1
|
Yang X, Zhang H, Wu Z, Chen Q, Zheng W, Shen Q, Wei Q, Shen JW, Guo Y. Tumor therapy utilizing dual-responsive nanoparticles: A multifaceted approach integrating calcium-overload and PTT/CDT/chemotherapy. J Control Release 2024; 376:646-658. [PMID: 39427774 DOI: 10.1016/j.jconrel.2024.10.029] [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: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
The advancement of rational nano drug delivery systems offers robust tools for achieving synergistic therapeutic outcomes in tumor treatment. In this study, we present the development of pH and near-infrared laser dual-responsive nanoparticles (DOX-CuS@CaCO3@PL-PEG, DCCP NPs) based on calcium carbonate, utilizing a one-pot gas diffusion reaction. These nanoparticles enable combined photothermal therapy (PTT), chemodynamic therapy (CDT), chemotherapy, and Ca2+-overloading synergistic therapy. Doxorubicin (DOX) and copper sulfide (CuS) NPs were co-loaded in CaCO3, followed by PEG surface functionalization. The presence of PEG enhanced the stability of DCCP NPs in aqueous environments. Controlled release of DOX, CuS NPs, and Ca2+ occurs specifically in the acidic tumor microenvironment. Released DOX enhances chemotherapy efficiency, while CuS NPs, upon laser irradiation, induce thermal damage, promoting further drug release and cellular uptake. Additionally, CuS NPs in our system consume excess GSH and generate toxic hydroxyl radicals (·OH) through a Fenton-like reaction, contributing to CDT. These radicals not only directly eliminate tumor cells but also disrupt mitochondrial Ca2+ buffering capacity. Furthermore, Ca2+ released from CaCO3 induces Ca2+-overloading, intensifying mitochondrial disruption and oxidative damage. The synergistic combination of PTT, CDT, chemotherapy, and Ca2+-overloading showcases significant therapeutic potential, indicating broad applications in tumor therapy. This multifaceted approach holds promise for advancing the field of tumor therapeutics.
Collapse
Affiliation(s)
- Xiaorong Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hong Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Zehua Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wei Zheng
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiying Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qiaolin Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; State Key Lab of Silicon Materials, Zhejiang University, Hangzhou 310027, China.
| | - Jia-Wei Shen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Yong Guo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| |
Collapse
|
2
|
Roy S, Gu J, Xia W, Mi C, Guo B. Advancements in manganese complex-based MRI agents: Innovations, design strategies, and future directions. Drug Discov Today 2024; 29:104101. [PMID: 39019428 DOI: 10.1016/j.drudis.2024.104101] [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: 04/21/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
This review focuses on the advancements in manganese (Mn) complex-based magnetic resonance imaging (MRI) agents for imaging different diseases. Here we emphasize the unique redox properties of Mn to deliver innovative MRI contrast agents, including small molecules, nanoparticles (NPs), metal-organic frameworks (MOFs), and polymer hybrids. Aspects of their rational design have been discussed, including size dependence, morphology tuning, surface property enhancement, etc., while also discussing the existing challenges and potential solutions. The present work will inspire and motivate scientists to emphasize MRI-guided applications and bring clinical success in the coming years.
Collapse
Affiliation(s)
- Shubham Roy
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055 China
| | - Jingsi Gu
- Education Center and Experiments and Innovations, Harbin Institute of Technology, Shenzhen 518055, China
| | - Wujiong Xia
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055 China
| | - Chao Mi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China; Shenzhen Light Life Technology Co., Ltd., Shenzhen 518107, China; School of Advanced Engineering, Great Bay Institute for Advanced Study, Great Bay University, Dongguan, Guangdong 523000, China.
| | - Bing Guo
- School of Science, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Harbin Institute of Technology, Shenzhen 518055 China.
