1
|
Ma X, Li Z, Ma H, Jiang K, Chen B, Wang W, Zhu Z, Wang J, Yang Z, Yunqing W, Dong S. Rotenone inhibited osteosarcoma metastasis by modulating ZO-2 expression and location via the ROS/Ca 2+/AMPK pathway. Redox Rep 2025; 30:2493556. [PMID: 40247635 PMCID: PMC12010658 DOI: 10.1080/13510002.2025.2493556] [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: 04/19/2025] Open
Abstract
BACKGROUND Pulmonary metastases in osteosarcoma (OS) are associated with a poor prognosis. Rotenone has shown anti-cancer activity. However, its effects on metastasis and the underlying mechanisms remain unknown. This study investigated the potential use of Rotenone for OS treatment. METHODS The effect of Rotenone and ROS/Ca2+/AMPK/ZO-2 pathway on metastasis and EMT was evaluated by Western blot, Transwell and Wound healing. Flow cytometer was employed to measure the intracellular Ros and Ca2+ levels. The subcellular location of ZO-2 was detected by IF, interaction between AMPK and ZO-2 were examined by Co-IP. Then, subcutaneous tumor and metastasis models were used to evaluate the function of Rotenone in OS metastasis. RESULTS Rotenone-induced ROS led to increased intracellular Ca2+, which promoted the EMT of OS cells through activation of AMPK and ZO-2 nuclear translocation. Inhibition of ROS production decreased intracellular Ca2+, restraining AMPK activity. Knock-down of ZO-2 significantly suppressed the anti-metastasis effects of Rotenone in OS cells. Moreover, Rotenone elevated p-AMPK and ZO-2 expression but inhibited EMT and lung metastasis in vivo.Conclusion These results provide evidence supporting an anti-metastatic effect of Rotenone. These findings support the use of Rotenone in the prevention of OS metastasis.
Collapse
Affiliation(s)
- Xiang Ma
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhen Li
- Department of Medical Oncology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Hengwei Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Kun Jiang
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Bao Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Weiquan Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Ziqiang Zhu
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jianqiang Wang
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Zuozhang Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Wang Yunqing
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Suwei Dong
- Department of Orthopaedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, People’s Republic of China
| |
Collapse
|
2
|
Feng J, Zhao M, Chen Z, Lin C, Jin J, Ma N, Bai C, Li ZH, Xu W, She C. STUB1-mediated ubiquitination of SLC25A10 regulates mitochondrial function and drives osteosarcoma progression: A novel therapeutic target. Cell Signal 2025; 132:111855. [PMID: 40349811 DOI: 10.1016/j.cellsig.2025.111855] [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: 03/03/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Osteosarcoma (OS) is a highly aggressive primary bone malignancy characterized by limited treatment options and poor clinical outcomes. Emerging evidence underscores the critical role of mitochondrial metabolism in tumor progression, positioning mitochondrial proteins as potential therapeutic targets. SLC25A10, a mitochondrial dicarboxylate carrier involved in redox homeostasis and fatty acid synthesis, has been implicated in various cancers; however, its role in OS remains unclear.In this study, we investigated the function of SLC25A10 in OS progression and its potential as a therapeutic target. Our results revealed that SLC25A10 expression is significantly upregulated in OS tissues and cell lines compared to normal bone tissue, and its elevated expression is associated with poor patient prognosis. Functional assays demonstrated that silencing SLC25A10 via shRNA or CRISPR/Cas9 significantly suppressed OS cell proliferation, migration, and mitochondrial function, resulting in mitochondrial membrane depolarization, oxidative damage, and apoptosis. In contrast, SLC25A10 overexpression promoted OS cell proliferation and migration. In vivo, knockout of SLC25A10 markedly inhibited the growth of subcutaneous OS xenografts in nude mice.Furthermore, we identified STUB1, an E3 ubiquitin ligase, as a negative regulator of SLC25A10. STUB1 knockdown reduced the ubiquitination of SLC25A10, leading to increased protein stability and elevated expression. Notably, lysine 254 (K254) was identified as a key site mediating STUB1-dependent ubiquitination of SLC25A10. STUB1-mediated downregulation of SLC25A10 suppressed OS cell proliferation and migration, indicating a tumor-suppressive role for STUB1 in OS through modulation of SLC25A10.Collectively, our findings demonstrate that SLC25A10 is essential for maintaining mitochondrial function and contributes to OS malignancy. Targeting SLC25A10 may represent a novel and promising therapeutic strategy for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Junchao Feng
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Department of Nuclear Accident Medical Emergency, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingzhi Zhao
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Department of Orthopedics, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zhanhong Chen
- The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Changjie Lin
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jieyu Jin
- Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Nan Ma
- The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Chaowen Bai
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Zhi-Hong Li
- The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China.
| | - Wei Xu
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.
| | - Chang She
- Department of Orthopedics, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Jian G, Wang S, Wang X, Lu Q, Zhu X, Wan S, Wang S, Li D, Wang C, He Q, Chen T, Song J. Enhanced sequential osteosarcoma therapy using a 3D-Printed bioceramic scaffold combined with 2D nanosheets via NIR-II photothermal-chemodynamic synergy. Bioact Mater 2025; 50:540-555. [PMID: 40391104 PMCID: PMC12088772 DOI: 10.1016/j.bioactmat.2025.04.029] [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: 11/08/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/21/2025] Open
Abstract
Background Osteosarcoma (OS) is a malignant tumor originating from primitive mesenchymal cells, characterized by rapid metastasis, high invasiveness, and significant mortality. The primary challenges in OS management include the effective elimination of residual tumor cells to prevent recurrence and the repair of extensive bone defects caused by surgical intervention. Objective This study aims to develop an innovative biomimetic 3D-printed bioactive glass ceramic (BGC) scaffold modified with two-dimensional nanosheets to address both tumor ablation and bone tissue repair. Materials and methods The nanosheets were constructed via ellagic acid (EA) and ruthenium (Ru) coordination, leveraging the non-topological adhesion properties of catechol in EA to deposit the nanosheets onto the BGC scaffold (EARu-BGC). The therapeutic effects of EARu-BGC were evaluated in vitro and in vivo. Results EARu-BGC sequentially responds to the local microenvironment during OS treatment. During the tumor ablation phase, EARu-BGC induced ferroptosis through the synergistic effects of photothermal and chemodynamic therapy, achieving over 90 % tumor cell ablation and significantly inhibiting tumor volume and weight. In the bone tissue repair phase, EARu-BGC exhibited adaptive ROS scavenging and facilitated a pro-healing microenvironment, promoting osteogenic differentiation. The gradual degradation of the BGC scaffold provided essential minerals and space for new bone formation. In vivo experiments demonstrated that EARu-BGC significantly enhanced osteogenesis, increasing the trabecular number to 1.51 ± 0.15/mm and reducing trabecular separation to 1.50 ± 0.04 mm. Conclusion The EARu-BGC scaffold presents a promising multifunctional platform for OS treatment by effectively balancing antitumor efficacy with bone repair capabilities.
Collapse
Affiliation(s)
- Guangyu Jian
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Si Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Xinlu Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Qinyi Lu
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Xingyu Zhu
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Shucheng Wan
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Shan Wang
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Dize Li
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Chao Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Beihang University, Beijing 100083, PR China
| | - Qingqing He
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Tao Chen
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| | - Jinlin Song
- The Affiliated Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases, Chongqing Municipal Key aboratory of Oral Biomedical Engineering of Higher Education, Chongqing Municipal Health Commission Key Laboratory of Oral Biomedical Engineering, Chongqing, 401147, PR China
| |
Collapse
|
4
|
Jiang M, Li H, Zhang Q, Xu T, Huang L, Zhang J, Yu H, Zhang J. The role of RGS12 in tissue repair and human diseases. Genes Dis 2025; 12:101480. [PMID: 40271194 PMCID: PMC12017852 DOI: 10.1016/j.gendis.2024.101480] [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: 02/05/2024] [Revised: 08/05/2024] [Accepted: 11/02/2024] [Indexed: 04/25/2025] Open
Abstract
Regulator of G protein signaling 12 (RGS12) belongs to the superfamily of RGS proteins defined by a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. As the largest family member, RGS12 is widely expressed in many cells and tissues. In the past few decades, it has been found that RGS12 affects the activity of various cells in the human body, participates in many physiological and pathological processes, and plays an important role in the pathogenesis of many diseases. Here, we set out to comprehensively review the role of RGS12 in human diseases and its mechanisms, highlighting the possibility of RGS12 as a therapeutic target for the treatment of human diseases.
Collapse
Affiliation(s)
- Min Jiang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongmei Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tongtong Xu
- General Department of Critical Care Medicine, Zhenjiang Traditional Chinese Medicine Hospital, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Zhenjiang, Jiangsu 212003, China
| | - Le Huang
- Army 72nd Group Military Hospital, Huzhou, Zhejiang 313000, China
| | - Jinghong Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Huiqing Yu
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Junhui Zhang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
5
|
Wang Y, Lu P. MOPSOGAT: Predicting CircRNA-Disease Associations via Improved Multi-objective Particle Swarm Optimization and Graph Attention Network. Interdiscip Sci 2025:10.1007/s12539-025-00725-3. [PMID: 40514639 DOI: 10.1007/s12539-025-00725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 06/16/2025]
Abstract
Recently increasing researches have discovered that circRNAs are remarkably reliable in organisms and play a crucial role as marker in many diseases. Although deep learning techniques has been universally applied to investigate the relationship of circRNA-disease, optimizing many parameters involved in these techniques for best performance has been a challenge. Therefore, we present, for the first time, a multi-objective particle swarm optimization algorithm to optimize the parameters in a graph attention network, ensuring that the model operates at peak efficiency. In addition, it also limits feature learning due to uneven distribution of different node types in heterogeneous graphs based on association relationships. We suggest a unique approach, MOPSOGAT, to overcome the aforementioned problems. MOPSOGAT is a method for predicting circRNA-disease associations utilizing the improved multi-objective particle swarm optimization (MOPSO) and the graph attention network. Initially, we obtain node sequences by utilizing multiple circRNA similarities and disease phenotypic similarities, and employing a heterogeneous graph with random walks incorporating jump and stay strategies. These sequences are then processed using word2vec to derive the neighbor vectors of the nodes, thus providing initial embeddings for circRNAs and diseases. Subsequently, in order to model convergence and diversity of the Pareto front solutions, an improved MOPSO algorithm is used to iteratively search for optimal solutions in the parameter space. After MOPSO optimization, parameters are fed into a graph attention network to further refine the model embedding. As a result, MOPSOGAT performs better than deep learning based methods, solely multi-objective optimization-based methods and machine learning-based ways. Moreover, the potential associations predicted by MOPSOGAT have been validated through case studies, further demonstrating the potential of MOPSOGAT in future biomedical research.
Collapse
Affiliation(s)
- Yuehao Wang
- School of Computer and Communication, Lanzhou University of Technology, Lanzhou, 730050, China
| | - Pengli Lu
- School of Computer and Communication, Lanzhou University of Technology, Lanzhou, 730050, China.
| |
Collapse
|
6
|
Li H, Bao T, Huang X, Zhou J, Zhang Z, Wang X, You W, Cao L, Han C. Overcoming drug resistance in osteosarcoma with MTX-CuB-NLC: An in vitro and in vivo study. Eur J Pharm Biopharm 2025:114779. [PMID: 40490044 DOI: 10.1016/j.ejpb.2025.114779] [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: 01/07/2025] [Revised: 02/11/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Osteosarcoma (OS) is the predominant bone tumor affecting pediatric and adolescent populations. The standard treatment regimen involves preoperative chemotherapy, surgical intervention, and postoperative chemotherapy. Methotrexate (MTX) serves as the first-line pharmacological agent for OS treatment; however, the emergence of tumor resistance to chemotherapeutic agents poses a significant challenge. Cucurbitacin B (CuB) exhibits intrinsic anti-OS properties and can synergistically enhance OS suppression by reversing drug resistance and augmenting the therapeutic effects of MTX. Nevertheless, the clinical application of CuB and MTX is hindered by their low aqueous solubility, necessitating the development of an effective drug delivery system to precisely target tumor tissues and maximize therapeutic efficacy. Consequently, this study focuses on the development of a nanostructured lipid carrier (NLC) co-loaded with MTX and CuB (MTX-CuB-NLC) to address these limitations. MTX-CuB-NLC is characterized as a spherical nanoparticle with a mean particle size of 44.13 ± 1.40 nm, a polydispersity index (PDI) of 0.279 ± 0.120, and a zeta potential of -17.10 ± 4.98 mV. The encapsulation efficiency (EE%) and drug loading (DL%) were determined to be 61.03 ± 2.40 % and 0.25 ± 0.02 % for MTX, and 81.02 ± 1.61 % and 0.23 ± 0.02 % for CuB, respectively. The formulation demonstrated substantial storage stability over a 14-day period. In vitro release studies indicated that MTX-CuB-NLC possesses sustained release capabilities. Furthermore, the nanoparticle exhibited significantly enhanced uptake and cytotoxicity against U-2 OS cells compared to the free drug. Notably, MTX-CuB-NLC displayed pronounced cytotoxic effects on methotrexate-resistant U-2 OS cells (U-2 OS/MTX), underscoring its potential to induce apoptosis and circumvent multidrug resistance in these cells. In an OS nude mouse model exhibiting drug resistance, MTX-CuB-NLC demonstrated superior tumor targeting and suppression efficacy. This research has culminated in the development of an effective continuous drug delivery system for osteosarcoma, presenting a promising strategy to combat drug resistance in this malignancy.
