1
|
Le G, Wen R, Fang H, Huang Z, Wang Y, Luo H. Exosomal miR-122 derived from M2 macrophages induces osteogenic differentiation of bone marrow mesenchymal stem cells in the treatment of alcoholic osteonecrosis of the femoral head. J Orthop Surg Res 2025; 20:107. [PMID: 39881350 PMCID: PMC11776149 DOI: 10.1186/s13018-025-05515-7] [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: 12/02/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Alcoholic osteonecrosis of the femoral head (AIONFH) is caused by long-term heavy drinking, which leads to abnormal alcohol and lipid metabolism, resulting in femoral head tissue damage, and then pathological necrosis of femoral head tissue. If not treated in time in clinical practice, it will seriously affect the quality of life of patients and even require hip replacement to treat alcoholic femoral head necrosis. This study will confirm whether M2 macrophage exosome (M2-Exo) miR-122 mediates alcohol-induced BMSCs osteogenic differentiation, ultimately leading to the inhibition of femoral head necrosis. M2 macrophages were identified by flow cytometry, and the isolated exosomes were characterized by transmission electron microscopy (TEM) and Nanoparticle Tracking Analysis (NTA). Next, miR-122 was overexpressed by transfecting miR-122 mimic, and the expression of miR-122 in M2 macrophages and their exosomes was evaluated. Subsequently, the effect of exosomal miR-122 on the osteogenic differentiation ability of BMSCs was detected, including cell proliferation, expression of osteogenic-related genes (RUNX2, BMP2, OPN, ALP), and calcium nodule formation. Finally, the therapeutic effect of M2-Exo was analyzed in a rat model of AIONFH, and bone repair and pathological damage were evaluated by Micro-CT, RT-qPCR, HE, Masson staining, and immunohistochemistry (COL I). The results showed that M2 macrophages were successfully polarized, with an average M2-Exo particle size of 156.4 nm and a concentration of 3.2E + 12 particles/mL. The expression of miR-122 in M2 macrophages is significantly higher than that in M0 macrophages, and miR-122 mimic can increase the content of miR-122 in M2-Exo. miR-122 in M2-Exo can promote osteogenic differentiation of rat bone marrow BMSCs, enhance cell viability, and increase the expression of osteogenesis-related genes. After being applied to the AIONFH rat model, the injection of M2-exo and miR-122 mimics significantly improved the repair effect of articular cartilage, alleviated pathological changes, and promoted the regeneration of bone tissue. M2-macrophage-derived exosomal miR-122 induces osteogenic differentiation of bone mesenchymal stem cells in treating AIONFH.
Collapse
Affiliation(s)
- Guoping Le
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Riyou Wen
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Huaixi Fang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Zhifa Huang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Yong Wang
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China
| | - Hanwen Luo
- Department of Joint Osteopathy, Liuzhou Worker's Hospital, Liuzhou, Guangxi Province, 545000, China.
| |
Collapse
|
2
|
Liu T, Liang X, Liu W, Yang S, Cui T, Yan F, Li Z. iRGD-Targeted Biosynthetic Nanobubbles for Ultrasound Molecular Imaging of Osteosarcoma. Int J Nanomedicine 2025; 20:791-805. [PMID: 39867315 PMCID: PMC11760272 DOI: 10.2147/ijn.s494151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/04/2025] [Indexed: 01/28/2025] Open
Abstract