| |
Collapse
|
3
|
Adzavon KP, Zhao W, He X, Sheng W. Ferroptosis resistance in cancer cells: nanoparticles for combination therapy as a solution. Front Pharmacol 2024; 15:1416382. [PMID: 38962305 PMCID: PMC11219589 DOI: 10.3389/fphar.2024.1416382] [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: 04/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Ferroptosis is a form of regulated cell death (RCD) characterized by iron-dependent lipid peroxidation. Ferroptosis is currently proposed as one of the most promising means of combating tumor resistance. Nevertheless, the problem of ferroptosis resistance in certain cancer cells has been identified. This review first, investigates the mechanisms of ferroptosis induction in cancer cells. Next, the problem of cancer cell resistance to ferroptosis, as well as the underlying mechanisms is discussed. Recently discovered ferroptosis-suppressing biomarkers have been described. The various types of nanoparticles that can induce ferroptosis are also discussed. Given the ability of nanoparticles to combine multiple agents, this review proposes nanoparticle-based ferroptosis cell death as a viable method of circumventing this resistance. This review suggests combining ferroptosis with other forms of cell death, such as apoptosis, cuproptosis and autophagy. It also suggests combining ferroptosis with immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Wang Sheng
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, China
| |
Collapse
|
4
|
Liu YJ, Dong SH, Hu WH, Chen QL, Zhang SF, Song K, Han ZC, Li MM, Han ZT, Liu WB, Zhang XS. A multifunctional biomimetic nanoplatform for image-guideded photothermal-ferroptotic synergistic osteosarcoma therapy. Bioact Mater 2024; 36:157-167. [PMID: 38463554 PMCID: PMC10924166 DOI: 10.1016/j.bioactmat.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Much effort has been devoted to improving treatment efficiency for osteosarcoma (OS). However, most current approaches result in poor therapeutic responses, thus indicating the need for the development of other therapeutic options. This study developed a multifunctional nanoparticle, PDA-MOF-E-M, an aggregation of OS targeting, programmed death targeting, and near-infrared (NIR)-aided targeting. At the same time, a multifunctional nanoparticle that utilises Fe-MOFs to create a cellular iron-rich environment and erastin as a ferroptosis inducer while ensuring targeted delivery to OS cells through cell membrane encapsulation is presented. The combination of PDA-MOF-E-M and PTT increased intracellular ROS and LPO levels and induced ferroptosis-related protein expression. A PDA-based PTT combined with erastin showed significant synergistic therapeutic improvement in the anti-tumour efficiency of the nanoparticle in vitro and vivo. The multifunctional nanoparticle efficiently prevents the osteoclasia progression of OS xenograft bone tumors in vivo. Finally, this study provides guidance and a point of reference for clinical approaches to treating OS.
Collapse
Affiliation(s)
- Yu-jie Liu
- Department of Orthopedic Oncology and Spine Tumor Center, Changzheng Hospital, Second Military Medical University, Shanghai, 200001, China
| | - Su-he Dong
- PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Wen-hao Hu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Qiao-ling Chen
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Shao-fu Zhang
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Kai Song
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Zhen-chuan Han
- PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Meng-meng Li
- Department of Anesthesiology, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Zhi-tao Han
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Jiangsu, China
| | - Wei-bo Liu
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| | - Xue-song Zhang
- Department of Orthopaedics, The Fourth Medical Centre, Chinese PLA General Hospital, 51 Fucheng Road, Haidian District, Beijing, 100048, China
| |
Collapse
|
5
|
Dhas N, Kudarha R, Tiwari R, Tiwari G, Garg N, Kumar P, Kulkarni S, Kulkarni J, Soman S, Hegde AR, Patel J, Garkal A, Sami A, Datta D, Colaco V, Mehta T, Vora L, Mutalik S. Recent advancements in nanomaterial-mediated ferroptosis-induced cancer therapy: Importance of molecular dynamics and novel strategies. Life Sci 2024; 346:122629. [PMID: 38631667 DOI: 10.1016/j.lfs.2024.122629] [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: 11/23/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Ferroptosis is a novel type of controlled cell death resulting from an imbalance between oxidative harm and protective mechanisms, demonstrating significant potential in combating cancer. It differs from other forms of cell death, such as apoptosis and necrosis. Molecular therapeutics have hard time playing the long-acting role of ferroptosis induction due to their limited water solubility, low cell targeting capacity, and quick metabolism in vivo. To this end, small molecule inducers based on biological factors have long been used as strategy to induce cell death. Research into ferroptosis and advancements in nanotechnology have led to the discovery that nanomaterials are superior to biological medications in triggering ferroptosis. Nanomaterials derived from iron can enhance ferroptosis induction by directly releasing large quantities of iron and increasing cell ROS levels. Moreover, utilizing nanomaterials to promote programmed cell death minimizes the probability of unfavorable effects induced by mutations in cancer-associated genes such as RAS and TP53. Taken together, this review summarizes the molecular mechanisms involved in ferroptosis along with the classification of ferroptosis induction. It also emphasized the importance of cell organelles in the control of ferroptosis in cancer therapy. The nanomaterials that trigger ferroptosis are categorized and explained. Iron-based and noniron-based nanomaterials with their characterization at the molecular and cellular levels have been explored, which will be useful for inducing ferroptosis that leads to reduced tumor growth. Within this framework, we offer a synopsis, which traverses the well-established mechanism of ferroptosis and offers practical suggestions for the design and therapeutic use of nanomaterials.