Collapse
Affiliation(s)
- Hao Li
- Qiqihar Medical University, Qiqihar 161006, China
| | - Tingqi Bao
- Qiqihar Medical University, Qiqihar 161006, China
| | - Xinyi Huang
- Qiqihar Medical University, Qiqihar 161006, China
| | - Jianwen Zhou
- Qiqihar Medical University, Qiqihar 161006, China
| | - Ziyu Zhang
- The First Affiliated Hospital of Qiqihar Medical University, Qiqihar 161042, China
| | - Xuefeng Wang
- The First Affiliated Hospital of Qiqihar Medical University, Qiqihar 161042, China
| | - Weifu You
- The First Affiliated Hospital of Qiqihar Medical University, Qiqihar 161042, China
| | - Lixin Cao
- The First Affiliated Hospital of Qiqihar Medical University, Qiqihar 161042, China.
| | - Cuiyan Han
- Qiqihar Medical University, Qiqihar 161006, China.
| |
Collapse
|
7
|
Park SH. Recent Research on the Role of Phytochemicals from Ginseng in Management of Osteosarcoma, Osteoporosis, and Osteoarthritis. Nutrients 2025; 17:1910. [PMID: 40507179 PMCID: PMC12158031 DOI: 10.3390/nu17111910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2025] [Revised: 05/30/2025] [Accepted: 05/31/2025] [Indexed: 06/16/2025] Open
Abstract
Ginseng phytochemicals have attracted considerable attention for their potential therapeutic applications in bone-related diseases including osteosarcoma, osteoporosis, and osteoarthritis. Recent research has highlighted the promising effects of ginsenosides and polysaccharides from ginseng by studying multi-target effects and combination therapies in osteosarcoma progression. Beyond osteosarcoma, ginseng phytochemicals have been explored for their effects on osteoporosis. Various ginsenosides and ginseng extract were shown to regulate signaling pathways involved in activating osteoblast and inhibiting osteoclast in vitro and in vivo models. Ginseng ginsenosides have also demonstrated potential anti-osteoarthritic properties. Recent studies discussed how ginsenoside reduced inflammation and cartilage degradation as a therapeutic candidate for osteoarthritis management. In this review, we examine the anti-osteosarcoma, anti-osteoporotic, and anti-osteoarthritic activities of ginseng-derived phytochemicals reported in studies published between 2014 and 2024. This review also provides a comprehensive overview of the working mechanisms of these compounds in various model systems. Furthermore, we address the limitations of current research approaches and outline future directions to maximize the therapeutic application of ginseng phytochemicals in the management of bone-related diseases.
Collapse
Affiliation(s)
- See-Hyoung Park
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
8
|
Wang Y, Jan H, Zhong Z, Zhou L, Teng K, Chen Y, Xu J, Xie D, Chen D, Xu J, Qin L, Tuan RS, Li ZA. Multiscale metal-based nanocomposites for bone and joint disease therapies. Mater Today Bio 2025; 32:101773. [PMID: 40290898 PMCID: PMC12033929 DOI: 10.1016/j.mtbio.2025.101773] [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: 03/09/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Bone and joint diseases are debilitating conditions that can result in significant functional impairment or even permanent disability. Multiscale metal-based nanocomposites, which integrate hierarchical structures ranging from the nanoscale to the macroscale, have emerged as a promising solution to this challenge. These materials combine the unique properties of metal-based nanoparticles (MNPs), such as enzyme-like activities, stimuli responsiveness, and photothermal conversion, with advanced manufacturing techniques, such as 3D printing and biohybrid systems. The integration of MNPs within polymer or ceramic matrices offers a degree of control over the mechanical strength, antimicrobial efficacy, and the manner of drug delivery, whilst concomitantly promoting the processes of osteogenesis and chondrogenesis. This review highlights breakthroughs in stimulus-responsive MNPs (e.g., photo-, magnetically-, or pH-activated systems) for on-demand therapy and their integration with biocomposite hybrids containing cells or extracellular vesicles to mimic the native tissue microenvironment. The applications of these composites are extensive, ranging from bone defects, infections, tumors, to degenerative joint diseases. The review emphasizes the enhanced load-bearing capacity, bioactivity, and tissue integration that can be achieved through hierarchical designs. Notwithstanding the potential of these applications, significant barriers to progress persist, including challenges related to long-term biocompatibility, regulatory hurdles, and scalable manufacturing. Finally, we propose future directions, including machine learning-guided design and patient-specific biomanufacturing to accelerate clinical translation. Multiscale metal-based nanocomposites, which bridge nanoscale innovations with macroscale functionality, are a revolutionary force in the field of biomedical engineering, providing personalized regenerative solutions for bone and joint diseases.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Hasnain Jan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region of China
| | - Zheng Zhong
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Kexin Teng
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
| | - Ye Chen
- Department of Chemistry, Faculty of Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Denghui Xie
- Department of Orthopedic Surgery, Center for Orthopedic Surgery, and Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, China
| | - Dexin Chen
- Institute of Advanced Wear & Corrosion Resistant and Functional Materials, Jinan University, Guangzhou, 510632, China
| | - Jiake Xu
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, Faculty of Medicine, and Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Rocky S. Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, NT, Hong Kong Special Administrative Region of China
- Institute for Tissue Engineering and Regenerative Medicine, and School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region of China
- Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, NT, Hong Kong Special Administrative Region of China
| |
Collapse
|
9
|
Guder WK, Engel NM, Hardes J, Podleska LE, Andreou D, Nottrott M, Streitbürger A. Limb salvage and complication management after (sub-) total humerus resection for primary malignant bone tumors in early childhood. J Orthop Surg Res 2025; 20:534. [PMID: 40426222 PMCID: PMC12117968 DOI: 10.1186/s13018-025-05956-0] [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: 02/03/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025] Open
Abstract
Limb-salvage after (sub-) total humerus resection in skeletally immature patients, aged less than 10 years, remains the domain of individual case decisions due to a small number of affected patients. Seven children aged ≤ 10 years at the time of (sub-) total bone sarcoma resection of the humerus between 2018 and 2023 were included in this retrospective study. Limb salvage with an intact motor function of the lower arm was achieved in 100% of cases at a mean follow-up of 89.8 months. Reconstruction survival was 28.6%. 61.8% (n = 8/13) of complications necessitated revision operations (n = 7). Distal instability and dislocation (n = 5) was the most frequently observed complication, followed by periprosthetic infection (n = 2). However, both fixed hinge prostheses and hemiprostheses with a highly cancellous implant body led to stable reconstructions of the elbow joint. Arm lengthening (5 cm) was successful in two patients who underwent implantation of a growing prosthesis. Patients developed a handedness contralateral to the operated arm in this study. The mean MSTS and TESS scores were 16.6 and 51.5. Patients suffered from a limited range of motion in the glenohumeral joint, limited fine motor function and strength despite an intact innervation of the executing muscles. Salvage of even short segments of the distal humerus has a high potential to decrease complication rates, whenever oncologically feasible. The use of fixed-hinge total humerus (growing) prostheses and custom-made total humerus hemiprostheses with a highly cancellous implant body showed the most promising results in terms of complications and functional outcomes. Arm lengthening using growing prostheses - albeit of a mostly cosmetic value - is an option in the upper extremity whenever the soft tissue coverage of the implant is adequate.
Collapse
Affiliation(s)
- Wiebke K Guder
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Nina M Engel
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Jendrik Hardes
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Lars E Podleska
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Dimosthenis Andreou
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Markus Nottrott
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Arne Streitbürger
- Department of Orthopedic Oncology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
10
|
Yin L, Xu P, Huang Y, Gu X, Sun L, Zhou H, Zhou W, Xie C, Fan Q. Glutathione-Responsive Near-Infrared-II Fluorescence Probe for Early and Accurate Detection of In Situ and Metastatic Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503257. [PMID: 40434227 DOI: 10.1002/smll.202503257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/14/2025] [Indexed: 05/29/2025]
Abstract
In situ and metastatic malignant tumors are primary diseases that threaten human life. Among all the metastases, liver metastasis is the most difficult to detect. As most imaging probes have high liver accumulation, it is difficult to distinguish tiny metastases from normal liver tissue with strong background signal. In this study, the design of a novel second near-infrared window (NIR-II) fluorescence probe for precise detection of carcinoma in situ and liver metastases is presented. The probe called Tg-RGD utilizes a commercially available cyanine dye IR-806 as the signaling moiety, a disulfide bond linker as the responsive moiety, an RGD-capped poly(ethylene glycol) (PEG) as the water soluble enhancer, and the tumor targeting moiety. Tg-RGD shows good glutathione (GSH) responsiveness and selectivity, where its NIR-II fluorescence intensity can enhance 50-fold after activation. In vivo study indicates that Tg-RGD shows much better imaging and targeting effects than Tg-PEG with a similar structure but without RGD moiety for both orthotopic breast cancer and osteosarcoma. Most importantly, Tg-RGD can detect tiny liver metastases with high signal-to-background ratio (3.2). Thus, this study reports a high-performance tumor-specific NIR-II fluorescence probe for in situ and tiny metastatic tumor detection, and may further broaden the applications into related tumor lesions.
Collapse
Affiliation(s)
- Likun Yin
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Pu Xu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Yuxin Huang
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Xuxuan Gu
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Liwen Sun
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Hui Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Wen Zhou
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Chen Xie
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| | - Quli Fan
- State Key Laboratory of Flexible Electronics (LoFE) & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, China
| |
Collapse
|
11
|
Lian Q, Liu H, Li J, Luo C, Liu C, Zhao H, Dai P, Wang B, Zhou H, Jiang X, Wang Z, Qiao S. Enhancing radiosensitivity of osteosarcoma by ITGB3 knockdown: a mechanism linked to enhanced osteogenic differentiation status through JNK/c-JUN/RUNX2 pathway activation. J Exp Clin Cancer Res 2025; 44:159. [PMID: 40410897 PMCID: PMC12102912 DOI: 10.1186/s13046-025-03417-4] [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] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 05/11/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND The prognosis of osteosarcoma has improved little over the past few decades, with radioresistance being a contributing factor. Effective radiosensitizing targets and novel mechanisms for treating osteosarcoma are urgently needed. Research on the impact of regulating differentiation levels on the radiosensitivity of malignant tumors is limited. This study aimed to explore the efficacy of ITGB3 as a novel radiosensitizing target in osteosarcoma and to explore whether the modulation of osteogenic differentiation plays a role in mediating the radiosensitizing effect. METHODS RNA sequencing was utilized to screen for potential targets that affect the radiosensitivity of osteosarcoma. In vitro assays examining cell viability, apoptosis, proliferation, migration, and invasion were conducted to verify the radiosensitizing effect of ITGB3-knockdown (KD). Furthermore, in vivo validation was performed by constructing mouse models with subcutaneous and orthotopic tibial tumors. Rescue experiments involving siRNAs and molecular inhibitors were performed to explore and validate the mechanisms through which ITGB3-KD exerts a radiosensitizing effect in vitro and in vivo. Additionally, osteogenic differentiation cultures of osteosarcoma cells were conducted as auxiliary validation for the radiosensitizing mechanism. RESULTS ITGB3-KD had a radiosensitizing effect on osteosarcoma in vitro by inhibiting cell viability, proliferation, migration, and invasion and promoting apoptosis. ITGB3-KD radiosensitized osteosarcoma in vivo in subcutaneous and orthotopic tibial tumor models. ITGB3-KD upregulated the JNK/c-JUN pathway, and rescue experiments with a JNK inhibitor revealed that the activation of this pathway was crucial for the upregulation of osteogenic markers such as RUNX2, OCN, and OPN, as well as for promoting apoptotic pathways. siRNA-based rescue experiments indicated that the upregulation of RUNX2 mediated the proapoptotic radiosensitizing effects of ITGB3-KD. Culture in osteogenic differentiation medium promoted osteosarcoma radiosensitization by enhancing the osteogenic differentiation status, working synergistically with ITGB3-KD. CONCLUSIONS Our findings indicate that ITGB3-KD enhances radiosensitivity in osteosarcoma by promoting osteogenic differentiation and apoptosis through activation of the JNK/c-JUN/RUNX2 pathway, identifying ITGB3 as a candidate therapeutic target and implicating JNK/c-JUN/RUNX2 signaling as a modulatory axis for improving the response to radiation of osteosarcoma.
