1
|
Khan M, Ullah R, Shah SM, Farooq U, Li J. Manganese-Based Nanotherapeutics for Targeted Treatment of Breast Cancer. ACS APPLIED BIO MATERIALS 2025; 8:3571-3600. [PMID: 40293195 DOI: 10.1021/acsabm.5c00040] [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: 04/30/2025]
Abstract
Breast cancer (BC) is one of the most common cancers among women and is associated with high mortality. Traditional modalities, including surgery, radiotherapy, and chemotherapy, have achieved certain advancements but continue to combat challenges including harm to healthy tissues, resistance to treatment, and adverse drug reactions. The rapid advancements in nanotechnology recently facilitated the exploration of innovative strategies for breast cancer therapy. Manganese-based nanotherapeutics have attracted great attention because of their unique characteristics such as tunable structures/morphologies, versatility, magnetic/optical properties, strong catalytic activities, excellent biodegradability, and biocompatibility. In this review, we highlighted different types of Mn-based nanotherapeutics to modulate TME, including metal-immunotherapy, alleviating tumor hypoxia, and increasing reactive oxygen species production, and we emphasized its role in magnetic resonance imaging (MRI)-guided therapy, photoacoustic imaging, and theranostic-based therapy along with a therapeutic carrier, all of which were discussed in the context of breast cancer. Hopefully, the present review will provide insights into the current landscape and future directions of multifunctional applications of Mn-based nanotherapeutics in the field of breast cancer treatment.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P.R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| | - Syed Mubassir Shah
- Department of Biotechnology, Abdul Wali Khan University, KPK, Mardan 23200, Pakistan
| | - Umar Farooq
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| | - Jun Li
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| |
Collapse
|
2
|
Wang Q, Wu F, Qiu X, Yu H, Lu M, Gu X, Liu J, Chen B, Zhang M, Du F. Zinc phthalocyanine and sulfasalazine coloaded nanoaggregates with Prussian blue functionalization for sensitizing tumor photothermal/photodynamic therapy. J Colloid Interface Sci 2025; 685:509-521. [PMID: 39953688 DOI: 10.1016/j.jcis.2025.01.111] [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: 10/29/2024] [Revised: 01/12/2025] [Accepted: 01/12/2025] [Indexed: 02/17/2025]
Abstract
Photothermal therapy/photodynamic therapy (PTT/PDT), as a noninvasive therapeutic modality, has been extensively applied in superficial tumor treatment. However, their curative effects were largely weakened due to hypoxia and an elevated glutathione (GSH) microenvironment. Herein, zinc phthalocyanine (ZnPc) and sulfasalazine (SAS) coloaded nanoaggregates (Z-S@B NAs) with Prussian blue (PB) functionalization (PB/Z-S@B NAs) were fabricated via self-assembly and using an in situ oxidative polymerization method for tumor PTT/PDT sensitization. The designed PB/Z-S@B NAs were capable of triggering local hyperthermia and generating substantial reactive oxygen species (ROS) under 660- and 808-nm laser irradiation. Notably, the PB/Z-S@B NAs exhibited favorable catalase-like (CAT-like) activity that decomposed hydrogen peroxide into oxygen, which further enhanced tumor cell sensitivity to PTT/PDT. Moreover, SAS from the PB/Z-S@B NAs remarkably decreased the antioxidant GSH level, resulting in a synergetic tumor-killing effect. Collectively, this study provides a versatile nanoplatform to overcome intrinsic antitumor effect and enhance tumor sensitivity to PTT/PDT.
