1
|
Yao Q, Wen J, Chen S, Wang Y, Wen X, Wang X, Li C, Zheng C, Li J, Ma Z, Zhan X, Xiao X, Bai Z. Shuangdan Jiedu Decoction improved LPS-induced acute lung injury by regulating both cGAS-STING pathway and inflammasome. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118661. [PMID: 39159837 DOI: 10.1016/j.jep.2024.118661] [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: 04/18/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shuangdan Jiedu Decoction (SJD) is a formula composed of six Chinese herbs with heat-removing and detoxifying, antibacterial, and anti-inflammatory effects, which is clinically used in the therapy of various inflammatory diseases of the lungs including COVID-19, but the therapeutic material basis of its action as well as its molecular mechanism are still unclear. AIM OF THE STUDY The study attempted to determine the therapeutic effect of SJD on LPS-induced acute lung injury (ALI), as well as to investigate its mechanism of action and assess its therapeutic potential for the cure of inflammation-related diseases in the clinical setting. MATERIALS AND METHODS We established an ALI model by tracheal drip LPS, and after the administration of SJD, we collected the bronchoalveolar lavage fluid (BALF) and lung tissues of mice and examined the expression of inflammatory factors in them. In addition, we evaluated the effects of SJD on the cyclic guanosine monophosphate-adenosine monophosphate synthase -stimulator of interferon genes (cGAS-STING) and inflammasome by immunoblotting and real-time quantitative polymerase chain reaction (RT-qPCR). RESULTS We demonstrated that SJD was effective in alleviating LPS-induced ALI by suppressing the levels of pro-inflammatory cytokines in the BALF, improving the level of lung histopathology and the number of neutrophils, as well as decreasing the inflammatory factor-associated gene expression. Importantly, we found that SJD could inhibit multiple stimulus-driven activation of cGAS-STING and inflammasome. Further studies showed that the Chinese herbal medicines in SJD had no influence on the cGAS-STING pathway and inflammasome alone at the formulated dose. By increasing the concentration of these herbs, we observed inhibitory effects on the cGAS-STING pathway and inflammasome, and the effect exerted was maximal when the six herbs were combined, indicating that the synergistic effects among these herbs plays a crucial role in the anti-inflammatory effects of SJD. CONCLUSIONS Our research demonstrated that SJD has a favorable protective effect against ALI, and its mechanism of effect may be associated with the synergistic effect exerted between six Chinese medicines to inhibit the cGAS-STING and inflammasome abnormal activation. These results are favorable for the wide application of SJD in the clinic as well as for the development of drugs for ALI from herbal formulas.
Collapse
Affiliation(s)
- Qing Yao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Jincai Wen
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Simin Chen
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Yan Wang
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Xinru Wen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Xianling Wang
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Chengwei Li
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Congyang Zheng
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Junjie Li
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China
| | - Zhijie Ma
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 100050, Beijing, PR China
| | - Xiaoyan Zhan
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China; National Key Laboratory of Kidney Diseases, Beijing 100005, PR China.
| | - Xiaohe Xiao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China; National Key Laboratory of Kidney Diseases, Beijing 100005, PR China.
| | - Zhaofang Bai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, PR China; Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, PR China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing, 100700, PR China; National Key Laboratory of Kidney Diseases, Beijing 100005, PR China.
| |
Collapse
|
2
|
Wang S, Wang T, Wang Z, Liu G, Ji R, Zang Y, Lin S, Lu J, Zhou H, Wang Q. An integrated bipolar electrode with shared cathode for dual-mode detection and imaging of CEA. Biosens Bioelectron 2024; 265:116704. [PMID: 39182411 DOI: 10.1016/j.bios.2024.116704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
In this paper, we designed a novel shared cathode bipolar electrode chip based on Ohm 's law and successfully constructed a dual-mode dual-signal biosensor platform (DD-cBPE). The device integrates ELISA, ECL, and ECL imaging to achieve highly sensitive detection and visual imaging of carcinoembryonic antigen (CEA). The unique circuit structure of the device not only realizes the dual signal detection of the target, but also breaks the traditional signal amplification concept. The total resistance of the system is reduced by series-parallel connection of BPE, and the total current in the circuit is increased. In addition, Au@NiCo2O4@MnO2 nanozyme activity probe was introduced into the common cathode to enhance the conductivity of the material. At the same time, due to the excellent peroxidase (POD) activity of NiCo2O4@MnO2, the decomposition of H2O2 was accelerated, so that more electrons flowed to the BPE anode, and finally the dual amplification of the ECL signal was realized. The device affects the current in the circuit by regulating the concentration of the co-reactant TPrA, thereby affecting the resistance of the system. Finally, different luminescent reagents emit light at the same potential and the luminous efficiency is similar. In addition, the chip does not need external resistance regulation, which improves the sensitivity of the immunosensor and meets the needs of timely detection. It provides a new idea for the deviceization of bipolar electrodes and has broad application prospects in biosensors, clinical detection, and environmental monitoring.
Collapse
Affiliation(s)
- Shumin Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China; Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, Shandong, 250012, China; Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Tengkai Wang
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China; Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, Shandong, 250012, China
| | - Zehua Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), 758 Hefei Road, Qingdao, Shandong, 266035, China
| | - Gengjun Liu
- Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, Shandong, 250012, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Rui Ji
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Yufei Zang
- Cheeloo College of Medicine, Shandong University, No. 44 Wenhua West Road, Jinan, Shandong, 250012, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Shengxiang Lin
- CHU de Québec Research Center and Department of Molecular Medicine, Laval University, Québec, QC, Canada
| | - Jiaoyang Lu
- Department of Gastroenterology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China
| | - Hong Zhou
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), 758 Hefei Road, Qingdao, Shandong, 266035, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Jin Y, Hu C, Xia J, Xie D, Ye L, Ye X, Jiang L, Song H, Zhu Y, Jiang S, Li W, Qi W, Yang Y, Hu Z. Bimetallic clusterzymes-loaded dendritic mesoporous silica particle regulate arthritis microenvironment via ROS scavenging and YAP1 stabilization. Bioact Mater 2024; 42:613-627. [PMID: 39314862 PMCID: PMC11417149 DOI: 10.1016/j.bioactmat.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Clusterzymes are synthetic enzymes exhibiting substantial catalytic activity and selectivity, which are uniquely driven by single-atom constructs. A dramatic increase in antioxidant capacity, 158 times more than natural trolox, is noted when single-atom copper is incorporated into gold-based clusterzymes to form Au24Cu1. Considering the inflammatory and mildly acidic microenvironment characteristic of osteoarthritis (OA), pH-dependent dendritic mesoporous silica nanoparticles (DMSNs) coupled with PEG have been employed as a delivery system for the spatial-temporal release of clusterzymes within active articular regions, thereby enhancing the duration of effectiveness. Nonetheless, achieving high therapeutic efficacy remains a significant challenge. Herein, we describe the construction of a Clusterzymes-DMSNs-PEG complex (CDP) which remarkably diminishes reactive oxygen species (ROS) and stabilizes the chondroprotective protein YAP by inhibiting the Hippo pathway. In the rabbit ACLT (anterior cruciate ligament transection) model, the CDP complex demonstrated inhibition of matrix metalloproteinase activity, preservation of type II collagen and aggregation protein secretion, thus prolonging the clusterzymes' protective influence on joint cartilage structure. Our research underscores the efficacy of the CDP complex in ROS-scavenging, enabled by the release of clusterzymes in response to an inflammatory and slightly acidic environment, leading to the obstruction of the Hippo pathway and downstream NF-κB signaling pathway. This study illuminates the design, composition, and use of DMSNs and clusterzymes in biomedicine, thus charting a promising course for the development of novel therapeutic strategies in alleviating OA.