Collapse
Affiliation(s)
- Qiujian Lian
- Department of Orthopedics, Fuzhou Second General Hospital, Fuzhou, Fujian, 350007, China
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Hu Liu
- Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jingyan Li
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Cheng Luo
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Chang Liu
- Department of Orthopedics, The 900th Hospital of Joint Logistic Support Force, Fuzhou, Fujian, 350025, China
| | - Haonan Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Peijun Dai
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Bingxuan Wang
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Huipeng Zhou
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China
| | - Xin Jiang
- Department of Anesthesiology, The Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Zhiwei Wang
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China.
| | - Suchi Qiao
- Department of Orthopedics, The Third Affiliated Hospital of Naval Medical University, Shanghai, 201805, China.
| |
Collapse
|
12
|
Huang T, Chen Y, Zhao Q, Wu X, Li H, Luo X, Su Y, Zhang S, Liu P, Tang N. Dual Regulation of Sprouty 4 Palmitoylation by ZDHHC7 and Palmitoyl-Protein Thioesterase 1: A Potential Therapeutic Strategy for Cisplatin-Resistant Osteosarcoma. RESEARCH (WASHINGTON, D.C.) 2025; 8:0708. [PMID: 40416361 PMCID: PMC12099059 DOI: 10.34133/research.0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/27/2025] [Accepted: 04/27/2025] [Indexed: 05/27/2025]
Abstract
Background: Osteosarcoma (OS) is a primary malignant bone tumor predominantly affecting adolescents. Chemotherapeutic agents, such as cisplatin, are commonly used in OS treatment; however, drug resistance markedly undermines treatment efficacy and contributes to reduced patient survival. The mechanisms underlying cisplatin resistance remain poorly understood. Recently, palmitoyl-protein thioesterase 1 (PPT1), a depalmitoylation enzyme, has attracted attention for its role in tumorigenesis and drug resistance. Investigating the mechanisms of PPT1 may offer new strategies to overcome resistance. Methods: This study analyzed multiple Gene Expression Omnibus datasets and utilized the OncoPredict tool to demonstrate the elevated expression of PPT1 in OS and its critical role in cisplatin resistance. By combining single-cell analysis with in vitro and in vivo experiments, we explored how PPT1 influences OS development through depalmitoylation and assessed the antitumor effects of the PPT1 inhibitor Ezurpimtrostat (GNS561), as well as its synergistic effects when combined with cisplatin. Results: We demonstrated that Sprouty 4 (SPRY4) undergoes a dynamic palmitoylation cycle regulated by zinc finger DHHC-type palmitoyl transferase 7 (ZDHHC7) and PPT1, which modulates mitogen-activated protein kinase (MAPK) signaling and subsequently affects tumor cell proliferation, migration, apoptosis, and drug resistance. Further validation confirmed the effectiveness of the PPT1 inhibitor GNS561 in overcoming cisplatin resistance. Notably, GNS561 exhibited a significant synergistic effect when used in combination with cisplatin, greatly enhancing the sensitivity of cisplatin-resistant cells. Conclusion: This study highlights the pivotal role of PPT1 in OS resistance mechanisms. PPT1 and ZDHHC7 regulate SPRY4 through a dynamic palmitoylation-depalmitoylation cycle that modulates MAPK signaling activation and contributes to OS cell proliferation, migration, and drug resistance. As a PPT1 inhibitor, GNS561 not only inhibits OS cell proliferation but also demonstrates synergistic effects with cisplatin, significantly enhancing cisplatin sensitivity in resistant cells and promoting apoptosis. Our findings offer a novel approach for targeting PPT1 in therapeutic strategies. GNS561 holds promise as an adjunctive therapy when combined with cisplatin, potentially overcoming resistance and improving efficacy, thereby enhancing the prognosis for OS patients. Future studies should further investigate the clinical potential of GNS561 and optimize OS treatment strategies.
Collapse
Affiliation(s)
- Tianlong Huang
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yifan Chen
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiangqiang Zhao
- Department of Hematology,
Liuzhou People’s Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
- Department of Hematology,
The Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Xin Wu
- Department of Spine Surgery, Third Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Hongxing Li
- Department of Orthopaedic,
The Central Hospital of Shaoyang, Shaoyang, China
- Changsha Medical University, Changsha, Hunan, China
| | - Xin Luo
- Orthopaedic Department,
The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Su
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shengqun Zhang
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pan Liu
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ning Tang
- Orthopaedic Department,
The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Orthopaedic Department,
The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Dkhar DS, Mahapatra S, Chandra P. An electrochemically charged nanoengineered bioelectronic immunosensing device for osteopontin detection in serum samples. NANOSCALE 2025. [PMID: 40405794 DOI: 10.1039/d5nr00253b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Osteopontin (OPN) is a crucial biomarker for osteosarcoma, an aggressive bone cancer. Elevated levels of OPN are found in osteosarcoma tissues and blood samples, associated with tumor growth, metastasis, and poor prognosis. The present electrochemical voltammetric detection methods are mainly assisted by redox couple/chemical mediators in a measuring solution. However, in resource-confined settings, such a detection strategy undermines the potential for the development of precise and easy-to-use point-of-care bioelectronic devices. To simplify the detection method, herein, we present a reliable and straightforward clinically deployable immunosensor via engineering an electrochemically charged surface that does not require mediators in the solution phase. Such an electroactive surface offers a simplistic label-free detection of the protein OPN, requiring only the use of a buffer solution to obtain the signals. The electroactive immunosensor probe was engineered using electrodeposited gold, bimetallic FeGdHCF redox nanoparticles, and oxidized graphene nanoplatelets for immobilizing the OPN antibodies. Since the detection is precisely dependent on the redox-active electrode surface for obtaining analytical signals, this strategy was further designed into a miniaturized device cascade for the direct and on-site detection of OPN. The immunosensor probe showed a low LOD of 0.437 (±0.002) (RSD < 4.09%, n = 5) pg mL-1 and a wide LDR of 5 × 102 to 2 × 106 pg mL-1. It was applied to detect OPN in serum samples and was also evaluated for its selectivity against various molecules coexisting in real clinical samples and was found to be stable for a period of six weeks.
Collapse
Affiliation(s)
- Daphika S Dkhar
- Laboratory of Bio-Physio Sensors and Nano-bioengineering, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India.
| | - Supratim Mahapatra
- Laboratory of Bio-Physio Sensors and Nano-bioengineering, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India.
| | - Pranjal Chandra
- Laboratory of Bio-Physio Sensors and Nano-bioengineering, School of Biochemical Engineering, Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh 221005, India.
| |
Collapse
|
14
|
Nair JJ, van Staden J. Cytotoxic lycorine alkaloids of the plant family Amaryllidaceae. Bioorg Chem 2025; 163:108619. [PMID: 40516169 DOI: 10.1016/j.bioorg.2025.108619] [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: 03/31/2025] [Revised: 05/14/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
The plant family Amaryllidaceae is embellished with a diverse array of antiproliferative alkaloid principles. Chief amongst these are the lycorine alkaloids, which have attracted considerable attention as potential anticancer drugs. This account tracks developments in the field with these substances encompassing the years 2015-2019. Twenty-nine compounds were screened against nearly eighty cancer cell lines representing seventeen different types of cancer. Submicromolar level activities were recorded against leukemia, myeloma and breast cancer cells. The response of lycorine (IC50 0.6 μM) to HL-60 myeloid leukemia cells was particularly striking. Promising activities were also documented from in vivo models of brain, lung, colon, prostate and breast cancer cells (in the 5-10 mg/kg/day dosage range). The screenings indicated these compounds to be efficacious without attendant detrimental effects towards control animals. Structure-activity relationship studies afforded useful insight to the elements of the anticancer pharmacophore, such as the necessity for the A-ring methylenedioxy and C-ring hydroxy functionalities. The mechanisms of action were intensively examined, with over twenty individual areas identified wherein such probes have been made. Of prominence here was the apoptosis-inducing abilities of lycorine against (amongst others) leukemia, pancreatic, bladder, liver and bone cancer cells, involving the modulation of key mediators such as caspase-3, p53, PARP, Bax and Bcl-2. Useful insights also emerged from docking studies undertaken with various cancer-related proteins, such as VEGF, HDAC, PI3Kα, c-Met kinase and EGFR. The lycorine alkaloids have proved to be highly versatile entities, readily embracing multiple facets of anticancer drug discovery.
Collapse
Affiliation(s)
- Jerald J Nair
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Private Bag X01, Scottsville 3209, South Africa
| | - Johannes van Staden
- Research Centre for Plant Growth and Development, School of Life Sciences, University of KwaZulu-Natal Pietermaritzburg, Private Bag X01, Scottsville 3209, South Africa.
| |
Collapse
|
15
|
Shen W, Ma Y, Yang C, Yan S, Ye K. Role of N6-methyladenosine methyltransferase component RBM15 in cancer progression and its therapeutic potential. Discov Oncol 2025; 16:855. [PMID: 40402374 PMCID: PMC12098234 DOI: 10.1007/s12672-025-02644-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/09/2025] [Indexed: 05/23/2025] Open
Abstract
Cancer ranks as a primary cause of mortality globally, and the study of its molecular markers and regulatory mechanisms holds paramount importance. N6-methyladenosine (m⁶A) represents the predominant modification in messenger RNA (mRNA), influencing key biological processes including RNA stability, splicing, and translation. The dynamic modulation of m⁶A modification is mediated by an array of enzymes comprising methyltransferases ("writers"), demethylases ("erasers"), and m⁶A-binding proteins ("readers").As a pivotal member of the m⁶A "writer" family, RNA binding motif protein 15 (RBM15) facilitates the recruitment of the methyltransferase complex (MTC) to mRNA, thus orchestrating the addition of m⁶A modifications. Although prior research has underscored the critical role of m⁶A in oncogenesis, the precise mechanisms through which RBM15 operates in cancer are yet to be elucidated. This study endeavors to elucidate the structural characteristics and functional roles of RBM15, investigate its potential regulatory mechanisms across diverse tumors, uncover its distinct functions in tumor genesis, progression, and metastasis, and evaluate the therapeutic potential of targeting RBM15 in cancer treatment.
Collapse
Affiliation(s)
- Wenxiang Shen
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yulong Ma
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Chunwang Yang
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Shishun Yan
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Kaishan Ye
- Department of Orthopedics, Second Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory of Bone and Joint Diseases of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
16
|
McGee LE, Pereira JS, McEachron TA, Mazcko C, LeBlanc AK, Beck JA. The tumor microenvironment of metastatic osteosarcoma in the human and canine lung. Commun Biol 2025; 8:756. [PMID: 40374715 DOI: 10.1038/s42003-025-07992-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/24/2025] [Indexed: 05/17/2025] Open
Abstract
Osteosarcoma is a rare but aggressive bone tumor that develops spontaneously in human and canine patients and most commonly metastasizes to the lung. The presence of lung metastases significantly decreases the survival rate of patients, with minimal benefit seen with available treatments. Canine osteosarcoma is clinically and molecularly similar to human osteosarcoma and develops approximately ten times more frequently than human osteosarcoma making dogs a promising natural model to study disease progression. The development of new therapies for pulmonary metastases requires an understanding of the interplay between tissue resident cells as well as recruited cell types and how those interactions impact seeding and progression within the new metastatic site. This review explores the tumor microenvironment surrounding pulmonary metastases and how current knowledge in canine and human patients can inform better treatments and outcomes for both populations.
Collapse
Affiliation(s)
- L E McGee
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - J S Pereira
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - T A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - C Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - A K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - J A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
17
|
Zhang J, Wang W. Glypican-3 regulated epithelial mesenchymal transformation-related genes in osteosarcoma: based on comprehensive tumor microenvironment profiling. Front Immunol 2025; 16:1566061. [PMID: 40433364 PMCID: PMC12106470 DOI: 10.3389/fimmu.2025.1566061] [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: 01/24/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Osteosarcoma (OS) is the most common primary bone malignancy, predominantly affecting children and adolescents. Current treatment approaches have limited efficacy, with a 5-year survival rate of approximately 60%. Epithelial-mesenchymal transition (EMT) plays a key role in the onset, progression, and metastasis of OS, potentially influencing patient prognosis. Methods We screened EMT-related genes from multiple transcriptomic datasets of OS and performed unsupervised consensus clustering of EMT-related gene sets. Key EMT-related genes were identified using weighted gene co-expression network analysis (WGCNA) and intersected with differentially expressed genes (DEGs) between OS and normal tissue samples. The least absolute shrinkage and selection operator (LASSO) algorithm was applied to screen candidate genes for developing a prognostic model. Single-cell RNA-Seq (scRNA-Seq) analysis was conducted on OS samples to identify cell populations expressing model genes. Functional validation was performed using si-GPC3 in the MG-63 cell line. Results The EMT-based prognostic model demonstrated strong predictive capacity across several validation cohorts. The model effectively predicted immune-related features and immunotherapy responses in high-risk and low-risk patient groups. Seven primary cell types were identified from scRNA-Seq data of OS samples, with the osteoblast population showing the highest proportion of cells positive for model genes. The OS_C3 subpopulation exhibited significantly higher scores and included nine gene modules associated with metabolism, structural integrity, proliferation, differentiation, adhesion, migration, immune responses, inflammatory reactions, and signal transduction. The model genes also demonstrated prognostic value across various cancer types. Knockdown of GPC3 in MG-63 cells resulted in decreased proliferation and migration ability. Conclusion This study provides new insights into the potential mechanisms of EMT in OS and its impact on the tumor immune microenvironment and response to immunotherapy. These findings may pave the way for novel personalized treatment strategies for OS patients.
