1
|
Wu Q, Feng Y, Lepoitevin M, Yu M, Serre C, Ge J, Huang Y. Metal-Organic Frameworks: Unlocking New Frontiers in Cardiovascular Diagnosis and Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416302. [PMID: 40270437 DOI: 10.1002/advs.202416302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/14/2025] [Indexed: 04/25/2025]
Abstract
Cardiovascular disease (CVD) is one of the most critical diseases which is the predominant cause of death in the world. Early screening and diagnosis of the disease and effective treatment after diagnosis play an important role in the patient's recovery. Metal-organic frameworks (MOFs), a kind of hybrid ordered micro or meso-porous materials, constructed by metal nodes or clusters with organic ligands, due to their special features like high porosity and specific surface area, open metal sites, or ligand tunability, are widely used in various areas including gas storage, catalysis, sensors, biomedicine. Recently, advances in MOFs are bringing new developments and opportunities for the healthcare industry including the theranostic of CVD. In this review, the applications of MOFs are illustrated in the diagnosis and therapy of CVD, including biomarker detection, imaging, drug delivery systems, therapeutic gas delivery platforms, and nanomedicine. Also, the toxicity and biocompatibility of MOFs are discussed. By providing a comprehensive summary of the role played by MOFs in the diagnosis and treatment of CVDs, it is hoped to promote the future applications of MOFs in disease theranostics, especially in CVDs.
Collapse
Affiliation(s)
- Qilu Wu
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Yuxiao Feng
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, P. R. China
| | - Mathilde Lepoitevin
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, 75005, France
| | - Meng Yu
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, 75005, France
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening & Guangdong-Hongkong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Christian Serre
- Institut des Matériaux Poreux de Paris, ENS, ESPCI Paris, CNRS, PSL University, Paris, 75005, France
| | - Jun Ge
- Key Lab for Industrial Biocatalysis, Ministry of Education, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, P. R. China
- Beijing, 100084, P. R. China
| | - Yuan Huang
- Cardiac Surgery Centre, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, P. R. China
| |
Collapse
|
2
|
He Y, Jin Z, Wang Y, Wu C, He X, Weng W, Cai X, Cheng K. Multifunctional Double-Loaded Oral Nanoplatform for Computed Tomography Imaging-Guided and Integrated Treatment of Inflammatory Bowel Disease. ACS NANO 2025; 19:14893-14913. [PMID: 40106686 DOI: 10.1021/acsnano.4c18865] [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: 03/22/2025]
Abstract
Excessive reactive oxygen species, disruption of the epithelial barrier, immune dysregulation, and gut microbiota imbalance are key factors driving the onset of inflammatory bowel disease (IBD) and complicating its treatment. Prompt diagnosis of diseases and precise delivery of therapeutic agents to inflamed intestinal sites offer promising targeted strategies for effectively treating IBD. Here, a barium sulfate-based nanoplatform (BaSO4@PDA@CeO2/DSP, BPCD) for synergistic delivery of nanozymes and drugs was developed. With enhanced colonic retention after oral drug delivery, this nanoplatform enables precise and effective targeting of inflammatory sites and CT imaging guidance to address multiple factors contributing to IBD. A comprehensive therapeutic effect was achieved through the synergistic action of cerium oxide with the optimized Ce3+/Ce4+ ratio and sustained release of dexamethasone sodium phosphate. Benefiting from superior gastrointestinal stability, the nanoplatform is highly effective in treating IBD by alleviating oxidative stress, modulating macrophage polarization balance, gut flora composition, and repairing the epithelial barrier. BPCD inhibits the development of IBD through multiple mechanisms and has superior biocompatibility, emerging as a practical alternative to traditional IBD therapies.