Collapse
Affiliation(s)
- Yang Jin
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Chuan Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Jiechao Xia
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Dingqi Xie
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Xinyi Ye
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Honghai Song
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Yutao Zhu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Sicheng Jiang
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Weiqing Li
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiming Qi
- Zhejiang Center for Medical Device Evaluation, Zhejiang Medical Products Administration Hangzhou 310009, Zhejiang, China
| | - Yannan Yang
- Institute of Optoelectronics, Fudan University, Shanghai, 200433, China
- South Australian ImmunoGENomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Zhijun Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| |
Collapse
|
4
|
Wang Q, Nie X, Song Y, Qiu H, Chen L, Zhu H, Zhang X, Yang M, Xu X, Chen P, Zhang C, Xu J, Ren Y, Shang W. Redox nanodrugs alleviate chronic kidney disease by reducing inflammation and regulating ROS. Biomater Sci 2024. [PMID: 39526526 DOI: 10.1039/d4bm00881b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Immune-mediated glomerular diseases lead to chronic kidney disease (CKD), primarily through mechanisms such as immune cell overactivation, mitochondrial dysfunction and imbalance of reactive oxygen species (ROS). We have developed an ultra-small nanodrug composed of Mn3O4 nanoparticles which is functionalized with biocompatible ligand citrate (C-Mn3O4 NPs) to maintain cellular redox balance in an animal model of oxidative injury. Furthermore, this ultra-small nanodrug, loaded with tacrolimus (Tac), regulated the activity of immune cells. We established a doxorubicin (DOX)-induced CKD model in SD rats using conditions of oxidative distress. The results demonstrate the ROS scavenging capability of Mn3O4 NPs, which mimics enzymatic activity, and the immunosuppressive effect of tacrolimus. This combination promotes targeted accumulation in the renal region with sustained drug release through the enhanced permeability and retention (EPR) effect. Tac@C-Mn3O4 protects the structural and functional integrity of mitochondria from oxidative damage while eliminating excess ROS to maintain cellular redox homeostasis, thereby suppressing the overexpression of pro-inflammatory cytokines to restore kidney function and preserve a normal kidney structure, reducing inflammation and regulating antioxidant stress pathways. This dual-pronged treatment strategy also provides novel strategies for CKD management and demonstrates substantial potential for clinical translational application.
Collapse
Affiliation(s)
- Qin Wang
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Xuedan Nie
- Department of Neurology, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong, 518116, China
| | - Yifan Song
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Haiyan Qiu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Liting Chen
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - He Zhu
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Xueli Zhang
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Mengru Yang
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Xiaohui Xu
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Peidan Chen
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Chao Zhang
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Jia Xu
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Yeping Ren
- Department of Nephrology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| | - Wenting Shang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
5
|
Leng T, Zhang L, Ma J, Qu X, Lei B. Intrinsically bioactive multifunctional Poly(citrate-curcumin) for rapid lung injury and MRSA infection therapy. Bioact Mater 2024; 41:158-173. [PMID: 39131630 PMCID: PMC11314446 DOI: 10.1016/j.bioactmat.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
Dysregulated inflammation after trauma or infection could result in the further disease and delayed tissue reconstruction. The conventional anti-inflammatory drug treatment suffers to the poor bioavailability and side effects. Herein, we developed an amphiphilic multifunctional poly (citrate-polyglycol-curcumin) (PCGC) nano oligomer with the robust anti-inflammatory activity for treating acute lung injury (ALI) and Methicillin-resistant staphylococcus aureus (MRSA) infected wound. PCGC demonstrated the sustained curcumin release, inherent photoluminescence, good cellular compatibility, hemocompatibility, robust antioxidant activity and enhanced cellular uptake. PCGC could efficiently scavenge nitrogen-based free radicals, oxygen-based free radicals, and intracellular oxygen species, enhance the endothelial cell migration and reduce the expression of pro-inflammatory factors through the NF-κB signal pathway. Combined the anti-inflammation and antioxidant properties, PCGC can shortened the inflammatory process. In animal model of ALI, PCGC was able to reduce the pulmonary edema, bronchial cell infiltration, and lung inflammation, while exhibiting rapid metabolic behavior in vivo. The MRSA-infection wound model showed that PCGC significantly reduced the expression of pro-inflammatory factors, promoted the angiogenesis and accelerated the wound healing. The transcriptome sequencing and molecular mechanism studies further demonstrated that PCGC could inhibit multiple inflammatory related pathways including TNFAIP3, IL-15RA, NF-κB. This work demonstrates that PCGC is efficient in resolving inflammation and promotes the prospect of application in inflammatory diseases as the drug-loaded therapeutic system.
Collapse
Affiliation(s)
- Tongtong Leng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Long Zhang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Junping Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Xiaoyan Qu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710054, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
6
|
Zhang R, Liu Y, Gao Y, Peng D, Luan Q, Li Z, Xia X, Xiang X. Flavonoid-rich sesame leaf extract-mediated synthesis of nanozymes: Extraction optimization, chemical composition identification and bioactivity evaluation. Food Chem 2024; 456:140021. [PMID: 38870817 DOI: 10.1016/j.foodchem.2024.140021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Sesame leaves contain rich phenolic acids and flavonoids. However, their potential in nanozyme synthesis has not been investigated yet. Herein, we report the preparation of flavonoid-rich sesame leaf extract (SLE), composition identification, and its use in the construction of iron (Fe)-based nanozymes (Fe-SLE CPNs). SLE was obtained with an extraction yield of ∼14.5% with a total flavonoid content (TFC) of ∼850.85 mg RE/g. There were 83 flavonoid compounds in SLE, primarily including scutellarin, apigenin-7-glucuronid, narcissin, and hyperoside. Fe-SLE CPNs exhibited nanodot morphology with a hydrodynamic size of 79.34 nm and good stability in various physiological solutions, pH levels, and temperatures. The Fe-SLE CPNs were more efficient in the scavenging ability of reactive oxygen species (ROS) than SLE alone. Furthermore, a stronger anti-inflammatory effect of the Fe-SLE CPNs was shown by modulating the MyD88-NF-κB-MAPK signaling pathways. These findings imply that SLE-based nanozymes hold great potential for diverse applications.
Collapse
Affiliation(s)
- Ruiying Zhang
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yufei Liu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Yiqiao Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Dengfeng Peng
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China
| | - Qian Luan
- Ningbo Innovation Center, Zhejiang University, Ningbo 315100, China
| | - Ziliang Li
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Xiaoyang Xia
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China.
| | - Xia Xiang
- Oil Crops Research Institute of Chinese Academy of Agricultural Sciences, Oil Crops and Lipids Process Technology National & Local Joint Engineering Laboratory, Key Laboratory of Oilseeds Processing, Ministry of Agriculture and Rural Affairs, Hubei Key Laboratory of Lipid Chemistry and Nutrition, Wuhan 430062, China.
| |
Collapse
|
7
|
Xiao Y, Tang Z, Zhang J, Saiding Q, Li Y, Du J, Tao W. One-Pot Synthesis of Fe-Norepinephrine Nanoparticles for Synergetic Thermal-Enhanced Chemodynamic Therapy. NANO LETTERS 2024; 24:13825-13833. [PMID: 39392201 DOI: 10.1021/acs.nanolett.4c04375] [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: 10/12/2024]
Abstract
Chemodynamic therapy (CDT) is an innovative and burgeoning strategy that utilizes Fenton-Fenton-like chemistry and specific microenvironments to produce highly toxic hydroxyl radicals (•OH), with numerous methods emerging to refine this approach. Herein, we report a coordination compound, Fe-norepinephrine nanoparticles (Fe-NE NPs), via a one-pot synthesis. The Fe-NE NPs are based on ferrous ions (Fe2+) and norepinephrine, which are capable of efficient Fe2+/Fe3+ delivery. Once internalized by tumor cells, the released Fe2+/Fe3+ exerts the Fenton reaction to specifically produce toxic •OH. Moreover, the internal photothermal conversion ability of Fe-NE NPs allows us to simultaneously introduce light to trigger local heat generation and then largely improve the Fenton reaction efficiency, which enables a synergetic photothermal and chemodynamic therapy to realize satisfactory in vivo antitumor efficiency. This proof-of-concept work offers a promising approach to developing nanomaterials and refining strategies for enhanced CDT against tumors.