Collapse
Affiliation(s)
- Namdev Dhas
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Ritu Kudarha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Ruchi Tiwari
- Pranveer Singh Institute of Technology (Pharmacy), Kalpi road, Bhauti, Kanpur 208020, Uttar Pradesh, India
| | - Gaurav Tiwari
- Pranveer Singh Institute of Technology (Pharmacy), Kalpi road, Bhauti, Kanpur 208020, Uttar Pradesh, India
| | - Neha Garg
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Praveen Kumar
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Jahnavi Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Soji Soman
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Aswathi R Hegde
- Faculty of Pharmacy, M S Ramaiah University of Applied Sciences, New BEL Road, MSR Nagar, Bangalore 560054, Karnataka, India
| | | | - Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India; Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Anam Sami
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Viola Colaco
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Tejal Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat 382481, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.
| |
Collapse
|
6
|
Sun B, Zheng X, Zhang X, Zhang H, Jiang Y. Oxaliplatin-Loaded Mil-100(Fe) for Chemotherapy-Ferroptosis Combined Therapy for Gastric Cancer. ACS OMEGA 2024; 9:16676-16686. [PMID: 38617668 PMCID: PMC11007804 DOI: 10.1021/acsomega.4c00658] [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: 01/20/2024] [Revised: 02/26/2024] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
Oxaliplatin (Oxa) is a commonly used chemotherapy drug in the treatment of gastric cancer, but its toxic side effects and drug resistance after long-term use have seriously limited its efficacy. Loading chemotherapy drugs with nanomaterials and delivering them to the tumor site are common ways to overcome the above problems. However, nanomaterials as carriers do not have therapeutic functions on their own, and the effect of single chemotherapy is relatively limited, so there is still room for progress in related research. Herein, we construct Oxa@Mil-100(Fe) nanocomposites by loading Oxa with a metal-organic framework (MOF) Mil-100(Fe) with high biocompatibility and a large specific surface area. The pore structure of Mil-100(Fe) is conducive to a large amount of Oxa loading with a drug-loading rate of up to 27.2%. Oxa@Mil-100(Fe) is responsive to the tumor microenvironment (TME) and can release Oxa and Fe3+ under external stimulation. On the one hand, Oxa can inhibit the synthesis of DNA and induce the apoptosis of gastric cancer cells. On the other hand, Fe3+ can clear overexpressed glutathione (GSH) in TME and be reduced to Fe2+, inhibiting the activity of glutathione peroxidase 4 (GPX4), leading to the accumulation of intracellular lipid peroxides (LPO), and at the same time releasing a large number of reactive oxygen species (ROS) through the Fenton reaction, inducing ferroptosis in gastric cancer cells. With the combination of apoptosis and ferroptosis, Oxa@Mil-100(Fe) shows a good therapeutic effect, and the killing effect on gastric cancer cells is obvious. In a nude mouse model of subcutaneous tumor transplantation, Oxa@Mil-100(Fe) shows a significant inhibitory effect on tumor growth, with an inhibition rate of nearly 60%. In addition to its excellent antitumor activity, Oxa@Mil-100(Fe) has no obvious toxic or side effects. This study provides a new idea and method for the combined treatment of gastric cancer.