Collapse
Affiliation(s)
- Yaoting He
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Center of Rehabilitation Biomedical Materials, Cyrus Tang Center for Sensor Materials and Applications, Zhejiang University, Hangzhou 310027, China
| | - Ziyang Jin
- Department of General Surgery, Minimally Invasive Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - YiFan Wang
- Department of General Surgery, Minimally Invasive Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Chengwei Wu
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Center of Rehabilitation Biomedical Materials, Cyrus Tang Center for Sensor Materials and Applications, Zhejiang University, Hangzhou 310027, China
| | - Xuzhao He
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Center of Rehabilitation Biomedical Materials, Cyrus Tang Center for Sensor Materials and Applications, Zhejiang University, Hangzhou 310027, China
| | - Wenjian Weng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Center of Rehabilitation Biomedical Materials, Cyrus Tang Center for Sensor Materials and Applications, Zhejiang University, Hangzhou 310027, China
| | - Xiujun Cai
- Department of General Surgery, Minimally Invasive Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Kui Cheng
- School of Materials Science and Engineering, State Key Laboratory of Silicon Materials, Center of Rehabilitation Biomedical Materials, Cyrus Tang Center for Sensor Materials and Applications, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
3
|
Zhao X, Qin Y, Li B, Wang Y, Liu J, Wang B, Zhao J, Yin J, Zhang L, Li J, Huang J, Chen K, Liu L, Wu Y. Genetically engineered biomimetic ATP-responsive nanozyme for the treatment of cardiac fibrosis. J Nanobiotechnology 2025; 23:10. [PMID: 39780203 PMCID: PMC11715444 DOI: 10.1186/s12951-024-03083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cardiac fibrosis plays a critical role in the progression of various forms of heart disease, significantly increasing the risk of sudden cardiac death. However, currently, there are no therapeutic strategies available to prevent the onset of cardiac fibrosis. METHODS AND RESULTS Here, biomimetic ATP-responsive nanozymes based on genetically engineered cell membranes are adapted to specifically recognize activated cardiac fibroblasts (CFs) for the treatment of cardiac fibrosis. By fusing the anti-FAP CAR genetically engineered cell membrane to zeolitic imidazole frameworks-90 (zif-90) cores loaded with antioxidant nanozymes CeO2 and siCTGF (siRNA targeting CTGF), these nanoparticles, called FM@zif-90/Ce/siR NPs, are demonstrated to effectively reduce the accumulation of myofibroblasts and the formation of fibrotic tissue, while restoring cardiac function. CONCLUSIONS These findings demonstrate that the combination of CeO2 and siCTGF has a beneficial curative effect on cardiac fibrosis, with significant translational potential.
Collapse
Affiliation(s)
- Xueli Zhao
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuze Qin
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bowen Li
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Wang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiao Liu
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Wang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Zhao
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiaqi Yin
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lanlan Zhang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jing Li
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Junzhe Huang
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Kun Chen
- Department of Anatomy, Histology and Embryology and K.K. Leung Brain Research Centre, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Liwen Liu
- Department of Ultrasound, Xijing Hypertrophic Cardiomyopathy Center, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Yuanming Wu
- Department of Laboratory, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Biochemistry and Molecular Biology, Shaanxi Provincial Key Laboratory of Clinical Genetics, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Li Y, Chen W, Yin J, Xia S, Jiang Y, Ge Q, Liu J, Wang M, Hou Z, Bai Y, Shi P. Biomineralized ZIF-8 Encapsulating SOD from Hydrogenobacter Thermophilus: Maintaining Activity in the Intestine and Alleviating Intestinal Oxidative Stress. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402812. [PMID: 39350464 DOI: 10.1002/smll.202402812] [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: 04/08/2024] [Revised: 09/17/2024] [Indexed: 12/13/2024]
Abstract
Oxidative stress is a major factor leading to inflammation and disease occurrence, and superoxide dismutase (SOD) is a crucial antioxidative metalloenzyme capable of alleviating oxidative stress. In this study, a novel thermostable SOD gene is obtained from the Hydrogenobacter thermophilus strain (HtSOD), transformed and efficiently expressed in Escherichia coli with an activity of 3438 U mg-1, exhibiting excellent thermal stability suitable for scalable production. However, the activity of HtSOD is reduced to less than 10% under the acidic environment. To address the acid resistance and gastrointestinal stability issues, a biomimetic mineralization approach is employed to encapsulate HtSOD within the ZIF-8 (HtSOD@ZIF-8). Gastrointestinal simulation results show that HtSOD@ZIF-8 maintained 70% activity in simulated gastric fluid for 2 h, subsequently recovering to 97% activity in simulated intestinal fluid. Cell and in vivo experiments indicated that HtSOD@ZIF-8 exhibited no cytotoxicity and do not impair growth performance. Furthermore, HtSOD@ZIF-8 increased the relative abundance of beneficial microbiota such as Dubosiella and Alistipes, mitigated oxonic stress and intestinal injury by reducing mitochondrial and total reactive oxygen species (ROS) levels in diquat-induced. Together, HtSOD@ZIF-8 maintains and elucidates activity in the intestine and biocompatibility, providing insights into alleviating oxidative stress in hosts and paving the way for scalable production.