Collapse
Affiliation(s)
- Yufen Xiao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - Zhongmin Tang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jiamin Zhang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Ou X, Wang Z, Yu D, Guo W, Zvyagin AV, Lin Q, Qu W. VEGF-loaded ROS-responsive nanodots improve the structure and function of sciatic nerve lesions in type II diabetic peripheral neuropathy. Biomaterials 2024; 315:122906. [PMID: 39488031 DOI: 10.1016/j.biomaterials.2024.122906] [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: 05/31/2024] [Revised: 09/27/2024] [Accepted: 10/20/2024] [Indexed: 11/04/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is a prevalent complication of diabetes mellitus (DM), significantly contributing to the risk of amputation and mortality. Reactive oxygen species (ROS) can induce both neurological and structural harm through direct impact and pyroptosis, underscoring the critical role of ROS regulation in mitigating DPN. In this research endeavor, we propose harnessing the inherent antioxidant properties of sulfhydryl groups by grafting them onto gold nanodots through an amidation reaction, resulting in the creation of ROS-responsive AuNDs. Additionally, we aim to synthesize AuNDs-VEGF, wherein VEGF is attached to AuNDs via electrostatic interactions, as a therapeutic strategy for addressing DPN in rat models. The results of in vivo experiments showed that AuNDs and AuNDs-VEGF nanoparticles could increase the nerve conduction velocity, shorten the latency of nerve conduction in the sciatic nerve, promote the regeneration of nerve trophectodermal vessels, improve the structure and function of the sciatic nerve, reduce the apoptosis of neural cells, and alleviate the atrophy of the gastrocnemius muscle. Thus, VEGF-loaded ROS-responsive nanodots present a promising avenue for ameliorating diabetic peripheral neuropathy. This innovative approach not only extends the application possibilities of nanodots but also introduces a novel avenue for the treatment of diabetic neuropathy.
Collapse
Affiliation(s)
- Xiaolan Ou
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China; Department of Plastic and Burn Surgery, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130000, China
| | - Daojiang Yu
- Department of Plastic and Burn Surgery, the Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Wenlai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Bio-photonics, Macquarie University, Sydney, NSW, 2109, Australia; Institute of Biology and Biomedicine, Lobachevsky Nizhny Novgorod State University, 603105. Nizhny Novgorod, Russia
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, 130000, China.
| | - Wenrui Qu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China; Joint International Research Laboratory of Ageing Active Strategy and Bionic Health in Northeast Asia of Ministry of Education, Changchun, 130041, China.
| |
Collapse
|
9
|
Zhang L, Chen S, Zheng Z, Lin Y, Wang C, Gong Y, Qin A, Su J, Tang S. Artificial Neutrophil-Mediated CEBPA-saRNA Delivery to Ameliorate ALI/ARDS. ACS APPLIED MATERIALS & INTERFACES 2024; 16:51957-51969. [PMID: 39305228 DOI: 10.1021/acsami.4c09022] [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: 10/04/2024]
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) still faces great challenges due to uncontrollable inflammation disorders, complicated causes of occurrence, and high mortality. Small-activating RNA (saRNA) has emerged as a novel and powerful gene-activating tool that may be useful in the treatment of ALI/ARDS. However, effective saRNA therapy is still challenged by the lack of effective and safe gene delivery vehicles. In this study, we develop a type of artificial neutrophil that is used to deliver saRNAs for ALI/ARDS treatment. The saRNA targeting CCAAT-enhancer binding protein α (CEBPA-saRNA) is complexed with H1 histone and further camouflaged with neutrophil membranes (NHR). Interestingly, we are the first to find that the H1 histone possesses the most effective binding capability to saRNA, compared to other subtypes. The prepared NHR shows excellent physicochemical properties, effective cellular uptake by the inflammatory M1 macrophages, and efficient activation of CEBPA, leading to significant M2 polarization. NHR shows an extended circulation lifetime and high-level accumulation in the inflamed lungs. The in vivo experiments indicate that NHR ameliorates ALI in a mouse model. This type of artificial neutrophil shows powerful inflammatory inhibition both in vitro and in vivo, which opens a new avenue for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Lingmin Zhang
- Department of Biomedical Engineering, Jinan University, No. 601 Huangpu Road, Guangzhou 510632, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Sheng Chen
- Department of Biomedical Engineering, Jinan University, No. 601 Huangpu Road, Guangzhou 510632, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - ZhouYikang Zheng
- Department of Biomedical Engineering, Jinan University, No. 601 Huangpu Road, Guangzhou 510632, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Yinshan Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chen Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Yingjie Gong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Aiping Qin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Jianfen Su
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Panyu Central Hospital and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Shunqing Tang
- Department of Biomedical Engineering, Jinan University, No. 601 Huangpu Road, Guangzhou 510632, China
| |
Collapse
|
10
|
He C, Li R, Zhang J, Chai W. Sivelestat protects against acute lung injury by up-regulating angiotensin-converting enzyme 2/angiotensin-(1-7)/Mas receptors. J Thorac Dis 2024; 16:6182-6195. [PMID: 39444885 PMCID: PMC11494560 DOI: 10.21037/jtd-24-1281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Background Acute lung injury (ALI) and its most severe manifestation of acute respiratory distress syndrome (ARDS) is a disease with a clinical mortality rate of up to 40% and is one of the most dangerous and common complications of severe coronavirus disease 2019 (COVID-19). Sivelestat (SIV) is the only licensed therapeutic medicine in the world for ALI/ARDS treatment. The angiotensin-converting enzyme 2 (ACE2)/angiotensin (Ang)-(1-7)/Mas receptor axis is critical in the prevention of ALI/ARDS. This study aims to investigate whether SIV alleviates lipopolysaccharides (LPS)-induced ALI by inhibiting the down-regulation of ACE2/Ang-(1-7)/Mas receptor axis expression. Methods In vivo, 90 male Sprague-Dawley rats were randomized into 5 groups. Then, we pretreated different groups of rats with dexamethasone (DEX) or SIV. Rats were sacrificed at three different time points (3, 6, and 12 hours) following LPS instillation. In vitro, RAW264.7 cells were divided into 11 groups. Different groups of cells were pretreated with DEX or SIV. And then added with LPS for 3, 6, and 12 hours. Next, we introduced A779, a potent Ang-(1-7) receptor antagonist, and DX600 as the ACE2 antagonist in different groups. Then the protein and messenger RNA (mRNA) expression levels of ACE2 in rat lung tissue and the expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and Ang-(1-7) in the rat serum and the cell culture supernatant were measured. And the data were statistically analyzed. Results In vivo, the rats pretreated with SIV or DEX had significantly lower lung wet/dry (W/D) ratios and lung pathological alterations than those exposed to LPS only. Both in vivo and in vitro, we observed that SIV or DEX significantly attenuated the LPS-induced up-regulation of IL-6 and TNF-α levels, and the down-regulation of ACE2 and Ang-(1-7) levels. In vitro, the pretreatment of the RAW264.7 cells with DX600 and A779 significantly reduced and even abolished the protective effects of SIV. Conclusions Therefore, it was concluded that SIV protected against LPS-induced ALI and decreased inflammatory cytokine release by up-regulating the ACE2/Ang-(1-7)/Mas receptor axis. Our results enrich the theoretical foundation for the clinical application of SIV and provide fresh ideas for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Changqing He
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ruoxin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jia Zhang
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Wenshu Chai
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
11
|
Yang X, Li J, Ma Y, Dong X, Qu J, Liang F, Liu J. Curcumin-mediated enhancement of lung barrier function in rats with high-altitude-associated acute lung injury via inhibition of inflammatory response. Respir Res 2024; 25:354. [PMID: 39342264 PMCID: PMC11439224 DOI: 10.1186/s12931-024-02975-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Exposure to a hypobaric hypoxic environment at high altitudes can lead to lung injury. In this study, we aimed to determine whether curcumin (Cur) could improve lung barrier function and protect against high-altitude-associated acute lung injury. METHODS Two hundred healthy rats were randomly divided into standard control, high-altitude control (HC), salidroside (40 mg/kg, positive control), and Cur (200 mg/kg) groups. Each group was further divided into five subgroups. Basic vital signs, lung injury histopathology, routine blood parameters, plasma lactate level, and arterial blood gas indicators were evaluated. Protein and inflammatory factor (tumor necrosis factor α (TNF-α), interleukin [IL]-1β, IL-6, and IL-10) concentrations in bronchoalveolar lavage fluid (BALF) were determined using the bicinchoninic acid method and enzyme-linked immunosorbent assay, respectively. Inflammation-related and lung barrier function-related proteins were analyzed using immunoblotting. RESULTS Cur improved blood routine indicators such as hemoglobin and hematocrit and reduced the BALF protein content and TNF-α, IL-1β, and IL-6 levels compared with those in the HC group. It increased IL-10 levels and reduced pulmonary capillary congestion, alveolar hemorrhage, and the degree of pulmonary interstitial edema. It increased oxygen partial pressure, oxygen saturation, carbonic acid hydrogen radical, and base excess levels, and the expression of zonula occludens 1, occludin, claudin-4, and reduced carbon dioxide partial pressure, plasma lactic acid, and the expression of phospho-nuclear factor kappa. CONCLUSIONS Exposure to a high-altitude environment for 48 h resulted in severe lung injury in rats. Cur improved lung barrier function and alleviated acute lung injury in rats at high altitudes.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
- Graduate School , Xinjiang Medical University, Urumqi, 830000, China
| | - Jiajia Li
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830000, China
| | - Yan Ma
- Department of Anesthesiology, Xinjiang Medical University Affiliated First Hospital, Urumqi, 830054, China
| | - Xiang Dong
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Jinquan Qu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Feixing Liang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China
| | - Jiangwei Liu
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjiang Military Command, Urumqi, 830000, China.