Collapse
Affiliation(s)
- Boyao Sun
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Xuewei Zheng
- Department
of Radiology, China-Japan Union Hospital
of Jilin University, Changchun 130031, P. R. China
| | - Xiaoyu Zhang
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Huaiyu Zhang
- Department
of Rehabilitation Medicine, China-Japan
Union Hospital of Jilin University, Changchun 130031, P. R. China
| | - Yang Jiang
- Department
of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, P. R. China
| |
Collapse
|
7
|
Wang J, Zhao W, Zhang Z, Liu X, Xie T, Wang L, Xue Y, Zhang Y. A Journey of Challenges and Victories: A Bibliometric Worldview of Nanomedicine since the 21st Century. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308915. [PMID: 38229552 DOI: 10.1002/adma.202308915] [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: 08/31/2023] [Revised: 11/18/2023] [Indexed: 01/18/2024]
Abstract
Nanotechnology profoundly affects the advancement of medicine. Limitations in diagnosing and treating cancer and chronic diseases promote the growth of nanomedicine. However, there are very few analytical and descriptive studies regarding the trajectory of nanomedicine, key research powers, present research landscape, focal investigative points, and future outlooks. Herein, articles and reviews published in the Science Citation Index Expanded of Web of Science Core Collection from first January 2000 to 18th July 2023 are analyzed. Herein, a bibliometric visualization of publication trends, countries/regions, institutions, journals, research categories, themes, references, and keywords is produced and elaborated. Nanomedicine-related academic output is increasing since the COVID-19 pandemic, solidifying the uneven global distribution of research performance. While China leads in terms of publication quantity and has numerous highly productive institutions, the USA has advantages in academic impact, commercialization, and industrial value. Nanomedicine integrates with other disciplines, establishing interdisciplinary platforms, in which drug delivery and nanoparticles remain focal points. Current research focuses on integrating nanomedicine and cell ferroptosis induction in cancer immunotherapy. The keyword "burst testing" identifies promising research directions, including immunogenic cell death, chemodynamic therapy, tumor microenvironment, immunotherapy, and extracellular vesicles. The prospects, major challenges, and barriers to addressing these directions are discussed.
Collapse
Affiliation(s)
- Jingyu Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Wenling Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Xingzi Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Tong Xie
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Lan Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Yuzhou Xue
- Department of Cardiology, Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, State Key Laboratory of Vascular Homeostasis and Remodeling Peking University, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yuemiao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| |
Collapse
|
8
|
Lin X, Chen H, Deng T, Cai B, Xia Y, Xie L, Wang H, Huang C. Improved Immune Response for Colorectal Cancer Therapy Triggered by Multifunctional Nanocomposites with Self-Amplifying Antitumor Ferroptosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13481-13495. [PMID: 38456402 DOI: 10.1021/acsami.3c16813] [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: 03/09/2024]
Abstract
Ferroptosis, as a type of regulated cell death, can trigger the release of damage-associated molecular patterns from cancer cells and lead to the enhancement of immune recognition. Fenton reaction-mediated chemodynamic therapy could initiate ferroptosis by generating lipid peroxides, but its efficiency would be greatly restricted by the insufficient H2O2 and antioxidant system within the tumor. Herein, this work reports the successful preparation of H2O2 self-supplied and glutathione (GSH)-depletion therapeutic nanocomposites (Cu2O@Au) through in situ growth of Au nanoparticles on the surface of cuprous oxide (Cu2O) nanospheres. Upon delivery into cancer cells, the released Cu2O could consume endogenous H2S within colorectal cancer cells to form Cu31S16 nanoparticles, while the released Au NPs could catalyze glucose to generate H2O2 and gluconic acid. The self-supplying endogenous H2O2 and lower acidity could amplify the Cu ion-induced Fenton-like reaction. Meanwhile, the consumption of glucose would reduce GSH generation by disrupting the pentose phosphate pathway. Additionally, the Cu2+/Cu+ catalytic cycle promotes the depletion of GSH, leading to lipid peroxide accumulation and ferroptosis. It was found that the onset of ferroptosis triggered by Cu2O@Au could initiate immunologic cell death, promote dendritic cell maturation and T-cell infiltration, and finally enhance the antitumor efficacy of the PD-L1 antibody. In summary, this collaborative action produces a remarkable antitumor effect, which provides a promising treatment strategy for colorectal cancer.