Collapse
Affiliation(s)
- Yuying Li
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
| | - Weihua Chen
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300384, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410000, China
| | - Siting Xia
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410000, China
| | - Yayun Jiang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410000, China
| | - Qianqian Ge
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410000, China
| | - Jinping Liu
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, 410000, China
| | - Mansheng Wang
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
| | - Zhenping Hou
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
| | - Yingguo Bai
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Pengjun Shi
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, Hunan, 410205, China
| |
Collapse
|
5
|
Han D, Wang F, Shen D. Nanomedicines as Guardians of the Heart: Unleashing the Power of Antioxidants to Alleviate Myocardial Ischemic Injury. Theranostics 2024; 14:5336-5370. [PMID: 39267789 PMCID: PMC11388064 DOI: 10.7150/thno.99961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Ischemic heart disease (IHD) is increasingly recognized as a significant cardiovascular disease with a growing global incidence. Interventions targeting the oxidative microenvironment have long been pivotal in therapeutic strategies. However, many antioxidant drugs face limitations due to pharmacokinetic and delivery challenges, such as short half-life, poor stability, low bioavailability, and significant side effects. Fortunately, nanotherapies exhibit considerable potential in addressing IHD. Nanomedicines offer advantages such as passive/active targeting, prolonged circulation time, enhanced bioavailability, and diverse carrier options. This comprehensive review explores the advancements in nanomedicines for mitigating IHD through oxidative stress regulation, providing an extensive overview for researchers in the field of antioxidant nanomedicines. By inspiring further research, this study aims to accelerate the development of novel therapies for myocardial injury.
Collapse
Affiliation(s)
- Dongjian Han
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Fuhang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Deliang Shen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
6
|
Liu W, Li Y, Wang Y, Feng Y. Bioactive Metal-Organic Frameworks as a Distinctive Platform to Diagnosis and Treat Vascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310249. [PMID: 38312082 DOI: 10.1002/smll.202310249] [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: 11/09/2023] [Revised: 01/07/2024] [Indexed: 02/06/2024]
Abstract
Vascular diseases (VDs) pose the leading threat worldwide due to high morbidity and mortality. The detection of VDs is commonly dependent on individual signs, which limits the accuracy and timeliness of therapies, especially for asymptomatic patients in clinical management. Therefore, more effective early diagnosis and lesion-targeted treatments remain a pressing clinical need. Metal-organic frameworks (MOFs) are porous crystalline materials formed by the coordination of inorganic metal ions and organic ligands. Due to their unique high specific surface area, structural flexibility, and functional versatility, MOFs are recognized as highly promising candidates for diagnostic and therapeutic applications in the field of VDs. In this review, the potential of MOFs to act as biosensors, contrast agents, artificial nanozymes, and multifunctional therapeutic agents in the diagnosis and treatment of VDs from the clinical perspective, highlighting the integration between clinical methods with MOFs is generalized. At the same time, multidisciplinary cooperation from chemistry, physics, biology, and medicine to promote the substantial commercial transformation of MOFs in tackling VDs is called for.
Collapse
Affiliation(s)
- Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, P. R. China
| | - Ying Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, P. R. China
| | - Yuanchao Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, P. R. China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin, 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin, 300072, China
| |
Collapse
|
7
|
Nam NN, Tran NKS, Nguyen TT, Trai NN, Thuy NP, Do HDK, Tran NHT, Trinh KTL. Classification and application of metal-based nanoantioxidants in medicine and healthcare. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:396-415. [PMID: 38633767 PMCID: PMC11022389 DOI: 10.3762/bjnano.15.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
Antioxidants play an important role in the prevention of oxidative stress and have been widely used in medicine and healthcare. However, natural antioxidants have several limitations such as low stability, difficult long-term storage, and high cost of large-scale production. Along with significant advances in nanotechnology, nanomaterials have emerged as a promising solution to improve the limitations of natural antioxidants because of their high stability, easy storage, time effectiveness, and low cost. Among various types of nanomaterials exhibiting antioxidant activity, metal-based nanoantioxidants show excellent reactivity because of the presence of an unpaired electron in their atomic structure. In this review, we summarize some novel metal-based nanoantioxidants and classify them into two main categories, namely chain-breaking and preventive antioxidant nanomaterials. In addition, the applications of antioxidant nanomaterials in medicine and healthcare are also discussed. This review provides a deeper understanding of the mechanisms of metal-based nanoantioxidants and a guideline for using these nanomaterials in medicine and healthcare.