Collapse
Affiliation(s)
| | - Wei Wang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Liaoning, Shenyang, China
| |
Collapse
|
18
|
Song X, Chen S, Cheng J, Li H, Wu R, Yan M, Wang M, Li J, Jin A, Wang W. Screening and identification Hub genes associated with immune cell infiltration and critical biomarkers in osteosarcoma. Mol Cell Probes 2025:102031. [PMID: 40374042 DOI: 10.1016/j.mcp.2025.102031] [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: 01/30/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
PURPOSE Osteosarcoma (OS) exhibits limited immune cell infiltration that directly contributes to poor prognosis. This study sought to screen and identify pivotal biomarkers of OS immune infiltration and early diagnosis of OS. METHODS The immune cell infiltration profiles with transcriptome sequencing data from 88 OS samples were explored with CIBERSORT algorithm. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Protein-protein interaction (PPI) network analyses were applied to identify hub genes, with the expressions confirmed by dual immunofluorescence in 50 OS samples. The new biomarker gene HTRA1 were examined by immunohistochemistry and validated by the Immune score and immune gene expression profile analyses. The impact of HTRA1 on OS prognosis was verified by Least absolute shrinkage and selection operator (LASSO) regression analysis. The biological effect of HTRA1 was characterized in MG63 cells. RESULT CD8+ T cells, activated memory CD4+ T cells and plasma cells were positively correlated with the prognosis of OS. Hub genes CCL5, CXCL9, CXCL13, and HTRA1, exhibited positive correlation with the infiltration of both CD8+ T cells and CD4+ T cells. HTRA1 expression was reduced in osteosarcoma tissues, which was positively correlated with immune scores and the expressions of immune-related genes. High levels of HTRA1 were associated with favorable OS prognosis, and could negatively impacted MG63 malignant characteristics. CONCLUSION CCL5, CXCL9, CXCL13, and HTRA1 were OS hub genes positively correlate with CD8+ T cell and CD4+ T cell infiltrations. HTRA1 can serve as an underlying biomarker for the prognosis and immunotherapy of OS.
Collapse
Affiliation(s)
- Xin Song
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China; Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Sihao Chen
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Junning Cheng
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou,311399, China
| | - Haiyu Li
- Institute of Intelligent Chinese Medicine,Chongqing University of Chinese Medicine,Chongqing 402760, China
| | - Ruixin Wu
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Min Yan
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Min Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University.Chongqing,400038, China
| | - Aishun Jin
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| | - Wang Wang
- Department of Immunology, College of Bassic Medicine; Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention,Chongqing Medical University, Chongqing,400010, China
| |
Collapse
|
19
|
Bishop MW. Osteosarcoma: Diagnosis, Treatment, and Emerging Opportunities. Hematol Oncol Clin North Am 2025:S0889-8588(25)00043-7. [PMID: 40368742 DOI: 10.1016/j.hoc.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Osteosarcoma is the most common primary bone malignancy of childhood and adolescence. Despite advances in cures for many pediatric neoplasms, outcomes for osteosarcoma have not significantly changed for nearly 4 decades and treatment is similar for patients with both standard and higher risk disease. Recent progress has been demonstrated with evidence of activity of multitargeted tyrosine kinase inhibitors for patients with advanced osteosarcoma; further study is needed to evaluate their use for newly diagnosed patients. Novel approaches seek to exploit common cell surface antigens (LRRC15 and B7-H3) as well as aberrant DNA repair mechanisms and cell cycle checkpoints.
Collapse
Affiliation(s)
- Michael W Bishop
- Department of Pediatrics, Section of Hematology/Oncology, Arkansas Children's Hospital, 1 Children's Way, Slot# 512-10, Little Rock, AR 72202, USA.
| |
Collapse
|
20
|
Zhang Y, Zhou Y, Zhang S, Zhang X, Li J, Zhan XK. miR-486-3p Suppresses Osteosarcoma Proliferation and Migration by Targeting the SPRED1-MAPK/ERK Pathway. Biochem Genet 2025:10.1007/s10528-025-11128-w. [PMID: 40358892 DOI: 10.1007/s10528-025-11128-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
Osteosarcoma (OS) is a common malignancy of the bone that originates from stromal cell lines. One of the key cellular pathways extensively studied in OS is the mitogen-activated protein kinase (MAPK) pathway, particularly ERK1/2, whose activation is closely associated with tumor growth and metastasis. MicroRNA (miRNA)-based detection and targeted therapies offer promising new strategies for the treatment of OS. In this study, we investigated the role of miR‑486‑3p in the regulation of the ERK1/2 pathway in OS. We examined the expression level of miR‑486‑3p in the GEO dataset (GSE65071) and clinical samples, and analyzed its regulation of the target gene SPRED1 in OS cells and tumor-bearing mice. Downregulation of miR‑486‑3p was confirmed in OS tissues, with its expression decreasing in line with the progression of clinical stages. Furthermore, the exogenous introduction of a miR-486-3p mimic attenuated the malignant behavior of OS cells, inhibiting their proliferation, migration, and invasion. Bioinformatic analysis revealed that miR‑486‑3p directly targets SPRED1 in OS, leading to alterations in epithelial-to-mesenchymal transition (EMT) markers, including E-cadherin, N-cadherin, and Vimentin. Functional loss- and gain-of-function experiments confirmed that miR‑486‑3p directly targets SPRED1 and inactivates the ERK1/2 pathway in both OS cells and tumor-bearing mice. This review demonstrates that downregulation of miR-486-3p leads to increased SPRED1 expression, which activates the ERK1/2 pathway in OS. Targeting miR-486-3p and SPRED1 could offer potential therapeutic benefits.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Orthopaedics, The First People's Hospital of Chengdu, Chengdu, Sichuan Province, China
| | - Yi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Sen Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiaoying Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Jiaxin Li
- Department of Gastrointestinal Surgery, Lishui Hospital Affiliated of Wenzhou Medical University & Lishui City People's Hospital, Lishui, Zhejiang Province, China.
| | - Xiao-Kai Zhan
- Department of Hematology and Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, P.R. China.
| |
Collapse
|
21
|
Alrouji M, Alshammari MS, Anwar S, Venkatesan K, Shamsi A. Mechanistic Roles of Transcriptional Cyclin-Dependent Kinases in Oncogenesis: Implications for Cancer Therapy. Cancers (Basel) 2025; 17:1554. [PMID: 40361480 PMCID: PMC12071579 DOI: 10.3390/cancers17091554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Cyclin-dependent kinases (CDKs) are pivotal in regulating cell cycle progression and transcription, making them crucial targets in cancer research. The two types of CDKs that regulate different biological activities are transcription-associated CDKs (e.g., CDK7, 8, 9, 12, and 13) and cell cycle-associated CDKs (e.g., CDK1, 2, 4, and 6). One characteristic of cancer is the dysregulation of CDK activity, which results in unchecked cell division and tumor expansion. Targeting transcriptional CDKs, which control RNA polymerase II activity and gene expression essential for cancer cell survival, has shown promise as a therapeutic approach in recent research. While research into selective inhibitors for transcriptional CDKs is ongoing, inhibitors that target CDK4/6, such as palbociclib and ribociclib, have demonstrated encouraging outcomes in treating breast cancer. CDK7, CDK8, and CDK9 are desirable targets for therapy since they have shown oncogenic roles in a variety of cancer types, such as colorectal, ovarian, and breast malignancies. Even with significant advancements, creating selective inhibitors with negligible off-target effects is still difficult. This review highlights the need for more research to optimize therapeutic strategies and improve patient outcomes by giving a thorough overview of the non-transcriptional roles of CDKs in cancer biology, their therapeutic potential, and the difficulties in targeting these kinases for cancer treatment.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Mohammed S. Alshammari
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia;
| | - Saleha Anwar
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, Saudi Arabia
| |
Collapse
|
22
|
Bhuia MS, Chowdhury R, Afroz M, Akbor MS, Al Hasan MS, Ferdous J, Hasan R, de Alencar MVOB, Mubarak MS, Islam MT. Therapeutic Efficacy Studies on the Monoterpenoid Hinokitiol in the Treatment of Different Types of Cancer. Chem Biodivers 2025; 22:e202401904. [PMID: 39776341 DOI: 10.1002/cbdv.202401904] [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: 08/06/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
Hinokitiol (HK), a monoterpenoid that naturally occurs in plants belonging to the Cupressaceae family, possesses important biological activities, including an anticancer effect. This review summarizes its anticancer potential and draws possible molecular interventions. In addition, it evaluates the biopharmaceutical, toxicological properties, and clinical application of HK to establish its viability for future advancement as a dependable anticancer medication. The assessment is based on the most recent information available from various databases. Findings demonstrate that HK possesses substantial therapeutic advantages against diverse types of cancer (colon, cervical, breast, bone, endometrial, liver, prostate, oral, and skin) through various molecular mechanisms. HK induces oxidative stress, cytotoxicity, apoptosis, cell-cycle arrest at the G and S phases, and autophagy through modulation of phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), p38/ERK/MAPK, nuclear factor kappa B, and c-Jun N-terminal kinase signaling pathways. Furthermore, this compound exhibits good oral bioavailability with excellent plasma clearance. Clinical uses of HK demonstrate therapeutic advantages without any significant negative effects. A thorough study of the pertinent data suggests that HK may serve as a viable candidate for developing novel cancer therapies. Consequently, more extensive studies are necessary to evaluate its cancer treatment efficacy, safety, and possible long-term hazards.
Collapse
Affiliation(s)
- Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Meher Afroz
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Showkot Akbor
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
| | - Rubel Hasan
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
| | | | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Dhaka, Bangladesh
- Phytochemistry and Biodiversity Research Laboratory, BioLuster Research Center Ltd, Gopalganj, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, Dhaka, Bangladesh
| |
Collapse
|
23
|
Yang L, Zhang J, Jiang Y, Zhang J, Wang Z, Wang L, Fan X, Ba G. Identifying KDM5B as the synthetic lethal target of KMT2D-mutated osteosarcoma. Chem Biol Interact 2025; 412:111451. [PMID: 40054828 DOI: 10.1016/j.cbi.2025.111451] [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: 10/28/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Osteosarcoma (OS) is a malignant bone tumor that occurs commonly in adolescents or children, previous studies have shown its complex epigenetic signature. Histone methyltransferases KMT2D loss-of-function mutation is common in various types of human cancer. Here we revealed that KMT2D loss promotes malignant phenotypes in osteosarcoma. Based on the result of epigenetic inhibitor library screening we discovered that KDM5B inhibitors selectively killed KMT2D-deficient cells. Also, the knockdown of KDM5B by shRNA could reduce cell proliferation, migration and induce apoptosis in KMT2D-KO cells, while no similar appearance was observed in wild-type cells. Furthermore, we testified the efficiency and safety of KDM5B inhibition in patient-derived xenografts (PDX) mouse models driven by KMT2D low-expressing patients. These results demonstrated KDM5B as a synthetic lethal factor of KMT2D-loss mutation. Our findings suggest a novel therapeutic strategy for treating KMT2D mutated osteosarcoma by targeting KDM5B.
Collapse
Affiliation(s)
- Liyu Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Jing Zhang
- Department of Bone & Soft tissue Oncology, People's Hospital of China Medical University (People's Hospital of Liaoning Province), Shenyang, Liaoning Province, China.
| | - Yiting Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jiayu Zhang
- Women and Children's Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Zhonghua Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Lihui Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China; Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Benxi, 117004, China.
| | - Xinyu Fan
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Gen Ba
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
24
|
Ma Y, Lai P, Sha Z, Li B, Wu J, Zhou X, He C, Ma X. TME-responsive nanocomposite hydrogel with targeted capacity for enhanced synergistic chemoimmunotherapy of MYC-amplified osteosarcoma. Bioact Mater 2025; 47:83-99. [PMID: 39897587 PMCID: PMC11783017 DOI: 10.1016/j.bioactmat.2025.01.006] [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: 08/24/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
The oncogene MYC is one of the most commonly activated oncogenic proteins in human tumors, with nearly one-fourth of osteosarcoma showing MYC amplification and exhibiting the worst clinical outcomes. The clinical efficacy of single radiotherapy, chemotherapy, and immunotherapy for such osteosarcoma is poor, and the dysregulation of MYC amplification and immune-suppressive tumor microenvironment (TME) may be potential causes of anti-tumor failure. To address the above issues, we developed an injectable TME-responsive nanocomposite hydrogel to simultaneously deliver an effective MYC inhibitor (NHWD-870) and IL11Rα-targeted liposomes containing cisplatin-loaded MnO2 (Cis/Mn@Lipo-IL11). After in situ administration, NHWD-870 effectively degrades MYC and downregulates CCL2 and IL13 cytokines to trigger M1 type activation of macrophages. Meanwhile, targeted delivery of Cis/Mn@Lipo-IL11 reacts with excess intratumoral GSH to generate Mn2+ and thus inducing excess active oxygen species (ROS) production through Fenton-like reaction, along with cisplatin, thereby inducing immunogenic cell death (ICD) to promote dendritic cell maturation. Through synergistic regulation of MYC and ICD levels, the immune microenvironment was reshaped to enhance immune infiltration. In the osteosarcoma-bearing model, the nanocomposite hydrogel significantly enhanced tumor T cell infiltration, induced effective anti-tumor immunity and attenuated lung metastasis. Therefore, our results reveal a powerful strategy for targeted combination therapy of MYC-amplified osteosarcoma.