Purpose Osteosarcoma is the most common primary malignant tumor of the bone. However, there is a lack of effective means for early diagnosis due to the heterogeneity of tumors and the complexity of tumor microenvironment. αvβ3 integrin, a crucial role in the growth and spread of tumors, is not only an effective biomarker for cancer angiogenesis, but also highly expressed in many tumor cells. Here, we selected it as the imaging target and fabricated iRGD-sGVs acoustic probe for the early-stage diagnosis of osteosarcoma. Materials and Methods Biological nanoscale gas vesicles (sGVs) were extracted from Serratia 39006. Their morphology was analyzed with phase contrast and transmission electron microscopes. Particle size and zeta potential were measured by a Zetasizer. iRGD-targeted molecular probes (iRGD-sGVs) were prepared by coupling iRGD to sGVs via Mal-PEG2000-NHS. Targeting efficiency of iRGD-sGVs was evaluated using flow cytometry and confocal microscopy on endothelial and K7M2 osteosarcoma cells. In vivo contrast-enhanced ultrasound imaging of iRGD-sGVs was performed in osteosarcoma-bearing mice, and the expression of avβ3 in osteosarcoma was detected through immunofluorescence staining assay. Biocompatibility of sGVs was assessed by hemolysis tests, CCK8 cytotoxicity assays, blood biochemical tests, and HE staining. Results sGVs from Serratia.39006 have smaller particle size (about 160 nm). Our in vitro and in vivo experiments showed the specifically binding ability of iRGD-sGVs to both vascular endothelial cells and tumor cells, producing the stronger and longer acoustic signals in tumors in comparison with the control probe. Immunofluorescence staining results indicated iRGD-sGVs were co-localized with highly expressed αvβ3 in tumor vasculature and osteosarcoma cells. Biocompatibility analysis showed no significant cytotoxicity of iRGD-sGVs to mice. Conclusion Our study provides a new strategy for early diagnosis of osteosarcoma.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Ultrasound, The second People’s Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, People’s Republic of China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530200, People’s Republic of China
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China
| | - Xiaoxin Liang
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People’s Republic of China
| | - Wei Liu
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, People’s Republic of China
| | - Shuai Yang
- Department of Clinical and Research, Shenzhen Mindray Bio-Medical Electronics Co, Ltd, Shenzhen, 518055, People’s Republic of China
| | - Tao Cui
- Medical Imaging center, Shenzhen Yunshan Yunli Hospital, Shenzhen, 518055, People’s Republic of China
| | - Fei Yan
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China
| | - Zhenzhou Li
- Department of Ultrasound, The second People’s Hospital of Shenzhen, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518061, People’s Republic of China
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530200, People’s Republic of China
| |
Collapse
|
3
|
Liu J, Wang C, Qiu S, Sun W, Yang G, Yuan L. Toward Ultrasound Molecular Imaging of Endothelial Dysfunction in Diabetes: Targets, Strategies, and Challenges. ACS APPLIED BIO MATERIALS 2024; 7:1416-1428. [PMID: 38391247 DOI: 10.1021/acsabm.4c00053] [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] [Indexed: 02/24/2024]
Abstract
Diabetes vasculopathy is a significant complication of diabetes mellitus (DM), and early identification and timely intervention can effectively slow the progression. Accumulating studies have shown that diabetes causes vascular complications directly or indirectly through a variety of mechanisms. Direct imaging of the endothelial molecular changes not only identifies the early stage of diabetes vasculopathy but also sheds light on the precise treatment. Targeted ultrasound contrast agent (UCA)-based ultrasound molecular imaging (UMI) can noninvasively detect the expression status of molecular biomarkers overexpressed in the vasculature, thereby being a potential strategy for the diagnosis and treatment response evaluation of DM. Amounts of efforts have been focused on identification of the molecular targets expressed in the vasculature, manufacturing strategies of the targeted UCA, and the clinical translation for the diagnosis and evaluation of therapeutic efficacy in both micro- and macrovasculopathy in DM. This review summarizes the latest research progress on endothelium-targeted UCA and discusses their promising future and challenges in diabetes vasculopathy theranostics.