| |
Collapse
|
12
|
Zhu Y, Huang X, Deng Z, Bai T, Gao B, Xu C, Fu J, Zhao Y, Zhang Y, Zhang M, Zhang M, Yang M, Chen L. Orally biomimetic metal-phenolic nanozyme with quadruple safeguards for intestinal homeostasis to ameliorate ulcerative colitis. J Nanobiotechnology 2024; 22:545. [PMID: 39238009 PMCID: PMC11378530 DOI: 10.1186/s12951-024-02802-z] [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: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is defined by persistent inflammatory processes within the gastrointestinal tract of uncertain etiology. Current therapeutic approaches are limited in their ability to address oxidative stress, inflammation, barrier function restoration, and modulation of gut microbiota in a coordinated manner to maintain intestinal homeostasis. RESULTS This study involves the construction of a metal-phenolic nanozyme (Cur-Fe) through a ferric ion-mediated oxidative coupling of curcumin. Cur-Fe nanozyme exhibits superoxide dismutase (SOD)-like and •OH scavenging activities, demonstrating significant anti-inflammatory and anti-oxidant properties for maintaining intracellular redox balance in vitro. Drawing inspiration from Escherichia coli Nissle 1917 (EcN), a biomimetic Cur-Fe nanozyme (CF@EM) is subsequently developed by integrating Cur-Fe into the EcN membrane (EM) to improve the in vivo targeting ability and therapeutic effectiveness of the Cur-Fe nanozyme. When orally administered, CF@EM demonstrates a strong ability to colonize the inflamed colon and restore intestinal redox balance and barrier function in DSS-induced colitis models. Importantly, CF@EM influences the gut microbiome towards a beneficial state by enhancing bacterial diversity and shifting the compositional structure toward an anti-inflammatory phenotype. Furthermore, analysis of intestinal microbial metabolites supports the notion that the therapeutic efficacy of CF@EM is closely associated with bile acid metabolism. CONCLUSION Inspired by gut microbes, we have successfully synthesized a biomimetic Cur-Fe nanozyme with the ability to inhibit inflammation and restore intestinal homeostasis. Collectively, without appreciable systemic toxicity, this work provides an unprecedented opportunity for targeted oral nanomedicine in the treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Xiaoling Huang
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Uygur Autonomous Region, Urumgi, 830001, China
| | - Zhichao Deng
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Ting Bai
- Department of Cardiovascular Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710077, China
| | - Bowen Gao
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Chenxi Xu
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Junlong Fu
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yuanru Zhao
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Mingxin Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Mei Yang
- Department of Thoracic Surgery, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Lina Chen
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China.
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
13
|
Qiu S, Cao L, Xiang D, Wang S, Wang D, Qian Y, Li X, Zhou X. Enhanced osteogenic differentiation in 3D hydrogel scaffold via macrophage mitochondrial transfer. J Nanobiotechnology 2024; 22:540. [PMID: 39237942 PMCID: PMC11375923 DOI: 10.1186/s12951-024-02757-1] [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: 03/22/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
To assess the efficacy of a novel 3D biomimetic hydrogel scaffold with immunomodulatory properties in promoting fracture healing. Immunomodulatory scaffolds were used in cell experiments, osteotomy mice treatment, and single-cell transcriptomic sequencing. In vitro, fluorescence tracing examined macrophage mitochondrial transfer and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs). Scaffold efficacy was assessed through alkaline phosphatase (ALP), Alizarin Red S (ARS) staining, and in vivo experiments. The scaffold demonstrated excellent biocompatibility and antioxidant-immune regulation. Single-cell sequencing revealed a shift in macrophage distribution towards the M2 phenotype. In vitro experiments showed that macrophage mitochondria promoted BMSCs' osteogenic differentiation. In vivo experiments confirmed accelerated fracture healing. The GAD/Ag-pIO scaffold enhances osteogenic differentiation and fracture healing through immunomodulation and promotion of macrophage mitochondrial transfer.
Collapse
Affiliation(s)
- Shui Qiu
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China
| | - Lili Cao
- Department of Medical Oncology, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, China
| | - Dingding Xiang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Shu Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Di Wang
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Yiyi Qian
- School of Mechanical Engineering and Automation, Foshan Graduate School of Innovation, Northeastern University, Shenyang, 110819, China
| | - Xiaohua Li
- Department of Orthopedics, Zhongmeng Hospital, Arong Banner, Hulunbuir City, Inner, Mongolia
| | - Xiaoshu Zhou
- Department of Orthopedics, First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning Province, China.
| |
Collapse
|
14
|
Fang F, Chen X. Carrier-Free Nanodrugs: From Bench to Bedside. ACS NANO 2024; 18:23827-23841. [PMID: 39163559 DOI: 10.1021/acsnano.4c09027] [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: 08/22/2024]
Abstract
Carrier-free nanodrugs with extraordinary active pharmaceutical ingredient (API) loading (even 100%), avoidable carrier-induced toxicity, and simple synthetic procedures are considered as one of the most promising candidates for disease theranostics. Substantial studies and the commercial success of "carrier-free" nanocrystals have demonstrated their strong clinical potential. However, their practical translations remain challenging and are impeded by unpredictable assembly processes, insufficient delivery efficiency, and an unclear in vivo fate. In this Perspective, we systematically outline the contemporary and emerging carrier-free nanodrugs based on diverse APIs, as well as highlight their opportunities and challenges in clinical translation. Looking ahead, further improvements in design and preparation, drug delivery, in vivo efficacy, and safety of carrier-free nanomedicines are essential to facilitate their translation from the bench to bedside.
Collapse
Affiliation(s)
- Fang Fang
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore 138667, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| |
Collapse
|
15
|
Guo S, Zhang J, Zhang Q, Xu S, Liu Y, Ma S, Hu X, Liu Y, Zhang X, Jiang R, Zhang Z, Zhang Z, Zhou Z, Wen L. Polygala tenuifolia willd. Extract alleviates LPS-induced acute lung injury in rats via TLR4/NF-κB pathway and NLRP3 inflammasome suppression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155859. [PMID: 38972239 DOI: 10.1016/j.phymed.2024.155859] [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: 11/06/2023] [Revised: 06/03/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Acute lung injury (ALI) has received considerable attention in the field of critical care as it can lead to high mortality rates. Polygala tenuifolia, a traditional Chinese medicine with strong expectorant properties, can be used to treat pneumonia. Owing to the complexity of its composition, the main active ingredient is not yet known. Thus, there is a need to identify its constituent compounds and mechanism of action in the treatment of ALI using advanced technological means. PURPOSE We investigated the anti-inflammatory mechanism and constituent compounds with regard to the effect of P. tenuifolia Willd. extract (EPT) in lipopolysaccharide (LPS)-induced ALI in vivo and in vitro. METHODS The UHPLC-Q-Exactive Orbitrap MS technology was used to investigate the chemical profile of EPT. Network pharmacology was used to predict the targets and pathways of action of EPT in ALI, and molecular docking was used to validate the binding of polygalacic acid to Toll-like receptor (TLR) 4. The main compounds were determined using LC-MS. A rat model of LPS-induced ALI was established, and THP-1 cells were stimulated with LPS and adenosine triphosphate (ATP) to construct an in vitro model. Pathological changes were observed using hematoxylin and eosin staining, Wright-Giemsa staining, and immunohistochemistry. The expression of inflammatory factors (NE, MPO, Ly-6 G, TNF-α, IL-1β, IL-6, and iNOS) was determined using enzyme-linked immunosorbent assay, real-time fluorescence quantitative polymerase chain reaction, and western blotting. The LPS + ATP-induced inflammation model in THP-1 cells was used to verify the in vivo experimental results. RESULTS Ninety-nine compounds were identified or tentatively deduced from EPT. Using network pharmacology, we found that TLR4/NF-κB may be a relevant pathway for the prevention and treatment of ALI by EPT. Polygalacic acid in EPT may be a potential active ingredient. EPT could alleviate LPS-induced histopathological lung damage and reduce the wet/dry lung weight ratio in the rat model of ALI. Moreover, EPT decreased the white blood cell and neutrophil counts in the bronchoalveolar lavage fluid and decreased the expression of genes and proteins of relevant inflammatory factors (NE, MPO, Ly-6 G, TNF-α, IL-1β, IL-6, and iNOS) in lung tissues. It also increased the expression of endothelial-type nitric oxide synthase expression. Western blotting confirmed that EPT may affect TLR4/NF-κB and NLRP3 signaling pathways in vivo. Similar results were obtained in THP-1 cells. CONCLUSION EPT reduced the release of inflammatory factors by affecting TLR4/NF-κB and NLRP3 signaling pathways, thereby attenuating the inflammatory response of ALI. Polygalacic acid is the likely compounds responsible for these effects.