Collapse
Affiliation(s)
- Yichao Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhou Sha
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Bing Li
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jiangpeng Wu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaojun Zhou
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Chuanglong He
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xiaojun Ma
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
25
|
Allen J, Alotaibi F, Alshamrani Y, Hesham A, Flowers A, Kim DD, Woo V. Destructive mandibular mass in a 60-year-old female. Oral Surg Oral Med Oral Pathol Oral Radiol 2025; 139:495-500. [PMID: 39638734 DOI: 10.1016/j.oooo.2024.11.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024]
Affiliation(s)
- Joshua Allen
- Department of Diagnostic Sciences, Texas A&M College of Dentistry, Dallas, TX, USA.
| | - Fawaz Alotaibi
- Department of Oral and Maxillofacial Surgery, Head and Neck Oncology and Microvascular Reconstructive Surgery, LSU Health Science Center Shreveport, Shreveport, LA, USA
| | - Yousef Alshamrani
- Department of Oral and Maxillofacial Surgery, Head and Neck Oncology and Microvascular Reconstructive Surgery, LSU Health Science Center Shreveport, Shreveport, LA, USA
| | - Abdulrahman Hesham
- Department of Oral and Maxillofacial Surgery, Head and Neck Oncology and Microvascular Reconstructive Surgery, LSU Health Science Center Shreveport, Shreveport, LA, USA
| | - Ashley Flowers
- Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport, Shreveport, LA, USA
| | - D David Kim
- Department of Oral and Maxillofacial Surgery, Head and Neck Oncology and Microvascular Reconstructive Surgery, LSU Health Science Center Shreveport, Shreveport, LA, USA
| | - Victoria Woo
- Department of Diagnostic Sciences, Texas A&M College of Dentistry, Dallas, TX, USA
| |
Collapse
|
26
|
Lin Y, Wang B. pH-responsive paclitaxel prodrug encapsulated in a polypeptide-chitosan polymer delivery system for osteosarcoma treatment. Carbohydr Res 2025; 551:109414. [PMID: 39923605 DOI: 10.1016/j.carres.2025.109414] [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: 12/27/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Osteosarcoma, a highly invasive and metastatic primary bone malignancy, remains a significant clinical challenge due to the limited improvement in overall survival despite advances in treatment strategies. This highlights the urgent need for the development of more effective therapeutic options. In response, we have developed a novel paclitaxel (PTX)-loaded nanodrug system, PLGA-CS-1@PTX, by incorporating a synthesized epoxy-tetrapeptide derivative (compound 1) with poly(lactic-co-glycolic acid) (PLGA) and chitosan (CS), forming the PLGA-CS-1 composite system. The system was thoroughly characterized for its physicochemical properties, including morphology, particle size, and in vitro release behavior. Scanning electron microscopy (SEM) confirmed the nanostructure of the particles, with particle sizes around 170 nm and a narrow PDI (<0.15), indicating a uniform distribution. In vitro release studies showed a pH-responsive release profile, with 84.8 % of PTX released at pH 5.4 after 65 h of incubation, compared to 68.1 % at pH 6.4 and 14.8 % at pH 7.4, demonstrating good drug release control in acidic environments. Biological assays demonstrated significant inhibition of osteosarcoma cell proliferation in both HOS and U2OS cell lines, with a dose-dependent reduction in SPICE1 expression, suggesting that PLGA-CS-1@PTX can effectively suppress the proliferative activity of osteosarcoma cells by modulating SPICE1 levels. The hydrophobic segment of the peptide enhanced the drug loading capacity and minimized side effects, improving the overall safety profile of the system. This composite system effectively integrates the strengths of each component, offering a promising, safe, and efficient strategy for osteosarcoma treatment with great potential for clinical application.
Collapse
Affiliation(s)
- Yunfei Lin
- Department of Orthopaedics, Peking University First Hospital, Beijing, China
| | - Bing Wang
- Department of Orthopaedics, Peking University First Hospital, Beijing, China.
| |
Collapse
|
27
|
Li S, Li Z, Wang J, Han X, Zhang L. Cardamom synergizes with cisplatin against human osteosarcoma cells by mTOR-mediated autophagy. Cancer Gene Ther 2025; 32:538-549. [PMID: 40140723 PMCID: PMC12086087 DOI: 10.1038/s41417-025-00894-9] [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] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
Cisplatin (DDP), a frontline chemotherapeutic agent in osteosarcoma (OS) treatment, is frequently paired with other compounds to enhance its therapeutic potency. Cardamom (CAR), a natural flavonoid, exhibits significant inhibitory effects on human OS cells while minimizing toxic side effects. In this study, we combined CAR and DDP to treat OS, revealing that the DDP/CAR combination synergistically inhibits the growth of human OS cells in vitro and in vivo. Network pharmacological analysis indicated that mammalian target of rapamycin (mTOR) may be an important cross-target for DDP/CAR combination. Notably, this combined treatment significantly reduced mTOR phosphorylation and elevated autophagy levels within OS cells. At the mechanistic level, the DDP/CAR regimen enhanced apoptosis and compromised the viability of OS cells by triggering autophagy. This impact was attenuated by the use of the mTOR activator MHY and the autophagy inhibitor hydroxychloroquine (HCQ). Furthermore, in DDP-resistant cell lines, CAR was able to mitigate DDP resistance by bolstering autophagy levels. In general, our results suggest that CAR bolstering autophagy levels DDP against OS cells through the induction of mTOR-mediated autophagy.
Collapse
Affiliation(s)
- Sheng Li
- Department of Respiratory Medicine, Shapingba Hospital affiliated to Chongqing University, Chongqing, China
| | - Ziyun Li
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jiayu Wang
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xueqian Han
- Key Laboratory of Clinical Laboratory Diagnostics Designated By Ministry of Education, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lulu Zhang
- Department of Clinical Laboratory, Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
28
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [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: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
29
|
Shi Y, Cui J, Xiaohan L. Vitamin B12 as a novel USP3 deubiquitinase inhibitor suppresses cell proliferation and growth in osteosarcoma. Biochem Biophys Res Commun 2025; 761:151640. [PMID: 40179740 DOI: 10.1016/j.bbrc.2025.151640] [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: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
Targeting oncogenic ubiquitin-specific proteases (USPs) has been regarded as a potential useful strategy for cancer treatment. In the current study, we aimed to screen candidate small molecule target oncogenic USPs in osteosarcoma. Initially, analysis of public datasets revealed that USP3 is significantly upregulated in osteosarcoma tissues. Functional studies further demonstrated that USP3 promotes osteosarcoma cell proliferation and tumor growth both in vitro and in vivo. Through structure-based virtual screening, we identified Vitamin B12 as a potential USP3 inhibitor. Biochemical assays confirmed that Vitamin B12 effectively suppresses USP3 activity at a level comparable to PR619, a broad-spectrum deubiquitinase inhibitor. Furthermore, Vitamin B12 mitigates the oncogenic effects of USP3 in osteosarcoma by inhibiting its deubiquitinase activity. In conclusion, our findings offer new insights into the role of USP3 in osteosarcoma and underscore the potential of Vitamin B12 as a promising therapeutic agent.
Collapse
Affiliation(s)
- Yuanzhen Shi
- Department of Foot and Ankle Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province, 250000, China
| | - Jiamin Cui
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Li Xiaohan
- Department of Foot and Ankle Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan City, Shandong Province, 250000, China.
| |
Collapse
|
30
|
Sharma A, Pettee D, Mella C, Hord C, Brockwell M, Hardy S, Ball HC, Safadi FF, Kuerbitz SJ. Epigenetic Inactivation of RIPK3-Dependent Necroptosis Augments Cisplatin Chemoresistance in Human Osteosarcoma. Int J Mol Sci 2025; 26:3863. [PMID: 40332549 PMCID: PMC12027565 DOI: 10.3390/ijms26083863] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy in children and adolescents. Unfortunately, drug resistance limits the efficacy of chemotherapeutic treatment and compromises therapeutic outcomes in a substantial proportion of cases. Aberrant CpG island methylation-associated transcriptional silencing contributes to chemoresistance in pediatric solid tumors. Here, using whole-genome DNA methylation screening on 16 human primary OS specimens, we identify receptor interacting protein kinase-3 (RIPK3), a molecular regulator of the necroptosis programmed cell death pathway, as a gene target of aberrant CpG methylation and demonstrate its role in human OS chemoresistance. We validated these findings via enforced expression and DsiRNA silencing, and evaluated the role of RIPK3 in cisplatin chemosensitivity and necroptosis activation through MLKL phosphorylation. We found that CpG island methylation results in RIPK3 silencing in primary human OS samples and cell lines. Enforced RIPK3 expression significantly enhanced cisplatin cytotoxicity in OS cells and DsiRNA knockdown reversed the cisplatin-sensitive phenotype. In cells with enforced RIPK3 expression, cisplatin treatment significantly increased phosphorylation of both RIPK3 and its target, MLKL, indicative of induction of necroptosis. Here, we identify RIPK3 as an important mediator of chemoresistance in OS and a potential pharmacologic target to improve chemotherapy efficacy in drug-resistant tumors.
Collapse
Affiliation(s)
- Aditya Sharma
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Daniel Pettee
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
| | - Catherine Hord
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Maximilian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Samantha Hardy
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| | - Fayez F. Safadi
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
- University Hospitals Health Systems, 11100 Euclid Ave, Cleveland, OH 44106, USA
| | - Steven J. Kuerbitz
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA; (A.S.); (D.P.); (C.M.); (S.J.K.)
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA; (C.H.); (M.B.); (S.H.); (F.F.S.)
| |
Collapse
|
31
|
Tang J, Guo Y, Lu H, Fang Y, Chen W. Prognostic nomogram for overall survival in pediatric osteosarcoma with pulmonary metastases: a SEER database analysis. Front Pediatr 2025; 13:1574034. [PMID: 40313679 PMCID: PMC12043881 DOI: 10.3389/fped.2025.1574034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Background Pulmonary metastasis (PM) is the most common site of distant metastasis in osteosarcoma (OS), particularly in pediatric cases, which are associated with poor prognosis. However, limited research has focused on identifying prognostic factors (PFs) for pediatric osteosarcoma with pulmonary metastasis (POPM). This study aims to identify clinical features and PFs of POPM and develop a validated nomogram to predict overall survival in POPM patients. Methods A retrospective analysis was conducted using OS cases from the Surveillance, Epidemiology, and End Results (SEER) database (2010-2021). Clinical characteristics were compared between patients with and without PM. PFs were identified using Least Absolute Shrinkage and Selection Operator (LASSO) regression and evaluated through Kaplan-Meier analysis. Patients were divided into training (N = 148) and validation (N = 64) cohorts. Independent PFs were determined via Cox regression to construct a prognostic nomogram, which was assessed using the concordance index (C-index), the area under the receiver operating characteristic curve (AUC-ROC), and calibration plots. Decision curve analysis (DCA) was used to evaluate clinical applicability. Results LASSO regression identified key PFs: AJCC stage, T stage, median household income, systemic therapy, and time from diagnosis to treatment. Among these, all except T stage were validated as independent PFs via Cox regression. The nomogram demonstrated strong predictive accuracy with C-index values of 0.68 (training) and 0.71 (validation). AUC values for 1-, 3-, and 5-year survival were 0.786, 0.709, and 0.711 in the training cohort and 0.780, 0.760, and 0.776 in the validation cohort. Calibration plots showed excellent concordance between predicted and actual survival, and DCA confirmed the nomogram's clinical relevance. Conclusion AJCC stage, median household income, systemic therapy, and time from diagnosis to treatment are significant PFs for POPM survival. The validated nomogram provides a valuable tool for personalized prognostic assessment and treatment decision-making in clinical practice.
Collapse
Affiliation(s)
- Jiaxiang Tang
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yun Guo
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Hongting Lu
- Department of Pediatric Surgery, Women and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yifan Fang
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Weiming Chen
- Department of Pediatric Surgery, Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
32
|
Cao L, Jia K, Van Tine BA, Yu Y, Peng Y, Chen X, Pan Q, Yang W, Zhang Z, Shao Z, Wu W. KPNA2 promotes osteosarcoma progression by regulating the alternative splicing of DDX3X mediated by YBX1. Oncogene 2025:10.1038/s41388-025-03375-3. [PMID: 40216969 DOI: 10.1038/s41388-025-03375-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025]
Abstract
Osteosarcoma (OS) is a rapidly progressive primary malignant bone tumor that occurs in children and adolescents aged between 15 and 19 years and adults aged over 60 years. As alternative splicing (AS) changes caused by abnormal splicing factors contribute to tumor progression, gene expression and AS analyses were performed on 44 osteosarcoma patients to create a genome-wide co-expression network of RNA-binding proteins (RBPs), AS events, and AS genes. A gain- or loss-of-function osteosarcoma cell model was established, and an interactive network analysis and enrichment analysis were performed. Karyopherin Subunit Alpha 2 (KPNA2) negatively correlated with patient survival. KPNA2 transports splicing factor Y-box Binding Protein 1 (YBX1) into the nucleus and YBX1 accelerates the degradation of the ATP-dependent RNA helicase DDX3X (DDX3X) through the nonsense-mediated decay (NMD) pathway to promote intron retention of the DDX3X gene, thus reducing DDX3X protein levels. KPNA2/YBX1 axis regulates the stability of DDX3X mRNA and cell cycle progression. KPNA2/YBX1/DDX3X axis might be potential targets for inhibiting disease progression and improving OS patient survival. It integrates AS control of DDX3X into the progression of OS and represents a potential prognostic biomarker and therapeutic target for OS therapy.