Collapse
Affiliation(s)
- Nguyen Nhat Nam
- Applied Biology Center, School of Agriculture and Aquaculture, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Nguyen Khoi Song Tran
- College of Korean Medicine, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| | - Tan Tai Nguyen
- Department of Materials Science, School of Applied Chemistry, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Nguyen Ngoc Trai
- Applied Biology Center, School of Agriculture and Aquaculture, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Nguyen Phuong Thuy
- Applied Biology Center, School of Agriculture and Aquaculture, Tra Vinh University, Tra Vinh City 87000, Vietnam
| | - Hoang Dang Khoa Do
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ward 13, District 04, Ho Chi Minh City 70000, Vietnam
| | - Nhu Hoa Thi Tran
- Faculty of Materials Science and Technology, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University Ho Chi Minh City, Vietnam
| | - Kieu The Loan Trinh
- BioNano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
| |
Collapse
|
8
|
Sun M, Hu N, Gao Y, Lv N, Fu X, Li Y, Zhai S, Zhang R. Platelet Membrane-Encapsulated Nanocomplexes Based on Profundity Scavenging ROS Strategy for Myocardial Infarction Therapy. Adv Healthc Mater 2024; 13:e2303101. [PMID: 38174837 DOI: 10.1002/adhm.202303101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/09/2023] [Indexed: 01/05/2024]
Abstract
Ischemia-induced myocardial injury has become a serious threat to human health, and its treatment remains a challenge. The occurrence of ischemic events leads to a burst release of reactive oxygen species (ROS), which triggers extensive oxidative damage and leads to dysfunctional autophagy, making it difficult for cells to maintain homeostasis. Antioxidants and modulation of autophagy have thus become promising strategies for the treatment of ischemic myocardial injury. This study proposes an antioxidant-activated autophagy therapeutic regimen based on combining melanin (Mel), an excellent antioxidant with metformin mimetic ploymetformin via electrostatic interactions, to obtain a nanocomplex (Met-Mel). The nanocomplex is finally encapsulated with platelet membranes (PMN) to construct a biomimetic nanoparticle (PMN@Met-Mel) capable of targeting injured myocardium. The prepared PMN@Met-Mel has good Mel loading capacity and optimal biosafety. It exhibits excellent antioxidant activity and autophagy activation, rapidly restoring mitochondrial function. Moreover, RNA sequencing (RNA-seq) analysis reveals that PMN@Met-Mel operates mechanistically by triggering the activation of the autophagy pathway. Subsequent in vivo experiments showcase promising cardioprotective effects of these nanoparticles. These discoveries present a newly devised nanoplatform with promising potential for the effective treatment of myocardial infarction.
Collapse
Affiliation(s)
- Meng Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
- Department of Cardiology, First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Nan Hu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Yangyang Gao
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Nan Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaohong Fu
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, 030001, China
| | - Yafeng Li
- The Nephrology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Shaodong Zhai
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
9
|
Yan Z, Zhang Y, Du L, Liu L, Zhou H, Song W. U(VI) exposure induces apoptosis and pyroptosis in RAW264.7 cells. CHEMOSPHERE 2023; 342:140154. [PMID: 37714482 DOI: 10.1016/j.chemosphere.2023.140154] [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: 07/25/2023] [Revised: 08/28/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
U(VI) pollution has already led to serious harm to the environment and human health with the increase of human activities. The viability of RAW264.7 cells was assessed under various U(VI) concentration stress for 24 and 48 h. The reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and superoxide dismutase (SOD) activities of RAW264.7 cells under U(VI) stress were measured. The results showed that U(VI) decreased cell activity, induced intracellular ROS production, abnormal MMP, and increased SOD activity. The flow cytometry with Annexin-V/PI double labeling demonstrated that the rate of late apoptosis increased with the increase of U(VI) concentration, resulting in decreased Bcl-2 expression and increased Bax expression. The morphology of RAW264.7 cells dramatically changed after 48 h U(VI) exposure, including the evident bubble phenomenon. Besides, U(VI) also increased the proportion of LDH releases and increased GSDMD, and Ras, p38, JNK, and ERK1/2 protein expression, which indicated that the MAPK pathway was also involved. Therefore, U(VI) ultimately led to apoptosis and pyroptosis in RAW264.7 cells. This study offered convincing proof of U(VI) immunotoxicity and established the theoretical framework for further fundamental studies on U(VI) toxicity.