Collapse
Affiliation(s)
- Shuyun Guo
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Jianguang Zhang
- Qinghai-Tibetan Plateau Ethnic Medicinal Resources Protection and Utilization Key Laboratory of National Ethnic Affairs Commission of the People's Republic of China, Southwest Minzu University, Chengdu 610225, China; Qinzhou Provincial Health School, Qinzhou 535000, China
| | - Qian Zhang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Shuang Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yuezhen Liu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Shangzhi Ma
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Xiaodi Hu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Yanju Liu
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Xiuqiao Zhang
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Ruixue Jiang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China
| | - Zhifeng Zhang
- Qinghai-Tibetan Plateau Ethnic Medicinal Resources Protection and Utilization Key Laboratory of National Ethnic Affairs Commission of the People's Republic of China, Southwest Minzu University, Chengdu 610225, China
| | - Zhihua Zhang
- School of Basic Medicine, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| | - Li Wen
- School of Pharmacy, Hubei University of Traditional Chinese Medicine, Huangjia Lake West Road 16, Wuhan 430065, China.
| |
Collapse
|
16
|
Ashique S, Mishra N, Mantry S, Garg A, Kumar N, Gupta M, Kar SK, Islam A, Mohanto S, Subramaniyan V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03392-1. [PMID: 39196392 DOI: 10.1007/s00210-024-03392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
A significant number of deaths and disabilities worldwide are brought on by inflammatory lung diseases. Many inflammatory lung disorders, including chronic respiratory emphysema, resistant asthma, resistance to steroids, and coronavirus-infected lung infections, have severe variants for which there are no viable treatments; as a result, new treatment alternatives are needed. Here, we emphasize how oxidative imbalance contributes to the emergence of provocative lung problems that are challenging to treat. Endogenic antioxidant systems are not enough to avert free radical-mediated damage due to the induced overproduction of ROS. Pro-inflammatory mediators are then produced due to intracellular signaling events, which can harm the tissue and worsen the inflammatory response. Overproduction of ROS causes oxidative stress, which causes lung damage and various disease conditions. Invasive microorganisms or hazardous substances that are inhaled repeatedly can cause an excessive amount of ROS to be produced. By starting signal transduction pathways, increased ROS generation during inflammation may cause recurrent DNA damage and apoptosis and activate proto-oncogenes. This review provides information about new targets for conducting research in related domains or target factors to prevent, control, or treat such inflammatory oxidative stress-induced inflammatory lung disorders.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, 713212, India.
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, MP, 474005, India
| | - Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Sanjeeb Kumar Kar
- Department of Pharmaceutical Chemistry, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
17
|
Zhao T, Zhou ZR, Wan HQ, Feng T, Hu XH, Li XQ, Zhao SM, Li HL, Hou JW, Li W, Lu DY, Qian MY, Shen X. Otilonium bromide ameliorates pulmonary fibrosis in mice through activating phosphatase PPM1A. Acta Pharmacol Sin 2024:10.1038/s41401-024-01368-8. [PMID: 39160244 DOI: 10.1038/s41401-024-01368-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/24/2024] [Indexed: 08/21/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic, progressive and irreversible interstitial lung disease characterized by unremitting pulmonary myofibroblasts activation, extracellular matrix (ECM) deposition and inflammatory recruitment. PF has no curable medication yet. In this study we investigated the molecular pathogenesis and potential therapeutic targets of PF and discovered drug lead compounds for PF therapy. A murine PF model was established in mice by intratracheal instillation of bleomycin (BLM, 5 mg/kg). We showed that the protein level of pulmonary protein phosphatase magnesium-dependent 1A (PPM1A, also known as PP2Cα) was significantly downregulated in PF patients and BLM-induced PF mice. We demonstrated that TRIM47 promoted ubiquitination and decreased PPM1A protein in PF progression. By screening the lab in-house compound library, we discovered otilonium bromide (OB, clinically used for treating irritable bowel syndrome) as a PPM1A enzymatic activator with an EC50 value of 4.23 μM. Treatment with OB (2.5, 5 mg·kg-1·d-1, i.p., for 20 days) significantly ameliorated PF-like pathology in mice. We constructed PF mice with PPM1A-specific knockdown in the lung tissues, and determined that by targeting PPM1A, OB treatment suppressed ECM deposition through TGF-β/SMAD3 pathway in fibroblasts, repressed inflammatory responses through NF-κB/NLRP3 pathway in alveolar epithelial cells, and blunted the crosstalk between inflammation in alveolar epithelial cells and ECM deposition in fibroblasts. Together, our results demonstrate that pulmonary PPM1A activation is a promising therapeutic strategy for PF and highlighted the potential of OB in the treatment of the disease.
Collapse
Affiliation(s)
- Tong Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi-Ruo Zhou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hui-Qi Wan
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tian Feng
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu-Hui Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Qian Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shi-Mei Zhao
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hong-Lin Li
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ji-Wei Hou
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, China
| | - Wei Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Da-Yun Lu
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Min-Yi Qian
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
18
|
曾 佳, 黄 颂, 杜 方, 曹 素, 高 杨, 邱 逦, 唐 远. [Advances in the Application of Nanozymes in Joint Disease Therapy]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:800-806. [PMID: 39170029 PMCID: PMC11334270 DOI: 10.12182/20240760105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Indexed: 08/23/2024]
Abstract
Nanozymes are nanoscale materials with enzyme-mimicking catalytic properties. Nanozymes can mimic the mechanism of natural enzyme molecules. By means of advanced chemical synthesis technology, the size, shape, and surface characteristics of nanozymes can be accurately regulated, and their catalytic properties can be customized according to the specific need. Nanozymes can mimic the function of natural enzymes, including catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx), to scavenge reactive oxygen species (ROS). Reported findings have shown that nanozymes have the advantages of excellent stability, low cost, and adjustable catalytic activity, thereby showing great potential and broad prospects in the application of disease treatment. Herein, we reviewed the advances in the application of nanozymes in the treatment of joint diseases. The common clinical manifestations of joint diseases include joint pain, swelling, stiffness, and limited mobility. In severe cases, joint diseases may lead to joint destruction, deformity, and functional damage, entailing crippling socioeconomic burdens. ROS is a product of oxidative stress. Increased ROS in the joints can induce macrophage M1 type polarization, which in turn induces and aggravates arthritis. Therefore, the key to the treatment of joint diseases lies in ROS scavenging and increasing oxygen (O2) content. Nanozymes have demonstrated promising application potential in the treatment of joint diseases, including rheumatoid arthritis, osteoarthritis, and gouty arthritis. However, how to ensure their biosafety, reduce the toxicity, and increase enzyme activity remains the main challenge in current research. Precise control of the chemical composition, size, shape, and surface modification of nanomaterials is the main development direction for the future.