Collapse
Affiliation(s)
- Li Cao
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ke Jia
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - B A Van Tine
- Division of Medical Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yihan Yu
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yizhong Peng
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xuanzuo Chen
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Qing Pan
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Wenbo Yang
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zhicai Zhang
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Zengwu Shao
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Wei Wu
- Department of Orthopaedic, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
33
|
Chu D, Huang R, Shi J, Xu R, Wei D. NETs-related genes predict prognosis and are correlated with the immune microenvironment in osteosarcoma. Front Oncol 2025; 15:1551074. [PMID: 40276062 PMCID: PMC12018237 DOI: 10.3389/fonc.2025.1551074] [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: 12/24/2024] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Background Osteosarcoma is the most common primary bone tumor. It has a high rate of early metastasis, and its treatment is one of the most challenging topics in the bone tumor field. Recent studies have shown that neutrophil extracellular traps play an important role in tumor metastasis and may provide new horizons for exploring metastasis in osteosarcoma. Methods OS data were downloaded from the TARGET database and Gene Expression Omnibus datasets. Univariate Cox regression was conducted to assess NETRGs. Patients were subsequently categorized into high- and low-risk groups on the basis of risk score values derived from multivariate Cox analysis, and prognostic models were established. The immune infiltration of relevant genes and drug sensitivity of key genes were also analyzed. Results A total of 15 NETs-related genes associated with osteosarcoma metastases were identified. Among them, a total of 4 genes were related to prognosis, namely, MAPK1, CFH, ATG7 and DDIT4, and a prognostic model based on these 4 genes was established. The prognosis was worse in the high-risk group, whose areas under the ROC curves (AUCs) were 0.857, 0.779, and 0.689 at 1, 3, and 5 years, respectively. The key genes were subsequently found to be associated with the infiltration of 20 types of immune cells. Finally, the small-molecule drug toxin c 10, an approximately 6700 mw protein, may target key genes. Finally, ATG7 was validated at the histological level by combining the results of the validation group dataset analysis. Conclusions A risk model based on 4 NETRDEGs is a reliable prognostic predictor for OS patients, and CFH and ATG7 may serve as a new diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Dawei Chu
- First Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Rui Huang
- First Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jiandang Shi
- Department of Orthopedic, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ruiqing Xu
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Daihao Wei
- First Clinical Medical College of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
34
|
Luo K, Tang H, Yan W, Li S, Luo X, Yang M, Li F, Liang J, Liao S, Liu Y, He J, Liu DH. Prognostic significance of T cells and NK cells in osteosarcoma: a dual-center retrospective study. World J Surg Oncol 2025; 23:130. [PMID: 40205405 PMCID: PMC11980234 DOI: 10.1186/s12957-025-03784-4] [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: 08/13/2024] [Accepted: 03/29/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND There is no study on the relationship between peripheral blood different lymphocyte subtypes and the prognosis of osteosarcoma (OS). Therefore, this study aims to investigate the predictive value of T cells and natural killer (NK) cells for the prognosis of OS patients. METHODS This study retrospectively analyzed the clinical data and preliminary laboratory indicators of patients with OS admitted from dual-center between January 2014 and January 2021. The receiver operating characteristic (ROC) curve was employed to determine optimal cutoff values for different lymphocyte subtypes, with T cells, NK cells, and B lymphocytes subsequently stratified into high- and low-proportion groups based on their respective optimal cutoff values. Kaplan-Meier curve was employed to analyze the impact of different lymphocyte on survival time and status. Univariate and multivariate Cox analyses were performed on clinical and laboratory indicators to identify independent prognostic factors influencing the prognosis of OS patients. RESULTS After screening 277 patients with OS, a total of 106 patients were eligible for this study. The median follow-up time was 36.00 months. At the last follow-up, patients were categorized as having a good prognosis if they survived or a poor prognosis if they died: good prognosis (n = 48) and poor prognosis (n = 58). Kaplan-Meier curve revealed that patients with a high proportion of T (Median overall survival: 41 months vs. 32 months, P = 0.007) and NK (Median overall survival: 44 months vs. 32 months, P = 0.004) cells had a better prognosis compared to those with a low proportion. Univariate analysis indicated that age, body mass index (BMI), C-reactive protein (CRP), tumor size, Enneking stage, surgical method, and the proportions of T, NK, and B cells were associated with the prognosis of OS patients (P < 0.05). Multivariate analysis indicated that Enneking stage (II vs. I, HR = 12.543, P = 0.015; III vs. I, HR = 29.078, P = 0.001), and the proportions of T and NK cells (HR = 0.466, P = 0.048; HR = 0.497, P = 0.029) were independent factors influencing the prognosis of OS patients (P < 0.05). CONCLUSION The proportions of T and NK cells may serve as efficient and practical prognostic indicators for OS patients, with higher proportions often associated with a better prognosis.
Collapse
Affiliation(s)
- Kai Luo
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Haijun Tang
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Weijie Yan
- Department of Bone and soft Tissue Oncology, Affiliated Cancer Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Shanhang Li
- Department of Bone and soft Tissue Oncology, Affiliated Cancer Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Xiaoting Luo
- Department of Pharmacy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Mingxiu Yang
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Feicui Li
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Jiming Liang
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Shijie Liao
- Department of Traumatic orthopedics Hand Surgery, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Yun Liu
- Department of Spine and Osteopathy, the First Affiliated Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China.
| | - Juliang He
- Department of Bone and soft Tissue Oncology, Affiliated Cancer Hospital of Guangxi Medical University, 530021, Nanning, Guangxi, China.
| | - Dehuai H Liu
- Department of Orthopaedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, 530001, Nanning, Guangxi, China.
| |
Collapse
|
35
|
Li D, Li Y, Cang J, Yan X, Wu F, Sun X, Zhang W. Synergistic chemo-immunotherapy for osteosarcoma via a pH-responsive multi-component nanoparticle system. Front Pharmacol 2025; 16:1584245. [PMID: 40264674 PMCID: PMC12011790 DOI: 10.3389/fphar.2025.1584245] [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: 02/27/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Osteosarcoma (OS) is the most common primary malignant bone tumor in pediatric populations. Its treatment is complicated by chemotherapy-induced toxicity and limited induction of immunogenic cell death (ICD). Methods To address these challenges, we developed a pH-responsive, multi-component nanoparticle system designed to co-deliver doxorubicin (DOX), monophosphoryl lipid A (MPLA), and a PD-1/PD-L1-targeting peptide, integrated with the immune-modulating polymer PEG-PC7A. The system was optimized using both one-factor-at-a-time (OFAT) and Box-Behnken design (BBD). Results The optimized nanoparticles had a hydrodynamic size of 110 nm, high encapsulation efficiency (97.15%), and pH-sensitive drug release (91% at pH 6.5). In vitro studies showed enhanced ICD markers, including calreticulin exposure and ATP/HMGB1 release, aswell as synergistic dendritic cell maturation via dual STING/TLR4 pathway activation. In an orthotopic LM8 osteosarcoma model, the nanoparticles significantly suppressed tumor growth, promoted cytotoxic T lymphocyte infiltration, reduced regulatory T cells, and established long-term immune memory. Discussion The combination of ICD induction, innate immune activation, and checkpoint blockade reprogrammed the tumor microenvironment, amplifying anti-tumor immune responses. These results demonstrate the potential of this multifunctional nanoparticle platform as an effective immunochemotherapeutic strategy for osteosarcoma, offering enhanced therapeutic efficacy and reduced systemic toxicity.
Collapse
Affiliation(s)
- Dapeng Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuanfan Li
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jie Cang
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xianwen Yan
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feipeng Wu
- Department of Spine Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenchao Zhang
- Department of Orthopedics, Affiliated Jintan Hospital of Jiangsu University, Jintan, Jiangsu, China
| |
Collapse
|
36
|
Zhang Y, Liu W, Liu D, Li X, Zhuang Q, Sun Q, Wu X, Li F. Multi-omics analysis of copper metabolism-related molecular subtypes and risk stratification for osteosarcoma. Discov Oncol 2025; 16:480. [PMID: 40192894 PMCID: PMC11977037 DOI: 10.1007/s12672-025-02273-0] [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: 11/06/2024] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND As the most common primary malignant bone tumor, further investigation into risk stratification for osteosarcoma (OS) prognosis is of significant clinical importance. Copper is essential for bone metabolism; however, its specific role in OS remains unclear. METHODS The expression characteristics of copper metabolism related genes (CORGs) in OS were revealed by single cell sequencing. Prognosis-associated CORGs were identified, and a CORG-related scoring system and risk model were established using bioinformatics approaches, including univariate and multivariate Cox regression analyses and LASSO analysis. We further analyzed immune microenvironment infiltration, molecular subtypes and clinicopathological characteristics. The impact of selected CORG with high-risk coefficient on OS cells was tested by qRT-PCR, western blot, siRNA, colony formation analysis and Transwell in vitro. RESULTS We successfully developed an OS scoring system related to copper metabolism and validated its independent prognostic value in patients with OS. The potential clinical value of CORG scoring system was analyzed. APOA4 was selected for in vitro experiments and its effect on the proliferation and invasion ability of OS cells was verified. CONCLUSION We established a copper metabolism-related scoring system to effectively stratify the risk of OS patients. Our results provide a new basis for the role of copper metabolism in OS and provide new potential targets for the treatment of OS.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Wen Liu
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Dayong Liu
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Xiaopeng Li
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Qingshan Zhuang
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Quan Sun
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China
| | - Xiaolin Wu
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
- Cancer Institute, the Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, China.
| | - Feng Li
- Department of Minimally Invasive Spine Surgery, Weifang People's Hospital, Weifang, China.
| |
Collapse
|
37
|
Zhang W, Li L, Wang Z, Nie Y, Yang Y, Li C, Zhang Y, Jiang Y, Kou Y, Zhang W, Lai Y. Injectable and adhesive MgO 2-potentiated hydrogel with sequential tumor synergistic therapy and osteogenesis for challenging postsurgical osteosarcoma treatment. Biomaterials 2025; 315:122959. [PMID: 39612764 DOI: 10.1016/j.biomaterials.2024.122959] [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: 07/30/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024]
Abstract
The clinical treatment of osteosarcoma faces great challenges of residual tumor cells leading to tumor recurrence and irregular bone defects difficult to repair after surgery removal of the primary tumor tissue. We developed an injectable and in-situ cross-linkable hydrogel (named MOG hydrogel) using MgO2 nanoparticles and dopamine-conjugated gelatin as main components. MgO2 was rationally designed as a multifunctional active ingredient to mediate in situ gelation, tumor therapy, and bone repair sequentially. The 10MOG (with 10 mg/mL MgO2 content) showed excellent gel stability, injectability, shape adaptability, tissue adhesion, and rapid hemostatic ability. Importantly, 10MOG exhibited ideal sequential H2O2 and Mg2+ release property. The released H2O2 synergized with photothermal therapy for enhanced tumor recurrence suppression, and the sustainable Mg2+ release efficiently promoted bone regeneration. The MOG hydrogel, possessing excellent on-demand antitumor and osteogenic capabilities in vitro and in vivo, exhibited tremendous potential in the clinical application for challenging postsurgical osteosarcoma treatment.
Collapse
Affiliation(s)
- Wenjing Zhang
- Shenzhen Clinical Research Center for Trauma Treatment, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China; National Center for Trauma Medicine, Beijing, 100000, China
| | - Long Li
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Guangdong Engineering Laboratory of Biomaterials Additive Manufacturing, Shenzhen, 518055, China
| | - Zishuo Wang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yangyi Nie
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yipei Yang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Cairong Li
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuyang Zhang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuxi Jiang
- Shenzhen Clinical Research Center for Trauma Treatment, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, China
| | - Yuhui Kou
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, 100044, China; National Center for Trauma Medicine, Beijing, 100000, China.
| | - Wei Zhang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Guangdong Engineering Laboratory of Biomaterials Additive Manufacturing, Shenzhen, 518055, China.
