrophage M1/M2 ratio increased from 3 to 10 during progressive-to end-stage. During the progressive-stage, new blood vessels formed in the reparative area, M2 macrophages accumulated in perivascular (M1/M2 ratio ∼0.05), while M1 macrophages were enriched in avascular areas (M1/M2 ratio ∼12). Furthermore, inflammatory cytokines were detected in synovial fluid and plasma using cytometric bead arrays. Interleukin (IL)-6 and IL-1β were persistently enriched in synovial fluid compared to plasma in patients with NONFH, and this difference was confirmed by immunohistochemistry staining. However, only IL-6 levels in plasma were higher in patients with progressive-stage NONFH than in osteoarthritis. Moreover, fibrosis tissues were observed in the necrotic area in progressive-stage and end-stage NONFH based on Sirius Red staining. Together, these findings indicate that macrophage M1/M2 imbalance facilitates the progression of NONFH, a chronic inflammatory disease characterized by chronic inflammation, osteonecrosis and tissue fibrosis in the local lesion. Inhibiting inflammation, promoting the resolution of inflammation, switching macrophages to an M2 phenotype, or inhibiting their adoption of an M1 phenotype may be useful therapeutic strategies against NONFH.
Collapse
Affiliation(s)
- Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yan Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Yingqi Chen
- Department of Bone and Joint Surgery, National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Youwen Liu
- Department of Orthopedic, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Maoxiao Ma
- Department of Orthopedic, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Zetao Ma
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Chao Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Hui Zeng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Bone and Joint Surgery, National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- *Correspondence: Lixiang Xue, ; Chen Yue, ; Deli Wang,
| | - Chen Yue
- Department of Orthopedic, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
- *Correspondence: Lixiang Xue, ; Chen Yue, ; Deli Wang,
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Bone and Joint Surgery, National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, China
- *Correspondence: Lixiang Xue, ; Chen Yue, ; Deli Wang,
| |
Collapse
|