Collapse
Affiliation(s)
- 佳 曾
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 颂雅 黄
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 方雪 杜
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 素娇 曹
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 杨 高
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 逦 邱
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| | - 远姣 唐
- 成都市双流区第一人民医院-四川大学华西空港医院 超声医学科 (成都 610200)Department of Ultrasound, The First People's Hospital of Shuangliu District, Chengdu & West China (Airport) Hospital, Sichuan University, Chengdu 610200, China
| |
Collapse
|
19
|
Yin Y, Zeng Z, Wei S, Shen Z, Cong Z, Zhu X. Using the sympathetic system, beta blockers and alpha-2 agonists, to address acute respiratory distress syndrome. Int Immunopharmacol 2024; 139:112670. [PMID: 39018694 DOI: 10.1016/j.intimp.2024.112670] [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: 05/31/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Senhao Wei
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhukai Cong
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China.
| | - Xi Zhu
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
20
|
Yin M, Lei D, Liu Y, Qin T, Gao H, Lv W, Liu Q, Qin L, Jin W, Chen Y, Liang H, Wang B, Gao M, Zhang J, Lu J. NIR triggered polydopamine coated cerium dioxide nanozyme for ameliorating acute lung injury via enhanced ROS scavenging. J Nanobiotechnology 2024; 22:321. [PMID: 38849841 PMCID: PMC11162040 DOI: 10.1186/s12951-024-02570-w] [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: 04/10/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Acute lung injury (ALI) is a life threatening disease in critically ill patients, and characterized by excessive reactive oxygen species (ROS) and inflammatory factors levels in the lung. Multiple evidences suggest that nanozyme with diversified catalytic capabilities plays a vital role in this fatal lung injury. At present, we developed a novel class of polydopamine (PDA) coated cerium dioxide (CeO2) nanozyme (Ce@P) that acts as the potent ROS scavenger for scavenging intracellular ROS and suppressing inflammatory responses against ALI. Herein, we aimed to identify that Ce@P combining with NIR irradiation could further strengthen its ROS scavenging capacity. Specifically, NIR triggered Ce@P exhibited the most potent antioxidant and anti-inflammatory behaviors in lipopolysaccharide (LPS) induced macrophages through decreasing the intracellular ROS levels, down-regulating the levels of TNF-α, IL-1β and IL-6, up-regulating the level of antioxidant cytokine (SOD-2), inducing M2 directional polarization (CD206 up-regulation), and increasing the expression level of HSP70. Besides, we performed intravenous (IV) injection of Ce@P in LPS induced ALI rat model, and found that it significantly accumulated in the lung tissue for 6 h after injection. It was also observed that Ce@P + NIR presented the superior behaviors of decreasing lung inflammation, alleviating diffuse alveolar damage, as well as promoting lung tissue repair. All in all, it has developed the strategy of using Ce@P combining with NIR irradiation for the synergistic enhanced treatment of ALI, which can serve as a promising therapeutic strategy for the clinical treatment of ROS derived diseases as well.
Collapse
Affiliation(s)
- Mingjing Yin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Doudou Lei
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yalan Liu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Tao Qin
- Department of Intensive Care Unit, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Huyang Gao
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Wenquan Lv
- Department of Emergency, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, Guangxi, 530022, China
| | - Qianyue Liu
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Weiqian Jin
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yin Chen
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Hao Liang
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Bailei Wang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China
| | - Ming Gao
- Life Sciences Institute, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jianfeng Zhang
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China.
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530007, China.
| |
Collapse
|
21
|
Ban W, Chen Z, Zhang T, Du T, Huo D, Zhu G, He Z, Sun J, Sun M. Boarding pyroptosis onto nanotechnology for cancer therapy. J Control Release 2024; 370:653-676. [PMID: 38735396 DOI: 10.1016/j.jconrel.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Pyroptosis, a non-apoptotic programmed cellular inflammatory death mechanism characterized by gasdermin (GSDM) family proteins, has gathered significant attention in the cancer treatment. However, the alarming clinical trial data indicates that pyroptosis-mediated cancer therapeutic efficiency is still unsatisfactory. It is essential to integrate the burgeoning biomedical findings and innovations with potent technology to hasten the development of pyroptosis-based antitumor drugs. Considering the rapid development of pyroptosis-driven cancer nanotherapeutics, here we aim to summarize the recent advances in this field at the intersection of pyroptosis and nanotechnology. First, the foundation of pyroptosis-based nanomedicines (NMs) is outlined to illustrate the reliability and effectiveness for the treatment of tumor. Next, the emerging nanotherapeutics designed to induce pyroptosis are overviewed. Moreover, the cross-talk between pyroptosis and other cell death modalities are discussed, aiming to explore the mechanistic level relationships to provide guidance strategies for the combination of different types of antitumor drugs. Last but not least, the opportunities and challenges of employing pyroptosis-based NMs in potential clinical cancer therapy are highlighted.
Collapse
Affiliation(s)
- Weiyue Ban
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhichao Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tengda Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Dianqiu Huo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Guorui Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China.
| |
Collapse
|
22
|
Aghakhani N, Soraya H. Curcumin supplementation as a complementary and alternative medicine for COVID-19 patients. Phytother Res 2024; 38:2724-2727. [PMID: 38520269 DOI: 10.1002/ptr.8194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/23/2024] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Affiliation(s)
- Nader Aghakhani
- Food and Beverages Safety Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
- Experimental and Applied Pharmaceutical Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
23
|
He S, Lin M, Zheng Q, Liang B, He X, Zhang Y, Xu Q, Deng H, Fan K, Chen W. Glucose Oxidase Energized Osmium with Dual-Active Centers and Triple Enzyme Activities for Infected Diabetic Wound Management. Adv Healthc Mater 2024; 13:e2303548. [PMID: 38507709 DOI: 10.1002/adhm.202303548] [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: 10/16/2023] [Revised: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Diabetic wounds are susceptible to bacterial infections, largely linked to high blood glucose levels (hyperglycemia). To treat such wounds, enzymes like glucose oxidase (GOx) can be combined with nanozymes (nanomaterials mimic enzymes) to use glucose effectively for purposes. However, there is still room for improvement in these systems, particularly in terms of process simplification, enzyme activity regulation, and treatment effects. Herein, the approach utilizes GOx to directly facilitate the biomineralized growth of osmium (Os) nanozyme (GOx-OsNCs), leading to dual-active centers and remarkable triple enzyme activities. Initially, GOx-OsNCs use vicinal dual-active centers, enabling a self-cascaded mechanism that significantly enhances glucose sensing performance compared to step-by-step reactions, surpassing the capabilities of other metal sources such as gold and platinum. In addition, GOx-OsNCs are integrated into a glucose-sensing gel, enabling instantaneous visual feedback. In the treatment of infected diabetic wounds, GOx-OsNCs exhibit multifaceted benefits by lowering blood glucose levels and exhibiting antibacterial properties through the generation of hydroxyl free radicals, thereby expediting healing by fostering a favorable microenvironment. Furthermore, the catalase-like activity of GOx-OsNCs aids in reducing oxidative stress, inflammation, and hypoxia, culminating in improved healing outcomes. Overall, this synergistic enzyme-nanozyme blend is user-friendly and holds considerable promise for diverse applications.
Collapse
Affiliation(s)
- Shaobin He
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Mengting Lin
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| | - Qionghua Zheng
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| | - Bo Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xinjie He
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
- School of Clinical Medicine, Fujian Medical University, Fuzhou, 350004, China
| | - Yin Zhang
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Qiuxia Xu
- Laboratory of Clinical Pharmacy, Department of Pharmacy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Haohua Deng
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, 451163, China
| | - Wei Chen
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350004, China
| |
Collapse
|
24
|
Ye J, Fan Y, She Y, Shi J, Yang Y, Yuan X, Li R, Han J, Liu L, Kang Y, Ji X. Biomimetic Self-Propelled Asymmetric Nanomotors for Cascade-Targeted Treatment of Neurological Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310211. [PMID: 38460166 PMCID: PMC11165487 DOI: 10.1002/advs.202310211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/19/2024] [Indexed: 03/11/2024]
Abstract
The precise targeted delivery of therapeutic agents to deep regions of the brain is crucial for the effective treatment of various neurological diseases. However, achieving this goal is challenging due to the presence of the blood‒brain barrier (BBB) and the complex anatomy of the brain. Here, a biomimetic self-propelled nanomotor with cascade targeting capacity is developed for the treatment of neurological inflammatory diseases. The self-propelled nanomotors are designed with biomimetic asymmetric structures with a mesoporous SiO2 head and multiple MnO2 tentacles. Macrophage membrane biomimetic modification endows nanomotors with inflammatory targeting and BBB penetration abilities The MnO2 agents catalyze the degradation of H2O2 into O2, not only by reducing brain inflammation but also by providing the driving force for deep brain penetration. Additionally, the mesoporous SiO2 head is loaded with curcumin, which actively regulates macrophage polarization from the M1 to the M2 phenotype. All in vitro cell, organoid model, and in vivo animal experiments confirmed the effectiveness of the biomimetic self-propelled nanomotors in precise targeting, deep brain penetration, anti-inflammatory, and nervous system function maintenance. Therefore, this study introduces a platform of biomimetic self-propelled nanomotors with inflammation targeting ability and active deep penetration for the treatment of neurological inflammation diseases.