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Guangdong Engineering Laboratory of Biomaterials Additive Manufacturing, Shenzhen, 518055, China.
| |
Collapse
|
38
|
Yin Z, Shen G, Fan M, Zheng P. Lipid metabolic reprogramming and associated ferroptosis in osteosarcoma: From molecular mechanisms to potential targets. J Bone Oncol 2025; 51:100660. [PMID: 39958756 PMCID: PMC11830322 DOI: 10.1016/j.jbo.2025.100660] [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: 12/01/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Osteosarcoma is a common bone tumor in adolescents, which is characterized by lipid metabolism disorders and plays a key role in tumorigenesis and disease progression. Ferroptosis is an iron-dependent form of programmed cell death associated with lipid peroxidation. This review provides an in-depth analysis of the complex relationship between lipid metabolic reprogramming and associated ferroptosis in OS from the perspective of metabolic enzymes and metabolites. We discussed the molecular basis of lipid uptake, synthesis, storage, lipolysis, and the tumor microenvironment, as well as their significance in OS development. Key enzymes such as adenosine triphosphate-citrate lyase (ACLY), acetyl-CoA synthetase 2 (ACSS2), fatty acid synthase (FASN) and stearoyl-CoA desaturase-1 (SCD1) are overexpressed in OS and associated with poor prognosis. Based on specific changes in metabolic processes, this review highlights potential therapeutic targets in the lipid metabolism and ferroptosis pathways, and in particular the HMG-CoA reductase inhibitor simvastatin has shown potential in inducing apoptosis and inhibiting OS metastasis. Targeting these pathways provides new strategies for the treatment of OS. However, challenges such as the complexity of drug development and metabolic interactions must be overcome. A comprehensive understanding of the interplay between dysregulation of lipid metabolism and ferroptosis is essential for the development of innovative and effective therapies for OS, with the ultimate goal of improving patient outcomes.
Collapse
Affiliation(s)
- Zhiyang Yin
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| | - Guanlu Shen
- School of Pharmacy, Jiangsu Ocean University, Lianyungang, Jiangsu, China
| | - Minjie Fan
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| | - Pengfei Zheng
- Department of Orthopaedics Surgery, Children’s Hospital of Nanjing Medical University, Nanjing 210000 Jiangsu Province, China
| |
Collapse
|
39
|
Wang L, Huang Y, Zhang X, Chen W, Dai Z. Exosomes derived from FN14-overexpressing BMSCs activate the NF-κB signaling pathway to induce PANoptosis in osteosarcoma. Apoptosis 2025; 30:880-893. [PMID: 39833632 PMCID: PMC11946957 DOI: 10.1007/s10495-024-02071-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] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Despite advances in treatment, the prognosis of osteosarcoma (OS) patients is unsatisfactory, and searching for possible targets is substantial. Fibroblast growth factor inducible type 14 (FN14), a plasma membrane protein, is involved in wound healing, angiogenesis, proliferation, apoptosis, and inflammation. However, its implication in OS development and progression has not been completely characterized. Herein, we explored the cell-to-cell communication of bone marrow mesenchymal stem cells (BMSCs) and OS cells mediated by FN14 in the tumor microenvironment of OS. To assess the interplay between FN14 expression levels and patient survival, FN14 expression was measured in both normal and OS tissues. The FN14 overexpressing BMSCs (OE) were constructed using lentivirus, and exosomes (EXO) were extracted. The uptake of FN14-containing EXO by OS cells was analyzed via flow cytometry and in vivo fluorescence imaging. In addition, high-throughput sequencing was performed to analyze the mechanisms by which EXO inhibits OS cell growth. Finally, the therapeutic effect of OE-EXO was evaluated in a mouse model of OS xenografts. The results showcased reduced FN14 expression in human and mouse OS tissues, suggesting its role may be involved in the malignant progression of OS. The FN14 expression was higher in BMSCs relative to OS cells, and FN14 was secreted and excreted by EXO. The OS cell progression was suppressed after the uptake of FN14-derived EXO from BMSCs. In addition, RNA sequencing revealed that FN14 in EXO activated NF-κB signaling, triggering PANoptosis in OS cells. In vivo, OE-EXO injection inhibited tumor growth in OS xenografts and significantly improved the long-term survival of mice. Our findings suggest that FN14 carried by EXO from BMSCs activates the NF-κB pathway to trigger PANoptosis in OS cells, providing a potential therapeutic strategy to inhibit OS progression.
Collapse
Affiliation(s)
- Liangming Wang
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yanbin Huang
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xiaolu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Wenkai Chen
- School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhangsheng Dai
- Department of Orthopedics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| |
Collapse
|
40
|
Manisundaram N, Ragan MV, Mayon L, Vaporciyan A, Austin MT. Surgical Margins and Oncologic Outcomes Following Wedge Resection of Pulmonary Metastases in Pediatric and Young Adult Patients with Sarcoma. Ann Surg Oncol 2025; 32:2883-2890. [PMID: 39789279 DOI: 10.1245/s10434-024-16800-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Children and young adults diagnosed with sarcoma often present with pulmonary metastases requiring wedge resection. It is important to balance the risk of pulmonary recurrence against the desire to limit resection of benign parenchyma. This study aims to determine the impact of resection margins on survival and recurrence among pediatric and young adult sarcoma patients. PATIENTS AND METHODS We conducted a retrospective cohort study of patients ages 25 years and younger with primary or recurrent osteogenic, Ewing's, or soft tissue sarcoma who underwent pulmonary metastasectomy (2006-2022). Margins were categorized as > 1 mm, ≥ 5 mm, or ≥ 10 mm length. Two-year overall survival (OS), disease-free survival (DFS), and regional disease-free survival, consisting of pulmonary recurrence following metastasectomy, were analyzed using the Kaplan-Meier method. Cox analysis utilized patient, tumor, and treatment factors to predict risk of death and/or recurrence. RESULTS In total, 122 patients were identified for analysis. The median number of wedge resections was 3.5, median nodule size was 12.5 mm, and median margin length was 3 mm. A 5-mm margin was associated with improvements in DFS and regional-DFS (10.6% vs. 29.7%, p = 0.01 and 10.7% versus 31.1%, p = 0.005, respectively). On Cox analysis, margin length was not associated with OS (p > 0.05); however, 5 mm (HR 0.46, p = 0.005) and 10-mm margins (HR 0.39, p = 0.04) were associated with improvements in regional DFS. Margin length was not associated with development of postoperative complications (p = 0.20). CONCLUSIONS Among pediatric and young adult sarcoma patients with pulmonary metastases, increased margin length was associated with decreasing risk of local recurrence but not the development of postoperative complications.
Collapse
Affiliation(s)
- Naveen Manisundaram
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Mecklin V Ragan
- Department of Surgery, Inova Fairfax, Falls Church, Falls Church, VA, USA
| | - Lauren Mayon
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara Vaporciyan
- Department of Cardiothoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mary T Austin
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
41
|
Griffin KH, Sagheb IS, Coonan TP, Fierro FA, Randall RL, Leach JK. Macrophage and osteosarcoma cell crosstalk is dependent on oxygen tension and 3D culture. BIOMATERIALS ADVANCES 2025; 169:214154. [PMID: 39708660 DOI: 10.1016/j.bioadv.2024.214154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/29/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Osteosarcoma (OS), the most common form of primary bone cancer in young adults, has had no improvements in clinical outcomes in 50 years. This highlights a critical need to advance mechanistic understanding of OS to further therapeutic discovery, which will only be possible with accurate models of the disease. Compared to traditional monolayer studies and preclinical models, in vitro models that better replicate the three-dimensional (3D) bone marrow microenvironment will facilitate methodical investigations of the events and factors that drive OS progression. Herein, we use fibrin-alginate interpenetrating network (FA IPN) hydrogels to model the hematological bone marrow environment. We interrogated the effects of oxygen tension, 3D culture, and macrophage phenotype on OS behavior and specifically examine the immunomodulatory crosstalk between OS and macrophages. We observe that OS is more sensitive to oxygen tension when cultured in 3D. Specifically, both highly and less metastatic OS exhibit decreased changes in DNA content over time in 3D, but then demonstrate diverging behaviors in heterotypic culture with macrophages. OS response to macrophages differs as a function of metastatic potential, where highly metastatic OS shows increased immunosuppression that varies with oxygen tension but relies on direct coculture conditions. To our knowledge, this is among the first work to report the effects of 3D culture on the interplay between OS and macrophages in a coculture microenvironment. Together, these data introduce FA IPNs as a promising platform for cancer research and emphasize the importance of novel models for the mechanistic study of OS.
Collapse
Affiliation(s)
- Katherine H Griffin
- School of Veterinary Medicine, University of California, Davis, CA, USA; Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - Isabel S Sagheb
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Thomas P Coonan
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Fernando A Fierro
- Department of Cell Biology and Human Anatomy, UC Davis Health, Sacramento, CA, USA
| | - R Lor Randall
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA, USA; Department of Biomedical Engineering, University of California, Davis, CA, USA.
| |
Collapse
|
42
|
Lai G, Zhao X, Chen Y, Xie T, Su Z, Lin J, Chen Y, Chen K. The origin and polarization of Macrophages and their role in the formation of the Pre-Metastatic niche in osteosarcoma. Int Immunopharmacol 2025; 150:114260. [PMID: 39938167 DOI: 10.1016/j.intimp.2025.114260] [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: 10/26/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Osteosarcoma, a primary malignant bone tumor commonly found in adolescents, is highly aggressive, with a high rate of disability and mortality. It has a profound negative impact on both the physical and psychological well-being of patients. The standard treatment approach, comprising surgery and chemotherapy, has seen little improvement in patient outcomes over the past several decades. Once relapse or metastasis occurs, prognosis worsens significantly. Therefore, there is an urgent need to explore new therapeutic approaches. In recent years, the successful application of immunotherapy in certain cancers has demonstrated its potential in the field of cancer treatment. Macrophages are the predominant components of the immune microenvironment in osteosarcoma and represent critical targets for immunotherapy. Macrophages exhibit dual characteristics; while they play a key role in maintaining tumor-promoting properties within the microenvironment, such as inflammation, angiogenesis, and immune suppression, they also possess antitumor potential as part of the innate immune system. A deeper understanding of macrophages and their relationship with osteosarcoma is essential for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Guisen Lai
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Xinyi Zhao
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanquan Chen
- Department of Orthopaedic Sun Yat-sen Memorial Hospital Sun Yat-sen University PR China
| | - Tianwei Xie
- The People's Hospital of Hezhou, No.150 Xiyue Street, Hezhou 542800 PR China
| | - Zepeng Su
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Jiajie Lin
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Yuanhai Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China
| | - Keng Chen
- Department of Orthopaedic The Eighth Affiliated Hospital Sun Yat-sen University PR China.
| |
Collapse
|
43
|
Ghufran SM, Brown ML, Beierle EA. Role of exosomes in diagnosis, prognostication, and treatment of pediatric solid tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200930. [PMID: 39895692 PMCID: PMC11783428 DOI: 10.1016/j.omton.2024.200930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cancer is the second leading cause of death in children, and solid tumors make up 30% of childhood cancers. Molecular profiling of pediatric solid tumors allows a personalized approach to therapy, but this approach mostly relies on surgical biopsy, which is invasive and carries the risk of complications. Liquid biopsy serves as a reliable alternative and a minimally invasive tool for diagnosing, prognosticating, and residual disease monitoring in childhood cancers. This review outlines the potential of exosomes as informative liquid biopsies in pediatric solid tumors. Studies highlighting the potential applications and clinical utility of exosomes and their molecular constituents as prognosticators and therapies in common childhood solid tumors, including neuroblastoma, medulloblastoma, sarcoma, and hepatoblastoma, have been overviewed. We also discuss the limitations and technical challenges of utilizing exosomes for pediatric solid tumors.
Collapse
Affiliation(s)
- Shaikh M. Ghufran
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Morgan L. Brown
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| | - Elizabeth A. Beierle
- University of Alabama at Birmingham, Department of Surgery, Division of Pediatric Surgery, Birmingham, AL 35233, USA
| |
Collapse
|
44
|
Zhou H, Shu R, Wu J, Zhou J, Yu Z, Cheng Q, Peng Z, Zhao M. Review of the role and potential clinical value of m6A methylation modifications in the biological process of osteosarcoma. Front Genet 2025; 16:1522622. [PMID: 40176793 PMCID: PMC11961878 DOI: 10.3389/fgene.2025.1522622] [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: 11/04/2024] [Accepted: 02/17/2025] [Indexed: 04/04/2025] Open
Abstract
Osteosarcoma (OS), an aggressive bone tumor, is a substantial threat to the quality of life and survival of affected individuals. Despite recent improvements in OS therapies, the considerable variability and chemotherapy resistance of this cancer necessitate continuous research to discover new treatment targets and biomarkers. Recent epigenetic advances highlight the crucial role of N6-methyladenosine (m6A) methylation in cancer. In OS, m6A methylation has been demonstrated to be a pivotal component in the pathogenesis. This review introduces new findings regarding the association between m6A methylation regulators and OS, and summarizes the potential clinical applications of OS and m6A methylation regulators, including the role of m6A methylation in OS proliferation, growth, apoptosis, and cell migration, invasion, and metastasis; relationship between m6A methylation and OS chemotherapy resistance; and relationship between m6A methylation and OS prognosis. Our review had certain limitations. The interaction between m6A methylation regulators and other oncogenic factors, such as lncRNAs and ncRNAs, is not fully understood. We hope that these potential methods will be translated into clinical applications and effective treatment.