Collapse
Affiliation(s)
- Qinxin Wang
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China; Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Feifei Wu
- Wuxi Traditional Chinese Medicine Hospital affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, PR China
| | - Xiaonan Qiu
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China
| | - Huijun Yu
- Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Mengke Lu
- Wuxi Traditional Chinese Medicine Hospital affiliated to Nanjing University of Chinese Medicine, Wuxi 214071, PR China
| | - Xuan Gu
- Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Baoding Chen
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China
| | - Miaomiao Zhang
- Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Fengyi Du
- Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China; Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
3
|
Li Z, Lu Y, Song J, Han P, Shi H, Yao X, Zhang X, Zhang G. An emodin-mediated multifunctional nanoplatform with augmented sonodynamic and immunoregulation for osteomyelitis therapy. J Colloid Interface Sci 2025; 684:122-137. [PMID: 39823728 DOI: 10.1016/j.jcis.2025.01.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 01/20/2025]
Abstract
Emodin (ED), as a traditional Chinese medicine, possesses a variety of biological activities and is also one of natural sonosensitizer. Whether emodin could react with titanium dioxide to enhance the sonodynamic activity for safely treating osteomyelitis remains to be explored. Hence, an ED-conjugated Mn-doped titanium dioxide (TOM) nanorod array is designed and prepared on titanium to eliminate bacterial infections under ultrasound (US) treatment. The doping of Mn and the formation of an inorganic-organic interfacial between ED and TOM cause an imbalance in the interfacial charge and reduce the band gap of TOM, thereby increasing the production of reactive oxygen species (ROS) under US. The generated ROS effectively destroys the bacterial cell membrane, accelerating Mn ions intake and thereby inducing ferroptosis-like death of MRSA. Moreover, the hybrid coating activates cAMP and cGMP-PKG pathways to modulate M1 polarization of macrophages to display potent phagocytosis to bacteria. After eliminating the bacterial infection, the coating also can facilitate macrophage M2 polarization to promote osteogenic differentiation. Overall, the Chinese medicine enhanced sonodynamic and immunotherapy have great promise in treating bacterial-infected osteomyelitis.
Collapse
Affiliation(s)
- Zehao Li
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Ying Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China
| | - Jianbo Song
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China
| | - Peide Han
- College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Huixian Shi
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China.
| | - Xiangyu Zhang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024 China; College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024 China.
| | - Guannan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032 China; Shanxi Provincial Key Laboratory for Translational Nuclear Medicine and Precision Protection, Taiyuan 030006 China.
| |
Collapse
|
4
|
Su W, Wang H, Pan J, Zhou Q. Advances in Sonodynamic Therapy: Focus on Ferroptosis. J Med Chem 2025; 68:5976-5992. [PMID: 40063557 DOI: 10.1021/acs.jmedchem.4c02603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Ferroptosis is a nonapoptotic form of cell death discovered in 2012. Noninvasive treatments regulating ferroptosis are important for a wide range of diseases. Among the noninvasive treatments, sonodynamic therapy (SDT) has become promising due to its strong tissue penetration and few side effects. In recent years, targeted drug delivery platforms constructed on the basis of SDT have provided an efficient delivery mode for the regulation of ferroptosis. Based on the latest research reports, this Perspective introduces the basic mechanism of SDT and the influencing factors of therapeutic effects, elucidates the significance of ferroptosis-targeted SDT, and summarizes the recent studies on ferroptosis-targeted SDT through different pathways. We also present innovative studies of composite ultrasound-responsive drug delivery platforms. Finally, a brief summary and outlook based on current ferroptosis-targeted SDT are presented.
Collapse
Affiliation(s)
- Wendi Su
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hao Wang
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Juhong Pan
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Zhou
- Echo Lab, Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
5
|
Xu W, Guan G, Yue R, Dong Z, Lei L, Kang H, Song G. Chemical Design of Magnetic Nanomaterials for Imaging and Ferroptosis-Based Cancer Therapy. Chem Rev 2025; 125:1897-1961. [PMID: 39951340 DOI: 10.1021/acs.chemrev.4c00546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Ferroptosis, an iron-dependent form of regulatory cell death, has garnered significant interest as a therapeutic target in cancer treatment due to its distinct characteristics, including lipid peroxide generation and redox imbalance. However, its clinical application in oncology is currently limited by issues such as suboptimal efficacy and potential off-target effects. The advent of nanotechnology has provided a new way for overcoming these challenges through the development of activatable magnetic nanoparticles (MNPs). These innovative MNPs are designed to improve the specificity and efficacy of ferroptosis induction. This Review delves into the chemical and biological principles guiding the design of MNPs for ferroptosis-based cancer therapies and imaging-guided therapies. It discusses the regulatory mechanisms and biological attributes of ferroptosis, the chemical composition of MNPs, their mechanism of action as ferroptosis inducers, and their integration with advanced imaging techniques for therapeutic monitoring. Additionally, we examine the convergence of ferroptosis with other therapeutic strategies, including chemodynamic therapy, photothermal therapy, photodynamic therapy, sonodynamic therapy, and immunotherapy, within the context of nanomedicine strategies utilizing MNPs. This Review highlights the potential of these multifunctional MNPs to surpass the limitations of conventional treatments, envisioning a future of drug-resistance-free, precision diagnostics and ferroptosis-based therapies for treating recalcitrant cancers.