Collapse
Affiliation(s)
- Jiahan Liu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Chen Wang
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Shuo Qiu
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Wenqi Sun
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| | - Guodong Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University Xi'an, Shaanxi 710032, China
| | - Lijun Yuan
- Department of Ultrasound Medicine, Tangdu Hospital, Fourth Military Medical University, Shaanxi 710038, China
| |
Collapse
|
4
|
Thirumalai A, Girigoswami K, Pallavi P, Harini K, Gowtham P, Girigoswami A. Cancer therapy with iRGD as a tumor-penetrating peptide. Bull Cancer 2023; 110:1288-1300. [PMID: 37813754 DOI: 10.1016/j.bulcan.2023.08.009] [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: 07/03/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
One of the primary threats in tumor treatment revolves around the limited ability to penetrate tumor sites, leading to reduced therapeutic effectiveness, which remains a critical concern. Recently gaining importance are novel peptides, namely CRGDK/RGPD/EC (iRGD), that possess enhanced tumor-penetrating and inhibitory properties. These peptides specifically target and penetrate tumors by binding to αvβ integrins, namely αvβ3 and αvβ5, as well as NRP-1 receptors. Remarkably abundant on both the vasculature and tumor cell surfaces, these peptides show promising potential for improving tumor treatment outcomes. As a result, iRGD penetrated deep into the tumor tissues with biological products, contrast agents (imaging agents), antitumor drugs, and immune modulators after co-injecting them with peptides or chemically linked to peptides. The synthesis of iRGD peptides is a relatively straightforward process compared to the synthesis of other traditional peptides, and they significantly improved tumor tissue penetration inhibiting tumor metastasis effectively. Recent studies demonstrate the effectiveness of iRGD-driven dual-targeting chemotherapeutics on cancer cells, and the nanocarriers were modified with iRGD, serving as a favorable delivery strategy of payloads for deeper tumor regions. This review aims to provide an overview to emphasize the recent advancements and advantages of iRGD in treating and imaging various cancers.
Collapse
Affiliation(s)
- Anbazhagan Thirumalai
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Koyeli Girigoswami
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Pragya Pallavi
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Karthick Harini
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Pemula Gowtham
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India
| | - Agnishwar Girigoswami
- Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Medical Bionanotechnology, Faculty of Allied Health Sciences, TN-603103 Kelambakkam, Chennai, India.
| |
Collapse
|
5
|
Xu HJ, Liu XZ, Yang L, Ning Y, Xu LL, Sun DM, Liao W, Yang Y, Li ZH. Runx2 overexpression promotes bone repair of osteonecrosis of the femoral head (ONFH). Mol Biol Rep 2023; 50:4769-4779. [PMID: 37029290 DOI: 10.1007/s11033-023-08411-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/28/2023] [Indexed: 04/09/2023]
Abstract
BACKGROUND Runt-related transcription factor-2 (Runx2) has been considered an inducer to improve bone repair ability of mesenchymal stem cells (MSCs). METHODS AND RESULTS Twenty-four rabbits were used to establish Osteonecrosis of the femoral head (ONFH) and randomly devided into four groups: Adenovirus Runx2 (Ad-Runx2) group, Runx2-siRNA group, MSCs group and Model group. At 1 week after model establishment, the Ad-Runx2 group was treated with 5 × 107 MSCs transfected through Ad-Runx2, the Runx2-siRNA group was treated with 5 × 107 MSCs transfected through Runx2-siRNA, the MSCs group was injected with 5 × 107 untreated MSCs, and the Model group was treated with saline. The injection was administered at 1 week and 3 weeks after model establishment. The expression of bone morphogenetic protein 2 (BMP-2), Runx2 and Osterix from the femoral head was detected at 3 and 6 weeks after MSCs being injected, and Masson Trichrome Staining, Gross Morphology, X-ray and CT images observation were used to evaluate the repair effect of ONFH. The data revealed that the expression of BMP-2, Runx2 and Osterix in the Runx2-siRNA group was reduced at 3 weeks compared with the MSCs group, and then the expression further reduced at 6 weeks, but was still higher than the Model group besides Osterix; The expression of these three genes in the Ad-Runx2 group was higher than in the MSCs group. Masson Trichrome Staining, Gross Morphology and X-ray and CT images observation revealed that necrotic femoral head of the MSCs group was more regular and smooth than the Runx2-siRNA group, which has a collapsed and irregular femoral head. In the Ad-Runx2 group, necrotic femoral head was basically completely repaired and covered by rich cartilage and bone tissue. CONCLUSIONS Overexpression of Runx2 can improve osteoblastic phenotype maintenance of MSCs and promote necrotic bone repair of ONFH.
Collapse
Affiliation(s)
- Hai-Jia Xu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiang-Zhong Liu
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Lu Yang
- Department of Anesthesiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Ning
- Department of Orthopedics, XiangYang Hospital of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Xiangyang, 441000, China
| | | | - Da-Ming Sun
- Wuhan Sports University, Wuhan, 430079, China
| | - Wen Liao
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yang
- Wuhan Sports University, Wuhan, 430079, China
| | - Zhang-Hua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|