Collapse
Affiliation(s)
- Huaqiang Zhou
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Rongbing Shu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Jianming Wu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Jiangjun Zhou
- Department of Orthopedic, The 908Th Hospital of Joint Logistic Support Force of PLA, Nanchang, China
| | - Zhuanyi Yu
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Qiuxin Cheng
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Zhihao Peng
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| | - Min Zhao
- Department of orthopaedic surgery, Yingtan People’s Hospital, YingTan, China
| |
Collapse
|
45
|
Zhang S, Chen X, Li J, Xu A, Bode AM, Luo X. The role of cryptochrome (CRY) in cancer: molecular mechanisms and Clock-based therapeutic strategies. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40109093 DOI: 10.3724/abbs.2025025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
The circadian rhythm is a phenomenon in which physiological, behavioral, and biochemical processes within an organism naturally fluctuate over a period of approximately 24 hours. This phenomenon is ubiquitous in living organisms. Disruption of circadian rhythms in mammals leads to different diseases, such as cancer, and neurodegenerative and metabolic disorders. In specific tissues, numerous genes have been found to have circadian oscillations, suggesting a broad role for rhythm genes in the regulation of gene expression. This review systematically summarizes the role of cryptochromes (CRYs) in the initiation and progression of different types of cancer and discusses the relationships between Clock genes and the tumor microenvironment (TME), as well as clock-based therapeutic strategies.
Collapse
Affiliation(s)
- Shuzhao Zhang
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Xue Chen
- Early Clinical Trial Center, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Jiayi Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Anan Xu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha 410078, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha 410078, China
| |
Collapse
|
46
|
Wang K, Shi C, Liu L, Yan H, Wang D, Ding M, Tong J, He Y, Hu Y, Chen C, Cao D, Zhang F, Zheng X, Liu Z. Design, synthesis, and biological evaluation of Flavokavain B derivatives as potent TRF2 inhibitors for the treatment of Osteosarcoma. Eur J Med Chem 2025; 286:117279. [PMID: 39874631 DOI: 10.1016/j.ejmech.2025.117279] [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: 09/16/2024] [Revised: 12/29/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025]
Abstract
Telomere repeat-binding factor 2 (TRF2) is a crucial component of the shelterin complex, commonly overexpressed in osteosarcoma (OS) and positively correlated with its progression. To date, effective TRF2 inhibitors for in vivo applications remain limited. In this study, a series of Flavokavain B derivatives were designed and synthesized, and their TRF2 inhibition and antitumor activity were evaluated. Among the tested compounds, the active compound F2 showed remarkable inhibition of TRF2 expression, along with potent antiproliferative activity in U2OS and MG63 cells, with IC50 values of 5.28 μM and 1.52 μM, respectively. Moreover, F2 significantly suppressed OS cell proliferation and induced apoptosis by accelerating telomere shortening and loss due to TRF2 inhibition. Mechanically, F2 selectively inhibited TRF2 protein expression and telomeric localization by directly binding to the TRF2TRFH domain. Furthermore, F2 demonstrated strong antitumor efficacy with minimal toxicity in an MG63-derived xenograft mouse model. These findings demonstrate that F2 is a promising drug candidate for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Kun Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Changgui Shi
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Lu Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Hao Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Dalong Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Meiqing Ding
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Jiaying Tong
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Yeying He
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Yina Hu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Chaoyue Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Di Cao
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China
| | - Fangjun Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, People's Republic of China.
| |
Collapse
|
47
|
Li J, Tang X, Wang L, Liu T. Vascularized fibular epiphyseal transfer for biological reconstruction of bone defects following resection in children with proximal humeral sarcoma. BMC Surg 2025; 25:95. [PMID: 40069703 PMCID: PMC11895305 DOI: 10.1186/s12893-025-02828-6] [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] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The functional reconstruction of bone defects following resection of proximal humerus tumors in children poses a significant challenge. This study utilized vascularized fibular epiphyseal transfer for proximal humerus reconstruction to evaluate the outcome, complications, and survival rates. METHODS In this study, we conducted a retrospective analysis of 13 pediatric patients who underwent vascularized fibular epiphyseal transfer for biological reconstruction following oncologic resection of the proximal humerus between 2019 and 2021. All patients received adequate preoperative preparation and evaluation, and complications were meticulously recorded. Regular functional follow-ups and imaging evaluations were performed. RESULTS A total of 13 patients with an average age of 9.8 years were included in this study. The average length of the humerus defect after surgical resection was 13.7 cm (9.4-17.8 cm). Delayed wound healing was observed in 2 patients, and one patient experienced brief common peroneal nerve palsy. There were 3 cases of graft fracture, all of which occurred within 1 year after operation. These cases were successfully managed through the application of draping plaster or brace fixation. The mean follow-up period was 39.8 months (ranging from 19 to 57 months). The mean Musculoskeletal Tumor Society (MSTS) score was 21.5 (18-24). All patients reported no persistent pain. CONCLUSION In conclusion, we assert that vascularized fibular epiphyseal transfer provides a reliable and promising option for reconstruction in pediatric patients undergoing proximal humerus tumor resection surgery. Graft fractures were the most prevalent complication, emphasizing the importance of cautionary measures to prevent falls or trauma. However, further validation through increased case numbers and extended follow-up periods is necessary.
Collapse
Affiliation(s)
- Jun Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, P. R. China
| | - Xianzhe Tang
- Department of Orthopedics, Chenzhou First People's Hospital, Chenzhou, Hunan, China
| | - Lu Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, P. R. China.
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, P. R. China.
| |
Collapse
|
48
|
Li B, Xu D, Lin H, Wu R, Wu S, Shao J, Zhang J, Dai H, Wei D, Huang B, Gao Z, Diao X. Domain adaptive detection framework for multi-center bone tumor detection on radiographs. Comput Med Imaging Graph 2025; 123:102522. [PMID: 40154010 DOI: 10.1016/j.compmedimag.2025.102522] [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: 12/07/2023] [Revised: 01/14/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
Automatic bone tumor detection on radiographs is crucial for reducing mortality from bone cancer. However, the performance of the detection methods may be considerably affected when deployed to bone tumor data in a distinct domain, which could be attributed to the differences in the imaging process and can be solved by training with a large amount of annotated data. However, these data are difficult to obtain in clinical practice. To address this challenge, we propose a domain-adaptive (DA) detection framework to effectively bridge the domain gap of bone tumor radiographs across centers, consisting of four parts: a multilevel feature alignment module (MFAM) for image-level alignment, Wasserstein distance critic (WDC) for quantization of feature distance, instance feature alignment module (IFAM) for instance-level alignment, and consistency regularization module (CRM), which maintains the consistency between the domain predictions of MFAM and IFAM. The experimental results indicated that our framework can improve average precision (AP) with an intersection over union threshold of 0.2 (AP@20) on the source and target domain test sets by 1 % and 8.9 %, respectively. Moreover, we designed a domain discriminator with an attention mechanism to improve the efficiency and performance of the domain-adaptative bone tumor detection model, which further improved the AP@20 on the source and target domain test sets by 2 % and 10.7 %, respectively. The proposed DA model is expected to bridge the domain gap and address the generalization problem across multiple centers.
Collapse
Affiliation(s)
- Bing Li
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China; Medical Imaging Department, The First Affiliated Hospital of Guangdong Pharmaceutical University, China
| | - Danyang Xu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hongxin Lin
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China
| | - Ruodai Wu
- Radiology Department, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Songxiong Wu
- Radiology Department, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, China
| | - Jingjing Shao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jinxiang Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haiyang Dai
- Department of Radiology, People's Hospital of Huizhou City Center, Huizhou, Guangdong, China
| | - Dan Wei
- Department of Radiology, Huiya Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Huizhou, Guangdong, China
| | - Bingsheng Huang
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China.
| | - Zhenhua Gao
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Radiology, Huiya Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Huizhou, Guangdong, China.
| | - Xianfen Diao
- Medical AI Lab, School of Biomedical Engineering, Medical School, Shenzhen University, Shenzhen, China; National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Medical School, Shenzhen University, Shenzhen, China.
| |
Collapse
|
49
|
Wei G, Jia H, Zhang Z, Qin J, Ao J, Qian H. O-GlcNAcylation: Sagacious Orchestrator of Bone-, Joint-, and Spine-Related Diseases. J Proteome Res 2025; 24:981-994. [PMID: 39921656 PMCID: PMC11894655 DOI: 10.1021/acs.jproteome.4c00859] [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: 10/18/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025]
Abstract
O-linked beta-N-acetylglucosamine glycosylation (O-GlcNAcylation), a post-translational modification of proteins, occurs in multiple physiological and pathological processes. Despite comprehensive study of protein modifications, such as phosphorylation, acetylation, and ubiquitination in musculoskeletal diseases, the role of O-GlcNAcylation in this field has been largely overlooked. However, in recent years, several studies have initially elucidated the biological mechanisms through which O-GlcNAcylation regulates the development and progress of musculoskeletal diseases, including osteoarthritis, osteoporosis, osteosarcoma, and intervertebral disc degeneration. This review aims to systematically and comprehensively summarize the existing evidence, sketching the contours of the underlying mechanisms and related signaling pathways, discussing the limitations and controversies, and providing guidance for future studies on the role of O-GlcNAcylation modifications in musculoskeletal diseases.
Collapse
Affiliation(s)
- Guihuo Wei
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Hao Jia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Zhuo Zhang
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Jianpu Qin
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Jun Ao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Hu Qian
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
50
|
Du H, Wu D, Zhang T, Zhong Y, Wu K, Guo X, Sheng L, Huang N, Gao C, Sun R. Ziyuglycoside II suppressed the progression of osteosarcoma by coordinating estrogen-related receptor gamma and p53 signaling pathway. Chin J Nat Med 2025; 23:354-367. [PMID: 40122665 DOI: 10.1016/s1875-5364(25)60847-8] [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: 12/26/2023] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 03/25/2025]
Abstract
Osteosarcoma (OS) is the most prevalent primary malignant bone tumor affecting children and adolescents. Despite ongoing research efforts, the 5-year survival rate has remained stagnant for many years, highlighting the critical need for novel drug development to enhance current treatment protocols. Ziyuglycoside II (ZYG II), a triterpenoid saponin extracted from S. officinalis, has recently demonstrated antitumor properties. This study evaluates the antitumor effect of ZYG II on osteosarcoma and elucidates its mechanism of action through the co-regulation of p53 and estrogen-related receptor gamma (ESRRG), which inhibits disease progression. The research employs in vitro experiments using multiple established osteosarcoma cell lines, as well as in vivo studies utilizing a nude mouse model of orthotopic xenograft osteosarcoma. Additionally, ESRRG shRNA was used to construct stable ESRRG-reducing OS cell lines to investigate the molecular mechanism by which ZYG II exerts its anti-osteosarcoma effects through the co-regulation of ESRRG and p53. Results indicate that ZYG II administration led to decreased OS cell viability and reduced tumor volumes. Furthermore, cell cycles were arrested at the G0/G1 phase, while the proportion of apoptotic cells increased. Expression of p53, ESRRG, p21, Bax, Cleaved Caspase-9, and Cleaved Caspase-3 proteins increased, while expression of CDK4, Cyclin D1, and Bcl-2 proteins decreased. Multiple ZYG II and ESRRG docking patterns were simulated through molecular docking. Comparing the pharmacodynamic response of ZYG II to OS cell lines with reduced ESRRG and normal expression demonstrated that ZYG II inhibits osteosarcoma progression, induces cell cycle arrest, and promotes cell apoptosis through the coordination of p53 and ESRRG. In conclusion, ZYG II inhibits osteosarcoma progression, leads to cell cycle arrest, and promotes cell apoptosis through synergistic regulation of p53 and ESRRG.
Collapse
Affiliation(s)
- Hang Du
- The Second Hospital of Shandong University, Jinan 250033, China
| | - Dongjin Wu
- The Second Hospital of Shandong University, Jinan 250033, China
| | - Tianyu Zhang
- The Second Hospital of Shandong University, Jinan 250033, China; Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese medicine Jinan 250355, China
| | - Ying Zhong
- The Second Hospital of Shandong University, Jinan 250033, China
| | - Kaiyi Wu
- The Second Hospital of Shandong University, Jinan 250033, China; School of Pharmacy, Tianjin University of Traditional Chinese medicine, Tianjin 301617, China
| | - Xin Guo
- The Second Hospital of Shandong University, Jinan 250033, China; School of Pharmacy, Tianjin University of Traditional Chinese medicine, Tianjin 301617, China
| | - Lisong Sheng
- Advanced Medical Research Institute, Shandong University, Jinan 250012, China
| | - Nana Huang
- The Second Hospital of Shandong University, Jinan 250033, China; Academy of Traditional Chinese Medicine, Shandong University of Traditional Chinese medicine Jinan 250355, China
| | - Chunzheng Gao
- The Second Hospital of Shandong University, Jinan 250033, China.
| | - Rong Sun
- The Second Hospital of Shandong University, Jinan 250033, China; Advanced Medical Research Institute, Shandong University, Jinan 250012, China.
| |
Collapse
|