Collapse
Affiliation(s)
- Wei Xu
- School of Life Science and Technology, Shandong Second Medical University, Weifang 261053, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Guoqiang Guan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Renye Yue
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei 230032, PR China
| | - Zhe Dong
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| | - Lingling Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China
| | - Heemin Kang
- Department of Materials Science and Engineering and College of Medicine, Korea University, 12 Seoul 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, PR China
| |
Collapse
|
6
|
Wang X, Gao M, Lu X, Lei Y, Sun J, Ren M, Xu T, Lin H. Resveratrol alleviates Mono-2-ethylhexyl phthalate-induced mitophagy, ferroptosis, and immunological dysfunction in grass carp hepatocytes by regulating the Nrf2 pathway. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 371:123235. [PMID: 39509968 DOI: 10.1016/j.jenvman.2024.123235] [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: 08/07/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Mono-2-ethylhexyl phthalate (MEHP) is the major biologically active metabolite of Di(2-ethylhexyl) phthalate (DEHP). This MEHP mono-ester metabolite can be transported through the bloodstream into tissues such as the liver, kidneys, fat, and testes and cause corresponding damage. Resveratrol (RSV) has anti-inflammatory, antioxidant, and detoxification characteristics. Our research examined whether RSV alleviates MEHP-induced grass carp hepatocyte (L8824 cell) injury and its relationship with the Nrf2 pathway, mitophagy, ferroptosis, and immune function. Therefore, we treated L8824 cells with 85 μM MEHP and/or 2 μM RSV. The findings indicated that exposing MEHP resulted in increased reactive oxygen species (ROS) content and decreased mitochondrial membrane potential in L8824 cells, which induced an up-regulation of the expression of mitophagy-related indicators (PINK1, Parkin, Beclin1, LC3B, and ATG5) and a down-regulation of P62. An up-regulation of the expression of the ferroptosis-related indicators TFR1 and COX-2, and GPX4 and FTH expression was down-regulated. In addition, there was a decrease in the expression of IL-2 and IFN-γ and an increase in the expression of inflammatory cytokines such as TNF-α, IL-1β, and IL-6 after exposure to MEHP. RSV activates the Nrf2 pathway and effectively alleviates MEHP-induced mitophagy, ferroptosis, and immunologic dysfunction of L8824 cells.