Collapse
Affiliation(s)
- Xiaosheng Lin
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Hongwu Chen
- Shantou University Medical College, Shantou 515041, China
| | - Tingting Deng
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Binghui Cai
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Yubin Xia
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Lei Xie
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Huaiming Wang
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Cong Huang
- The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
9
|
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. [PMID: 38462885 DOI: 10.1111/odi.14925] [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: 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
|
10
|
Oryani MA, Nosrati S, Javid H, Mehri A, Hashemzadeh A, Karimi-Shahri M. Targeted cancer treatment using folate-conjugated sponge-like ZIF-8 nanoparticles: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1377-1404. [PMID: 37715816 DOI: 10.1007/s00210-023-02707-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/02/2023] [Indexed: 09/18/2023]
Abstract
ZIF-8 (zeolitic imidazolate framework-8) is a potential drug delivery system because of its unique properties, which include a large surface area, a large pore capacity, a large loading capacity, and outstanding stability under physiological conditions. ZIF-8 nanoparticles may be readily functionalized with targeting ligands for the identification and absorption of particular cancer cells, enhancing the efficacy of chemotherapeutic medicines and reducing adverse effects. ZIF-8 is also pH-responsive, allowing medication release in the acidic milieu of cancer cells. Because of its tunable structure, it can be easily functionalized to design cancer-specific targeted medicines. The delivery of ZIF-8 to cancer cells can be facilitated by folic acid-conjugation. Hence, it can bind to overexpressed folate receptors on the surface of cancer cells, which holds the promise of reducing unwanted deliveries. As a result of its importance in cancer treatment, the folate-conjugated ZIF-8 was the major focus of this review.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shamim Nosrati
- Department of Clinical Biochemistry, Faculty of Medicine, Azad Shahroud University, Shahroud, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Mehri
- Endoscopic and Minimally Invasive Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
11
|
Li C, Ding Z, Han Y. Mn-Doped Nano-Hydroxyapatites as Theranostic Agents with Tumor pH-Amplified MRI-Signal Capabilities for Guiding Photothermal Therapy. Int J Nanomedicine 2023; 18:6101-6118. [PMID: 37915749 PMCID: PMC10617543 DOI: 10.2147/ijn.s429336] [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: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Background The integration of diagnostic and therapeutic functions into a biosafe nanoplatform with intelligent response functions at the tumor microenvironment (TME) is a promising strategy for cancer therapy. Methods Mn-doped nano-hydroxyapatite (nHAPMn) nanoparticles were successfully prepared via a simple coprecipitation method for magnetic resonance imaging (MRI)-guided photothermal therapy. This study is the first to report on the use of Mn to render biodegradable hydroxyapatite suitable for MRI and effective photothermal therapy (PTT) simultaneously by regulating the pH of nHAPMn during the preparation process. Results Combined with near-infrared (NIR) laser irradiation, a photothermal conversion efficiency of 26% and effective photothermal lethality in vitro were achieved. Moreover, the degradation of nHAPMn led to the release of Mn ions and amplified the MRI signals in an acidic TME, which confirmed that nHAPMn had a good pH-responsive MRI capacity in solid tumors. In animal experiments, tumors in the nHAPMn5+NIR group completely abated after 14 days of treatment, with no significant recurrence during the experiment. Conclusion Therefore, nHAPMn is promising as a nanotheranostic agent and can be effective in clinical diagnosis and therapy for treating cancer.
Collapse
Affiliation(s)
- Chengyu Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, People’s Republic of China
| | - Ziyou Ding
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, People’s Republic of China
| | - Yingchao Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, People’s Republic of China
| |
Collapse
|
12
|
Wang J, Yang J, Liu K, Yuan J, Shi Y, Li H, Zhao L. Tumor targeted cancer membrane-camouflaged ultra-small Fe nanoparticles for enhanced collaborative apoptosis and ferroptosis in glioma. Mater Today Bio 2023; 22:100780. [PMID: 37680585 PMCID: PMC10480784 DOI: 10.1016/j.mtbio.2023.100780] [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: 05/31/2023] [Revised: 07/28/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023] Open
Abstract
Glioma is recognized as the most common and aggressive primary brain tumor in adults. Owing to the occurrence of drug resistance and the failure of drug to penetrate the blood-brain barrier (BBB), there is no effective strategy for the treatment of glioma. The main objective of this study was to develop a biomimetic glioma C6 cell membrane (C6M) derived nanovesicles (DOX-FN/C6M-NVs) loaded with doxorubicin (DOX) and ultra-small Fe nanoparticles (FN) for accomplishing the effective brain tumor-targeted delivery of DOX and improving anti-cancer efficacy via inducing collaborative apoptosis and ferroptosis. The findings revealed that employing C6M-NVs as a carrier significantly improved the therapeutic efficacy by enabling evasion of immune surveillance, facilitating targeted drug delivery to tumor sites, and minimizing cardiotoxicity and adverse effects associated with DOX. DOX-FN/C6M-NVs exhibited more potent anti-tumor effects as compared with free DOX by promoting DOX-mediated apoptosis and accelerating ferroptosis via the mediation of FN. This study suggested that DOX-FN/C6M-NVs as the potential inducer of ferroptosis and apoptosis conferred effective tumor suppression in the treatment of glioma.
