1
|
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
|
2
|
Hu Q, Ren A, Zhang X, Tang Z, Wang R, Wang DY, Huang T, Liu J, Ming J. Manganese-Loaded pH-Responsive DNA Hydrogels Enable Tg-Guided Thyroid Tumor Targeted Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13403-13414. [PMID: 39996966 DOI: 10.1021/acsami.4c19676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The diagnosis of metastatic and recurrent occult thyroid cancer presents a significant challenge. This study introduces a DNA-Mn hydrogel (M-TDH) that specifically targets thyroglobulin (Tg). This nanogel is loaded with paramagnetic Mn2+ for facilitating magnetic resonance (MR) imaging. As a cofactor of DNA polymerase, Mn2+ promotes the extension of long-strand DNA and forms Mn2PPi nuclei with PPi4- in the system. The synthesis of M-TDH is achieved through Mn2PPi nucleation and growth with long-strand DNA acting as the structural framework. The X-scaffold functions as a junction point, thereby enhancing structural stability. The Tg aptamer sequence is incorporated into M-TDH, ensuring specific targeting of thyroid cancer cells. Furthermore, M-TDH demonstrates an extended residence time at the thyroid tumor site, thus increasing the duration of enhanced MR imaging. Overall, this study introduces an aptamer-based, thyroid tumor-targeted DNA nanogel for MR imaging diagnostic applications, with the potential to advance a multifunctional magnetic nanosystem toward clinical application.
Collapse
Affiliation(s)
- Qingyi Hu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Anwen Ren
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Ximeng Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Zimei Tang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Rong Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Dong-Yuan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Jie Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| |
Collapse
|
3
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
4
|
Topçu BT, Bozdağ Pehlivan S, Akdağ Y, Mut M, Öner L. Antibody Conjugated Nano-Enabled Drug Delivery Systems Against Brain Tumors. J Pharm Sci 2024; 113:1455-1469. [PMID: 38555997 DOI: 10.1016/j.xphs.2024.03.017] [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: 01/18/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
The use of antibody-conjugated nanoparticles for brain tumor treatment has gained significant attention in recent years. Nanoparticles functionalized with anti-transferrin receptor antibodies have shown promising results in facilitating nanoparticle uptake by endothelial cells of brain capillaries and post-capillary venules. This approach offers a potential alternative to the direct conjugation of biologics to antibodies. Furthermore, studies have demonstrated the potential of antibody-conjugated nanoparticles in targeting brain tumors, as evidenced by the specific binding of these nanoparticles to brain cancer cells. Additionally, the development of targeted nanoparticles designed to transcytoses the blood-brain barrier (BBB) to deliver small molecule drugs and therapeutic antibodies to brain metastases holds promise for brain tumor treatment. While the use of nanoparticles as a delivery method for brain cancer treatment has faced challenges, including the successful delivery of nanoparticles to malignant brain tumors due to the presence of the BBB and infiltrating cancer cells in the normal brain, recent advancements in nanoparticle-mediated drug delivery systems have shown potential for enhancing the efficacy of brain cancer therapy. Moreover, the development of brain-penetrating nanoparticles capable of distributing over clinically relevant volumes when administered via convection-enhanced delivery presents a promising strategy for improving drug delivery to brain tumors. In conclusion, the use of antibody-conjugated nanoparticles for brain tumor treatment shows great promise in overcoming the challenges associated with drug delivery to the brain. By leveraging the specific targeting capabilities of these nanoparticles, researchers are making significant strides in developing effective and targeted therapies for brain tumors.
Collapse
Affiliation(s)
- Beril Taş Topçu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| | - Sibel Bozdağ Pehlivan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey.
| | - Yagmur Akdağ
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| | - Melike Mut
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Levent Öner
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University 06100, Ankara, Turkey
| |
Collapse
|