Collapse
Affiliation(s)
- Xiaodan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Meichen Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Xiunan Lu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yutian Lei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Jiatong Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Mengyao Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tong Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Laboratory of Embryo Biotechnology, College of Life Science, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
7
|
Wei W, Wang H, Ren C, Deng R, Qin Q, Ding L, Li P, Liu Y, Chang M, Chen Y, Zhou Y. Ultrasmall Enzyodynamic PANoptosis Nano-Inducers for Ultrasound-Amplified Hepatocellular Carcinoma Therapy and Lung Metastasis Inhibition. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409618. [PMID: 39225412 DOI: 10.1002/adma.202409618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Addressing the inefficiency of current therapeutic approaches for hepatocellular carcinoma is an urgent and pressing challenge. PANoptosis, a form of inflammatory programmed cell death, presents a dependable strategy for combating cancer by engaging multiple cell death pathways (apoptosis, pyroptosis, and necroptosis). In this study, an ultrasmall Bi2Sn2O7 nanozyme with ultrasound-magnified multienzyme-mimicking properties is designed and engineered as a PANoptosis inducer through destroying the mitochondrial function of tumor cells and enhancing the intracellular accumulation of toxic reactive oxygen species, finally triggering the activation of PANoptosis process. The role of PANoptosis inducer has been verified by the expression of related proteins, including cleaved Caspase 3, NLRP3, N-GSDMD, cleaved Caspase 1, p-MLKL, and RIPK3. The inclusion of external ultrasonic irradiation significantly augments the enzyodynamic therapeutic efficiency. In vitro and in vivo antineoplastic efficacy, along with inhibition of lung metastasis, validate the benefits of the Bi2Sn2O7-mediated PANoptosis pathway. This study not only elucidates the intricate mechanisms underlying Bi2Sn2O7 as a PANoptosis inducer, but also offers a novel perspective for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wuyang Wei
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Hai Wang
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Chunrong Ren
- Department of Gastroenterology, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Ruxi Deng
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Qiaoxi Qin
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Li Ding
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Pan Li
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ying Liu
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yang Zhou
- Department of Ultrasound, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, 610031, China
| |
Collapse
|
8
|
Chen H, Cui H, Liu W, Li BW, Tian Z, Zhao YY, Yu GT. Manganese drives ferroptosis of cancer cells via YAP/TAZ phase separation activated ACSL4 in OSCC. Oral Dis 2024; 30:4898-4908. [PMID: 38462885 DOI: 10.1111/odi.14925] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/02/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVE Ferroptosis has been defined as a novel form of regulated cell death characterized by iron-dependent lipid peroxidation. Manganese has been used to induce ferroptosis in cancer cells recently. This study aims to investigate whether manganese can induce ferroptosis in oral squamous cell carcinoma (OSCC) and the underlying biological mechanisms. MATERIALS AND METHODS Cancer cells with or without manganese treatment were analyzed by RNA-sequencing to identify ferroptosis-related genes. Next, the activation of YAP/TAZ/ACSL4-ferroptosis signaling pathway was detected. Bioinformatic analysis and immunofluorescence assay were used to explore the phase separation of YAP/TAZ. Finally, specimens of OSCC patients were applied to analyze the clinical significance of YAP/TAZ/ACSL4. RESULTS RNA-sequencing analysis showed the ferroptosis-related genes and YAP/TAZ were upregulated after manganese treatment. The results of immunofluorescence, ELISA, western blotting, etc. further confirmed that manganese-induced ferroptosis depends on YAP/TAZ/ACSL4 signaling pathway. Moreover, the activation of ACSL4 was achieved by YAP/TAZ phase separation. The survival analysis in OSCC specimen suggested that the higher level of YAP/TAZ-ACSL4 axis expression indicates longer survival. CONCLUSIONS Manganese induces YAP/TAZ phase separation and subsequent ACSL4 activation via YAP/TAZ nuclear translocation, which facilitates ferroptosis of OSCC. Then YAP/TAZ-ACSL4 axis can be used as a potential prognostic predictor of OSCC patients.
Collapse
Affiliation(s)
- Hao Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Wei Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo-Wen Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhen Tian
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yu-Yue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Guang-Tao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Dai X, Yang Y. Metal-organic frameworks: potential synergies with cold atmospheric plasmas for cancer control. J Mater Chem B 2024; 12:10770-10785. [PMID: 39350546 DOI: 10.1039/d4tb00968a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Metal-organic frameworks (MOFs) have attracted increasing attention for cancer treatment due to their unique characteristics such as crystallized porous structures, high surface area, and diverse and modifiable chemical properties. Despite the plethora of reports on MOF-based onco-therapeutic designs, these nanocomposites have rarely been launched for clinical use, given, at least, one unavoidable concern, i.e., biosafety. Among the diverse possibilities that MOFs can be engaged for cancer treatment, one unignorable opportunity is how MOFs can be combined with other emerging anti-cancer approaches as one treatment modality to resolve issues of either one for surpassed treatment efficacy. Taking cold atmospheric plasmas (CAPs) as an example, this review delineates the unique features of MOFs and discusses the possible synergies they can create with CAPs for mutual benefits. By providing one example on how MOFs can help overcome the issues of other pre-clinical cancer treatment regimens, this review identifies one research niche that may thrive the field of plasma medicine and revolutionize the schema of MOFs for biological applications.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China.