Collapse
Affiliation(s)
- Jingchen Wang
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Jian Yang
- Life Science Institution, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Kang Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Jiayu Yuan
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Hongdan Li
- Life Science Institution, Jinzhou Medical University, Jinzhou, 121000, PR China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000, PR China
| |
Collapse
|
13
|
Ou R, Aodeng G, Ai J. Advancements in the Application of the Fenton Reaction in the Cancer Microenvironment. Pharmaceutics 2023; 15:2337. [PMID: 37765305 PMCID: PMC10536994 DOI: 10.3390/pharmaceutics15092337] [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: 08/11/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer is a complex and multifaceted disease that continues to be a global health challenge. It exerts a tremendous burden on individuals, families, healthcare systems, and society as a whole. To mitigate the impact of cancer, concerted efforts and collaboration on a global scale are essential. This includes strengthening preventive measures, promoting early detection, and advancing effective treatment strategies. In the field of cancer treatment, researchers and clinicians are constantly seeking new approaches and technologies to improve therapeutic outcomes and minimize adverse effects. One promising avenue of investigation is the utilization of the Fenton reaction, a chemical process that involves the generation of highly reactive hydroxyl radicals (·OH) through the interaction of hydrogen peroxide (H2O2) with ferrous ions (Fe2+). The generated ·OH radicals possess strong oxidative properties, which can lead to the selective destruction of cancer cells. In recent years, researchers have successfully introduced the Fenton reaction into the cancer microenvironment through the application of nanotechnology, such as polymer nanoparticles and light-responsive nanoparticles. This article reviews the progress of the application of the Fenton reaction, catalyzed by polymer nanoparticles and light-responsive nanoparticles, in the cancer microenvironment, as well as the potential applications and future development directions of the Fenton reaction in the field of tumor treatment.
Collapse
Affiliation(s)
| | | | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (R.O.); (G.A.)
| |
Collapse
|
14
|
Wang J, Chu Y, Zhao Z, Zhang C, Chen Q, Ran H, Cao Y, Wu C. Piezoelectric enhanced sulfur doped graphdiyne nanozymes for synergistic ferroptosis-apoptosis anticancer therapy. J Nanobiotechnology 2023; 21:311. [PMID: 37660123 PMCID: PMC10474662 DOI: 10.1186/s12951-023-02059-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/11/2023] [Indexed: 09/04/2023] Open
Abstract
Graphdiyne has excellent potential due to its enzymatic properties. Metal-free sulfur-doped Graphdiyne (S-GDY) has piezoelectric characteristics, and ultrasonic excitation of S-GDY enhances peroxidase activity. It can turn hydrogen peroxide into toxic hydroxyl radicals and induce apoptosis in 4T1 cells. More importantly, the ultrasound (US) enhanced nanozyme induced 4T1 cell ferroptosis by promoting an imbalanced redox reaction due to glutathione depletion and glutathione peroxidase 4 inactivation. S-GDY exhibited enhanced nanozyme activity in vitro and in vivo that may directly trigger apoptosis-ferroptosis for effective tumor therapy. Altogether, this study was expected to provide new insights into the design of piezoelectric catalytic nanozyme and expand their application in the catalytic therapy of tumors.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yinzhu Chu
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhiyu Zhao
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Cong Zhang
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Qi Chen
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, State Key Laboratory of Ultrasound in Medicine and Engineering,, Chongqing Medical University, Chongqing, 400010, China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, State Key Laboratory of Ultrasound in Medicine and Engineering,, Chongqing Medical University, Chongqing, 400010, China.