Collapse
Affiliation(s)
- Zhuna Yan
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, PR China; Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Yan Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, PR China; Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Liang Du
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Lei Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China
| | - Han Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China.
| | - Wencheng Song
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, PR China; Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health & Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei 230031, PR China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions and School for Radiological and Interdisciplinary Sciences, Soochow University, 215123, Suzhou, PR China.
| |
Collapse
|
10
|
Guo J, Wang H, Li Y, Zhu S, Hu H, Gu Z. Nanotechnology in coronary heart disease. Acta Biomater 2023; 171:37-67. [PMID: 37714246 DOI: 10.1016/j.actbio.2023.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Coronary heart disease (CHD) is one of the major causes of death and disability worldwide, especially in low- and middle-income countries and among older populations. Conventional diagnostic and therapeutic approaches have limitations such as low sensitivity, high cost and side effects. Nanotechnology offers promising alternative strategies for the diagnosis and treatment of CHD by exploiting the unique properties of nanomaterials. In this review, we use bibliometric analysis to identify research hotspots in the application of nanotechnology in CHD and provide a comprehensive overview of the current state of the art. Nanomaterials with enhanced imaging and biosensing capabilities can improve the early detection of CHD through advanced contrast agents and high-resolution imaging techniques. Moreover, nanomaterials can facilitate targeted drug delivery, tissue engineering and modulation of inflammation and oxidative stress, thus addressing multiple aspects of CHD pathophysiology. We discuss the application of nanotechnology in CHD diagnosis (imaging and sensors) and treatment (regulation of macrophages, cardiac repair, anti-oxidative stress), and provide insights into future research directions and clinical translation. This review serves as a valuable resource for researchers and clinicians seeking to harness the potential of nanotechnology in the management of CHD. STATEMENT OF SIGNIFICANCE: Coronary heart disease (CHD) is the one of leading cause of death and disability worldwide. Nanotechnology offers new strategies for diagnosing and treating CHD by exploiting the unique properties of nanomaterials. This review uses bibliometric analysis to uncover research trends in the use of nanotechnology for CHD. We discuss the potential of nanomaterials for early CHD detection through advanced imaging and biosensing, targeted drug delivery, tissue engineering, and modulation of inflammation and oxidative stress. We also offer insights into future research directions and potential clinical applications. This work aims to guide researchers and clinicians in leveraging nanotechnology to improve CHD patient outcomes and quality of life.
Collapse
Affiliation(s)
- Junsong Guo
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hao Wang
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Ying Li
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100190, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Houxiang Hu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Zhanjun Gu
- Academician Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nano-safety, Institute of High Energy Physics, Beijing 100049, China; Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
11
|
Li X, Ou W, Xie M, Yang J, Li Q, Li T. Nanomedicine-Based Therapeutics for Myocardial Ischemic/Reperfusion Injury. Adv Healthc Mater 2023; 12:e2300161. [PMID: 36971662 PMCID: PMC11468948 DOI: 10.1002/adhm.202300161] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/05/2023] [Indexed: 03/29/2023]
Abstract
Myocardial ischemic/reperfusion (IR) injury is a global cardiovascular disease with high mortality and morbidity. Therapeutic interventions for myocardial ischemia involve restoring the occluded coronary artery. However, reactive oxygen species (ROS) inevitably impair the cardiomyocytes during the ischemic and reperfusion phases. Antioxidant therapy holds great promise against myocardial IR injury. The current therapeutic methodologies for ROS scavenging depend predominantly on administering antioxidants. Nevertheless, the intrinsic drawbacks of antioxidants limit their further clinical transformation. The use of nanoplatforms with versatile characteristics greatly benefits drug delivery in myocardial ischemic therapy. Nanoplatform-mediated drug delivery significantly improves drug bioavailability, increases therapeutic index, and reduces systemic toxicity. Nanoplatforms can be specifically and reasonably designed to enhance molecule accumulation at the myocardial site. The present review initially summarizes the mechanism of ROS generation during the process of myocardial ischemia. The understanding of this phenomenon will facilitate the advancement of innovative therapeutic strategies against myocardial IR injury. The latest developments in nanomedicine for treating myocardial ischemic injury are then discussed. Finally, the current challenges and perspectives in antioxidant therapy for myocardial IR injury are addressed.