Collapse
Affiliation(s)
- Jiamin Ye
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yaoguang She
- Department of General Surgerythe First Medical CenterChinese People's Liberation Army General HospitalBeijing100853China
| | - Jiacheng Shi
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Yiwen Yang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Xue Yuan
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Ruiyan Li
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Jingwen Han
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Luntao Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjin100730China
| | - Yong Kang
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjin300072China
- Medical CollegeLinyi UniversityLinyi276000China
| |
Collapse
|
25
|
Li X, Lin Z, Xu S, Zhang N, Zhou J, Liao B. Knockdown of KBTBD7 attenuates septic lung injury by inhibiting ferroptosis and improving mitochondrial dysfunction. Int Immunopharmacol 2024; 133:112129. [PMID: 38652964 DOI: 10.1016/j.intimp.2024.112129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Lung injury in sepsis is caused by an excessive inflammatory response caused by the entry of pathogenic microorganisms into the body. It is also accompanied by the production of large amounts of ROS. Ferroptosis and mitochondrial dysfunction have also been shown to be related to sepsis. Finding suitable sepsis therapeutic targets is crucial for sepsis research. BTB domain-containing protein 7 (KBTBD7) is involved in regulating inflammatory responses, but its role and mechanism in the treatment of septic lung injury are still unclear. In this study, we evaluated the role and related mechanisms of KBTBD7 in septic lung injury. In in vitro studies, we established an in vitro model by inducing human alveolar epithelial cells with lipopolysaccharide (LPS) and found that KBTBD7 was highly expressed in the in vitro model. KBTBD7 knockdown could reduce the inflammatory response by inhibiting the secretion of pro-inflammatory factors and inhibit the production of ROS, ferroptosis and mitochondrial dysfunction. Mechanistic studies show that KBTBD7 interacts with FOXA1, promotes FOXA1 expression, and indirectly inhibits SLC7A11 transcription. In vivo studies have shown that knocking down KBTBD7 improves lung tissue damage in septic lung injury mice, inhibits inflammatory factors, ROS production and ferroptosis. Taken together, knockdown of KBTBD7 shows an alleviating effect on septic lung injury in vitro and in vivo, providing a potential therapeutic target for the treatment of septic lung injury.
Collapse
Affiliation(s)
- Xiang Li
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Zhao Lin
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - ShiYu Xu
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Ning Zhang
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Jun Zhou
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Bo Liao
- Department of Critical Care Medicine, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| |
Collapse
|
26
|
Jia Y, Zhang L, Xu J, Xiang L. Recent advances in cell membrane camouflaged nanotherapeutics for the treatment of bacterial infection. Biomed Mater 2024; 19:042006. [PMID: 38697197 DOI: 10.1088/1748-605x/ad46d4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/01/2024] [Indexed: 05/04/2024]
Abstract
Infectious diseases caused by bacterial infections are common in clinical practice. Cell membrane coating nanotechnology represents a pioneering approach for the delivery of therapeutic agents without being cleared by the immune system in the meantime. And the mechanism of infection treatment should be divided into two parts: suppression of pathogenic bacteria and suppression of excessive immune response. The membrane-coated nanoparticles exert anti-bacterial function by neutralizing exotoxins and endotoxins, and some other bacterial proteins. Inflammation, the second procedure of bacterial infection, can also be suppressed through targeting the inflamed site, neutralization of toxins, and the suppression of pro-inflammatory cytokines. And platelet membrane can affect the complement process to suppress inflammation. Membrane-coated nanoparticles treat bacterial infections through the combined action of membranes and nanoparticles, and diagnose by imaging, forming a theranostic system. Several strategies have been discovered to enhance the anti-bacterial/anti-inflammatory capability, such as synthesizing the material through electroporation, pretreating with the corresponding pathogen, membrane hybridization, or incorporating with genetic modification, lipid insertion, and click chemistry. Here we aim to provide a comprehensive overview of the current knowledge regarding the application of membrane-coated nanoparticles in preventing bacterial infections as well as addressing existing uncertainties and misconceptions.
Collapse
Affiliation(s)
- Yinan Jia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Li Zhang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junhua Xu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
27
|
Huang Y, Liu X, Zhu J, Chen Z, Yu L, Huang X, Dong C, Li J, Zhou H, Yang Y, Tan W. Enzyme Core Spherical Nucleic Acid That Enables Enhanced Cuproptosis and Antitumor Immune Response through Alleviating Tumor Hypoxia. J Am Chem Soc 2024; 146:13805-13816. [PMID: 38552185 DOI: 10.1021/jacs.3c14247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Cuproptosis, a copper-dependent cell death process, has been confirmed to further activate the immune response and mediate the immune resistance. However, hypoxic tumor microenvironment hampers cuproptosis sensitivity and suppresses the body's antitumor immune response. Herein, we have successfully immobilized and functionalized catalase (CAT) with long single-stranded DNA containing polyvalent CpG sequences through rolling circle amplification (RCA) techniques, obtaining an enzyme-cored spherical nucleic acid nanoplatform (CAT-ecSNA-Cu) to deliver copper ions for cuproptosis. The presence of long-stranded DNA-protected CAT enhances mitochondrial respiration by catalyzing the conversion of H2O2 to O2, thereby sensitizing cuproptosis. Meanwhile, increased tumor oxygenation suppresses the expression of the hypoxia-inducible factor-1 (HIF-1) protein, resulting in the alleviation of the immunosuppressive tumor microenvironment. Of note, cuproptosis induces immunogenic cell death (ICD), which facilitates dendritic cell (DC) maturation and enhances antigen presentation through polyCpG-supported Toll-like receptor 9 (TLR9) activation. Furthermore, cuproptosis-induced PD-L1 upregulation in tumor cells complements checkpoint blockers (αPD-L1), enhancing antitumor immunity. The strategy of enhancing cuproptosis-mediated antitumor immune responses by alleviating hypoxia effectively promotes the activation and proliferation of effector T cells, ultimately leading to long-term immunity against cancer.
Collapse
Affiliation(s)
- Yuting Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Jiawei Zhu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Zhejie Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Lu Yu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Xin Huang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Chuhuang Dong
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Jiabei Li
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Huayuan Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
28
|
Xu Y, Lv L, Wang Q, Yao Q, Kou L, Zhang H. Emerging application of nanomedicine-based therapy in acute respiratory distress syndrome. Colloids Surf B Biointerfaces 2024; 237:113869. [PMID: 38522285 DOI: 10.1016/j.colsurfb.2024.113869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are serious lung injuries caused by various factors, leading to increased permeability of the alveolar-capillary barrier, reduced stability of the alveoli, inflammatory response, and hypoxemia. Despite several decades of research since ARDS was first formally described in 1967, reliable clinical treatment options are still lacking. Currently, supportive therapy and mechanical ventilation are prioritized, and there is no medication that can be completely effective in clinical treatment. In recent years, nanomedicine has developed rapidly and has exciting preclinical treatment capabilities. Using a drug delivery system based on nanobiotechnology, local drugs can be continuously released in lung tissue at therapeutic levels, reducing the frequency of administration and improving patient compliance. Furthermore, this novel drug delivery system can target specific sites and reduce systemic side effects. Currently, many nanomedicine treatment options for ARDS have demonstrated efficacy. This review briefly introduces the pathophysiology of ARDS, discusses various research progress on using nanomedicine to treat ARDS, and anticipates future developments in related fields.