| | - Changjun Wu
- Department of Ultrasound, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
15
|
Dai X, Li L, Li M, Yan X, Li J, Mao H, Wang C, Xu H. One pot preparation of muti-mode nanoplatform to combat ovarian cancer. Biomed Pharmacother 2023; 165:115172. [PMID: 37473681 DOI: 10.1016/j.biopha.2023.115172] [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: 05/02/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
Ovarian cancer is one of the most common gynecological cancers with high mortality rate. The battle against ovarian cancer usually impaired by the evolved multidrug resistance (MDR) phenotype as well as metastasis in cancers, which urgently call for the development of multi-mode strategies to overcome the MDR and reduce metastasis. Considering the good benefits of ferroptosis and photothermal therapy (PTT) in cancer management, we herein proposed a facile way to construct nanoparticle platform (Fe-Dox/PVP) composed of ferric chloride, doxorubicin (Dox) and polyvinyl pyrrolidone (PVP) for the multi-mode therapy of ovarian cancer using chemotherapy, ferroptosis and mild hypothermia PTT. Our results demonstrated that Fe-Dox/PVP with mild hypothermia was shown to have improved endosomal escape/drug delivery, enhanced ferroptosis induction and good tumor targeting effects. Most importantly, the integration of all three effects into one platform provided increased anti-metastasis effect and promising in vitro/in vivo anticancer performance with high biocompatibility. In this study, we offer a facile and robust way to prepare a multi-mode nanoplatform to combat ovarian cancer, which can be further extended for the management of many other cancers.
Collapse
Affiliation(s)
- Xiuliang Dai
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Lingjun Li
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Minhui Li
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Xiaomeng Yan
- Obstetrics and Gynecology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Jinhang Li
- Dalian Medical University, Dalian, China
| | - Hao Mao
- Dalian Medical University, Dalian, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China.
| | - Hongbin Xu
- Obstetrics and Gynecology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China; School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China.
| |
Collapse
|
16
|
Wen S, Shi Y, Zhang Y, Chang Q, Hu H, Deng X, Xie Y. pH-Activated Ce-Doped Molybdenum Oxide Nanoclusters for Tumor Microenvironment Responsive Photothermal and Chemodynamic Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023. [PMID: 37436959 DOI: 10.1021/acs.langmuir.3c01075] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Molybdenum-based nanomaterials have shown promise for anticancer treatment due to their strong photothermal and redox-activated capabilities. Herein, we have fabricated cerium-doped MoOx (Ce-MoOv) with tunable Mo/Ce molar ratios by a one-pot method and investigated their effect on chemodynamic therapy (CDT) and photothermal therapy (PTT). It is found that Ce-MoOv can self-assemble into nanoclusters in acidic conditions and the increasing Ce amount will generate oxygen vacancy defects and induce the valence change of Mo6+/Mo5+ and Ce4+/Ce3+, which leads to strong near-infrared absorption with high photothermal conversion efficiency of 71.31 and 49.86% for 808 and 1064 nm. Other than photothermal conversion, the materials demonstrate pH-/glutathione (GSH)-activated photoacoustic (PA) imaging capability in vitro. In addition, Ce-MoOv acts as a CDT reagent capable of converting endogenous H2O2 to two types of reactive oxygen species (•OH, 1O2) while depleting GSH. Ce-MoOv demonstrates an excellent therapeutic effect against HCT116 cells and effectively reduces the intracellular GSH level and significantly increases the number of reactive radicals under 1064 nm laser irradiation as compared with the no-laser group in vitro. This work provides a new paradigm using lanthanide-doped polymetallic oxides for pH-/GSH-responsive photothermal/chemodynamic therapy with PA imaging ability.