Collapse
Affiliation(s)
- Xi Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Wei Ou
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
- Department of AnesthesiologyNanchong Central HospitalNanchong637000P. R. China
| | - Maodi Xie
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Jing Yang
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Qian Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Tao Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| |
Collapse
|
12
|
Li D, Zhang G, Wang Z, Guo J, Liu Y, Lu Y, Qin Z, Xu Y, Cao C, Wang B, Guo Q, Wang Y, Liu G, Cui X, Zhang J, Tang J. Idebenone attenuates ferroptosis by inhibiting excessive autophagy via the ROS-AMPK-mTOR pathway to preserve cardiac function after myocardial infarction. Eur J Pharmacol 2023; 943:175569. [PMID: 36740037 DOI: 10.1016/j.ejphar.2023.175569] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of mortality worldwide. As a type of CVDs, myocardial infarction (MI) induces ischemia hypoxia, which leads to excessive reactive oxygen species (ROS), resulting in multiple cell deaths and contributing to the subsequent development of heart failure or premature death. Recent evidence indicates that ROS-induced lipid peroxidation promotes autophagy and ferroptosis, leading to the loss of healthy myocardium and resulting in the dysfunction of cardiac tissue. Theoretically, cardiac function would be preserved after MI by inhibiting autophagy and ferroptosis. As an analog of coenzyme Q10 (CoQ10) and a clinically approved drug, idebenone would be used to inhibit ferroptosis and preserve cardiac function due to its capacity to improve mitochondrial physiology with antioxidant and anti-inflammatory properties. Here, we confirmed that the addition of idebenone inhibited H2O2-induced and RSL3-induced ferroptosis. Furthermore, the ROS-AMPK-mTOR pathway axis was identified as the signaling pathway that idebenone stimulated to prevent excessive autophagy and consequent ferroptosis. In the MI animal model, idebenone demonstrated a cardioprotective role by regulating ROS-dependent autophagy and inhibiting ferroptosis, which paves the way for the future clinical translation of idebenone in MI management.
Collapse
Affiliation(s)
- Demin Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zeyu Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Jiacheng Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yu Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Yongzheng Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yanyan Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Chang Cao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Qianqian Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yunzhe Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Guozhen Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China.
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China.
| |
Collapse
|
13
|
Cold Atmospheric Plasma Inhibits the Proliferation of CAL-62 Cells through the ROS-Mediated PI3K/Akt/mTOR Signaling Pathway. SCIENCE AND TECHNOLOGY OF NUCLEAR INSTALLATIONS 2022. [DOI: 10.1155/2022/3884695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study aimed to investigate the inhibitory effects of cold atmospheric plasma (CAP) on anaplastic thyroid cancer cells (CAL-62 cells) and to reveal the molecular mechanism. The effects of CAP on CAL-62 cells were evaluated by cell viability, superoxide dismutase activity, apoptosis, cell cycle, and protein expression level, and the role of reactive oxygen species (ROS) produced by plasma was also investigated. The results showed that CAP dose-dependently inhibited cell viability and promotes cell apoptosis and G2/M arrest by increasing cell ROS levels. The activity of superoxide dismutase (SOD) was enhanced by CAP which indicated that the antioxidant system of the cell was activated. Additionally, the ROS produced by CAP can inhibit CAL-62 cell proliferation by inhibiting the PI3K/Akt/mTOR signaling pathway. Therefore, these findings will provide useful support for the application of CAP for treating anaplastic thyroid cancer.
Collapse
|