| | - Yixuan Yang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China.
| |
Collapse
|
10
|
He J, Zhang X, Xing H, Tan J, Zhang L, Xu Z, Kang Y, Xue P. Nanoparticle-mediated synergistic disruption of tumor innervation and redox homeostasis for potent antineoplastic therapy. J Control Release 2024; 376:457-469. [PMID: 39437966 DOI: 10.1016/j.jconrel.2024.10.039] [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: 06/28/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Innervation is closely linked to several biological processes that promote tumor growth, making it an increasingly promising therapeutic target. In this study, biomimetic hollow MnO2 nanocarriers camouflaged with tumor cell membranes (HMLC) are developed to encapsulate lidocaine, an innervation inhibitor, for effective antineoplastic therapy. This approach aims to suppress nerve fiber growth and induce intracellular redox imbalance. Benefiting from the tumor-homing effect, HMLC accumulates in cancerous tissue during circulation and is endocytosed by tumor cells through homologous membrane fusion. Once inside the cells, MnO2 can be degraded by the overproduced glutathione and H2O2, leading to the tumor-specific release of Mn2+ and lidocaine. The Mn2+-mediated Fenton-like reaction promotes the accumulation of reactive oxygen species, and the resulting oxidative stress, combined with glutathione depletion, exacerbates redox imbalance. Simultaneously, the released lidocaine downregulates nerve growth factor and neuronatin. The reduction in nerve growth factor significantly inhibits nerve fiber formation and infiltration in tumor tissue, while the decrease in neuronatin reduces intracellular Ca2+, which helps prevent metastasis. Overall, this strategy highlights the potential of nanoparticle-based tumor innervation disruptors in antineoplastic therapy.
Collapse
Affiliation(s)
- Jie He
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Xingli Zhang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Haiyan Xing
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Jiangwei Tan
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Lei Zhang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400715, China
| | - Zhigang Xu
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yuejun Kang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Peng Xue
- School of Materials and Energy, Southwest University, Chongqing 400715, China; Yibin Academy of Southwest University, Yibin 644005, China.
| |
Collapse
|
11
|
Chen Z, Zhang D, Huang H, Chen J, Li Z, Hu Y, Liu R. NIR Absorbing Organic Chromophores Combination with NSAIDs for Remodeling of the Inflammatory Microenvironment to Amplify Tumor Ferroptosis-Photothermal Synergistic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400361. [PMID: 38708879 DOI: 10.1002/smll.202400361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Indexed: 05/07/2024]
Abstract
Photothermal therapy has emerged as a promising approach for cancer treatment, which can cause ferroptosis to enhance immunotherapeutic efficacy. However, excessively generated immunogenicity will induce serious inflammatory response syndrome, resulting in a discounted therapeutic effect. Herein, a kind of NIR absorption small organic chromophore nanoparticles (TTHM NPs) with high photothermal conversion efficiency (68.33%) is developed, which can induce mitochondria dysfunction, generate mitochondrial superoxide, and following ferroptosis. TTHM NPs-based photothermal therapy is combined with Sulfasalazine (SUZ), a kind of nonsteroidal anti-inflammatory drugs, to weaken inflammation and promote ferroptosis through suppressing glutamate/cystine (Glu/Cys) antiporter system Xc- (xCT). Additionally, the combination of SUZ with PTT can induce immunogenic cell death (ICD), followed by promoting the maturation of DCs and the attraction of CD8+ T cell, which will secrete IFN-γ and trigger self-amplified ferroptosis via inhibiting xCT and simulating Acyl-CoA synthetase long-chain family member 4 (ACSL4). Moreover, the in vivo results demonstrate that this combination therapy can suppress the expression of inflammatory factors, enhance dendritic cell activation, facilitate T-cell infiltration, and realize effective thermal elimination of primary tumors and distant tumors. In general, this work provides an excellent example of combined medication and stimulates new thinking about onco-therapy and inflammatory response.
Collapse
Affiliation(s)
- Zhian Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Huilin Huang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jian Chen
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhenhao Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yanfeng Hu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Ruiyuan Liu
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|