Collapse
Affiliation(s)
- Shuangyan Wen
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yejiao Shi
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Yanan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Qing Chang
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Honggang Hu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiaoyong Deng
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Yijun Xie
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
17
|
Zhang X, Sathiyaseelan A, Naveen KV, Lu Y, Wang MH. Research progress in green synthesis of manganese and manganese oxide nanoparticles in biomedical and environmental applications - A review. CHEMOSPHERE 2023:139312. [PMID: 37354955 DOI: 10.1016/j.chemosphere.2023.139312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023]
Abstract
Nanomaterials and nanotechnology have this unassailable position for environmental remediation and medicine. Currently, global environmental pollution and public health problems are increasing and need to be urgently addressed. Manganese (Mn) is one of the essential metal elements for plants and animals, it is necessary to integrate with nanotechnology. Mn and Mn oxide (MnO) nanoparticles (NPs) have applications in dye degradation, biomedicine, electrochemical sensors, plant and animal growth, and catalysis. However, the current research is limited, especially in terms of optimal synthesis of Mn and MnO NPs, separation, purification conditions, and the development of potential application areas is too basic and do not support by in-depth studies. Hence, this review comprehensively discusses the classification, green synthesis methods, and applications of Mn and MnO NPs in biomedical, environmental, and other fields and gives a perspective for the future.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Anbazhagan Sathiyaseelan
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Kumar Vishven Naveen
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Yuting Lu
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
18
|
Zhang R, Kang R, Tang D. Ferroptosis in gastrointestinal cancer: From mechanisms to implications. Cancer Lett 2023; 561:216147. [PMID: 36965540 DOI: 10.1016/j.canlet.2023.216147] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Ferroptosis is a form of regulated cell death that is initiated by excessive lipid peroxidation that results in plasma membrane damage and the release of damage-associated molecular patterns. In recent years, ferroptosis has gained significant attention in cancer research due to its unique mechanism compared to other forms of regulated cell death, especially caspase-dependent apoptotic cell death. Gastrointestinal (GI) cancer encompasses malignancies that arise in the digestive tract, including the stomach, intestines, pancreas, colon, liver, rectum, anus, and biliary system. These cancers are a global health concern, with high incidence and mortality rates. Despite advances in medical treatments, drug resistance caused by defects in apoptotic pathways remains a persistent challenge in the management of GI cancer. Hence, exploring the role of ferroptosis in GI cancers may lead to more efficacious treatment strategies. In this review, we provide a comprehensive overview of the core mechanism of ferroptosis and discuss its function, regulation, and implications in the context of GI cancers.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
19
|
Liu F, Tang L, Li Q, Chen L, Pan Y, Yin Z, He J, Tian J. Single-cell transcriptomics uncover the key ferroptosis regulators contribute to cancer progression in head and neck squamous cell carcinoma. Front Mol Biosci 2022; 9:962742. [PMID: 36003082 PMCID: PMC9393303 DOI: 10.3389/fmolb.2022.962742] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
The mechanism underlying the association between the development of head and neck squamous cell carcinoma (HNSCC) and ferroptosis is unclear. We analyzed the transcriptomes of 5902 single cells from a single-cell RNA-sequencing (scRNA-seq) dataset. They then aggregate into B cells, epithelial cells, fibroblasts, germ cells, mesenchymal cells, cancer stem cells, stem cells, T cells and endometrial cells, respectively. Our study shows that multiple pathways are significantly enriched in HNSCC development including extracellular matrix structural components, humoral immune responses, and muscle contraction. Differentially expressed genes analysis in Pseudotime analysis, pathway and biological function indicated that there was a significant correlation in the ferroptosis pathway. Furthermore, higher ferroptosis potential index (FPI) scores were significantly associated with worse overall survival prognosis in HNSCC patients. Pseudo-temporal, survival analyses and immunohistochemistry identified multiple central genes in HNSCC development, including ACSL1, SLC39A14, TFRC, and PRNP genes, and indicated associated ferroptosis. Overall, our study detected ferroptosis-related features is closely correlated with HNSCC prognosis and development, and deserved candidates suitable for immunotherapy treatment strategies determination for HNSCC patients.
Collapse
Affiliation(s)
- Fei Liu
- Cancer Screening Center, Department of Health Management, Guangdong Second Provincial General Hospital, Guangdong, China
| | - Lindong Tang
- Institute of Hematology School of Medicine Jinan University, Guangdong, China
| | - Qing Li
- Department of Stomatology Guangdong Second Provincial General Hospital, Guangdong, China
| | - Leihui Chen
- Department of Stomatology Guangdong Second Provincial General Hospital, Guangdong, China
| | - Yuyue Pan
- Department of Stomatology Guangdong Second Provincial General Hospital, Guangdong, China
| | - Zhao Yin
- Department of Hematology Guangdong Second Provincial General Hospital, Guangdong, China
- *Correspondence: Zhao Yin, ; Jingjun He, ; Junzhang Tian,
| | - Jingjun He
- Cancer Screening Center, Department of Health Management, Guangdong Second Provincial General Hospital, Guangdong, China
- *Correspondence: Zhao Yin, ; Jingjun He, ; Junzhang Tian,
| | - Junzhang Tian
- Cancer Screening Center, Department of Health Management, Guangdong Second Provincial General Hospital, Guangdong, China
- *Correspondence: Zhao Yin, ; Jingjun He, ; Junzhang Tian,
| |
Collapse
|