Collapse
Affiliation(s)
- Yitianhe Xu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Leyao Lv
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Qian Wang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Hailin Zhang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Department of Children's Respiration Disease, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
29
|
Haynes ME, Sullivan DP, Muller WA. Neutrophil Infiltration and Function in the Pathogenesis of Inflammatory Airspace Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:628-636. [PMID: 38309429 PMCID: PMC11074974 DOI: 10.1016/j.ajpath.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 02/05/2024]
Abstract
Neutrophils are an important cell type often considered the body's first responders to inflammatory insult or damage. They are recruited to the tissue of the lungs in patients with inflammatory airspace diseases and have unique and complex functions that range from helpful to harmful. The uniqueness of these functions is due to the heterogeneity of the inflammatory cascade and retention in the vasculature. Neutrophils are known to marginate, or remain stagnant, in the lungs even in nondisease conditions. This review discusses the ways in which the recruitment, presence, and function of neutrophils in the airspace of the lungs are unique from those of other tissues, and the complex effects of neutrophils on pathogenesis. Inflammatory mediators produced by neutrophils, such as neutrophil elastase, proresolving mediators, and neutrophil extracellular traps, dramatically affect the outcomes of patients with disease of the lungs.
Collapse
Affiliation(s)
- Maureen E Haynes
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David P Sullivan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William A Muller
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
30
|
Shan J, Jin X, Zhang C, Huang M, Xing J, Li Q, Cui Y, Niu Q, Chen XL, Wang X. Metal natural product complex Ru-procyanidins with quadruple enzymatic activity combat infections from drug-resistant bacteria. Acta Pharm Sin B 2024; 14:2298-2316. [PMID: 38799629 PMCID: PMC11121202 DOI: 10.1016/j.apsb.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 05/29/2024] Open
Abstract
Bacterial infection hampers wound repair by impeding the healing process. Concurrently, inflammation at the wound site triggers the production of reactive oxygen species (ROS), causing oxidative stress and damage to proteins and cells. This can lead to chronic wounds, posing severe risks. Therefore, eliminating bacterial infection and reducing ROS levels are crucial for effective wound healing. Nanozymes, possessing enzyme-like catalytic activity, can convert endogenous substances into highly toxic substances, such as ROS, to combat bacteria and biofilms without inducing drug resistance. However, the current nanozyme model with single enzyme activity falls short of meeting the complex requirements of antimicrobial therapy. Thus, developing nanozymes with multiple enzymatic activities is essential. Herein, we engineered a novel metalloenzyme called Ru-procyanidin nanoparticles (Ru-PC NPs) with diverse enzymatic activities to aid wound healing and combat bacterial infections. Under acidic conditions, due to their glutathione (GSH) depletion and peroxidase (POD)-like activity, Ru-PC NPs combined with H2O2 exhibit excellent antibacterial effects. However, in a neutral environment, the Ru-PC NPs, with catalase (CAT) activity, decompose H2O2 to O2, alleviating hypoxia and ensuring a sufficient oxygen supply. Furthermore, Ru-PC NPs possess exceptional antioxidant capacity through their superior superoxide dismutase (SOD) enzyme activity, effectively scavenging excess ROS and reactive nitrogen species (RNS) in a neutral environment. This maintains the balance of the antioxidant system and prevents inflammation. Ru-PC NPs also promote the polarization of macrophages from M1 to M2, facilitating wound healing. More importantly, Ru-PC NPs show good biosafety with negligible toxicity. In vivo wound infection models have confirmed the efficacy of Ru-PC NPs in inhibiting bacterial infection and promoting wound healing. The focus of this work highlights the quadruple enzymatic activity of Ru-PC NPs and its potential to reduce inflammation and promote bacteria-infected wound healing.
Collapse
Affiliation(s)
- Jie Shan
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xu Jin
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Cong Zhang
- Division of Gastroenterology, the First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Muchen Huang
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Jianghao Xing
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qingrong Li
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuyu Cui
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Qiang Niu
- The Second Clinical Medical College, Anhui Medical University, Hefei 230022, China
| | - Xu Lin Chen
- Department of Burns, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
- College and Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| |
Collapse
|
31
|
Yang J, Dong L, Wang Y, Gong L, Gao H, Xie Y. Targeted degradation of hexokinase 2 for anti‑inflammatory treatment in acute lung injury. Mol Med Rep 2024; 29:83. [PMID: 38516767 PMCID: PMC10975098 DOI: 10.3892/mmr.2024.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Acute lung injury (ALI) is an acute inflammatory lung disease associated with both innate and adaptive immune responses. Hexokinase 2 (HK2) is specifically highly expressed in numerous types of inflammation‑related diseases and models. In the present study in vitro and in vivo effects of targeted degradation of HK2 on ALI were explored. The degradation of HK2 by the targeting peptide TAT (transactivator of transcription protein of HIV‑1)‑ataxin 1 (ATXN1)‑chaperone‑mediated autophagy‑targeting motif (CTM) was demonstrated by ELISA and western blotting in vitro and in vivo. The inhibitory effects of TAT‑ATXN1‑CTM on lipopolysaccharide (LPS)‑induced inflammatory responses were examined using ELISAs. The therapeutic effects of TAT‑ATXN1‑CTM on LPS‑induced ALI were examined via histological examination and ELISAs in mice. 10 µM TAT‑ATXN1‑CTM administration decreased HK2 protein expression and the secretion of proinflammatory cytokines (TNF‑α and IL‑1β) without altering HK2 mRNA expression in LPS‑treated both in vitro and in vivo, while pathological lung tissue damage and the accumulation of leukocytes, neutrophils, macrophages and lymphocytes in ALI were also significantly suppressed by 10 µM TAT‑ATXN1‑CTM treatment. TAT‑ATXN1‑CTM exhibited anti‑inflammatory activity in vitro and decreased the severity of ALI in vivo. HK2 degradation may represent a novel therapeutic approach for ALI.
Collapse
Affiliation(s)
- Jiayan Yang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Liangliang Dong
- Department of Pulmonology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| | - Yifan Wang
- Department of Pulmonology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Lifen Gong
- Department of Pulmonology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| | - Hongwei Gao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Yicheng Xie
- Department of Pulmonology, Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052, P.R. China
| |
Collapse
|
32
|
Huang X, Zhu J, Dong C, Li Y, Yu Q, Wang X, Chen Z, Li J, Yang Y, Wang H. Polyvalent Aptamer-Functionalized NIR-II Quantum Dots for Targeted Theranostics in High PD-L1-Expressing Tumors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21571-21581. [PMID: 38636085 DOI: 10.1021/acsami.4c01486] [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: 04/20/2024]
Abstract
Ag2S quantum dots (QDs) show superior optical properties in the NIR-II region and display significant clinical potential with favorable biocompatibility. However, inherent defects of low targeting and poor solubility necessitate practical modification methods to achieve the theranostics of Ag2S QDs. Herein, we used rolling circle amplification (RCA) techniques to obtain long single-stranded DNA containing the PD-L1 aptamer and C-rich DNA palindromic sequence. The C-rich DNA palindromic sequences can specifically chelate Ag2+ and thus serve as a template to result in biomimetic mineralization and formation of pApt-Ag2S QDs. These QDs enable specific targeting and illuminate hot tumors with high PD-L1 expression effectively, serving as excellent molecular targeted probes. In addition, due to the high NIR-II absorption of Ag2S QDs, pApt-Ag2S QDs exhibit remarkable photothermal properties. And besides, polyvalent PD-L1 aptamers can recognize PD-L1 protein and effectively block the inhibitory signal of PD-L1 on T cells, enabling efficient theranostics through the synergistic effect of photothermal therapy and immune checkpoint blocking therapy. Summary, we enhance the biological stability and antibleaching ability of Ag2S QDs using long single-stranded DNA as a template, thereby establishing a theranostic platform that specifically targets PD-L1 high-expressing inflamed tumors and demonstrates excellent performance both in vitro and in vivo.
Collapse
Affiliation(s)
- Xin Huang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Jiawei Zhu
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Chuhuang Dong
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yuqing Li
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Qing Yu
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Xuan Wang
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Zhejie Chen
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Jiabei Li
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
33
|
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
|
34
|
Duan Z, Zhou W, He S, Wang W, Huang H, Yi L, Zhang R, Chen J, Zan X, You C, Gao X. Intranasal Delivery of Curcumin Nanoparticles Improves Neuroinflammation and Neurological Deficits in Mice with Intracerebral Hemorrhage. SMALL METHODS 2024:e2400304. [PMID: 38577823 DOI: 10.1002/smtd.202400304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/24/2024] [Indexed: 04/06/2024]
Abstract
Intracerebral hemorrhage (ICH) represents one of the most severe subtypes of stroke. Due to the complexity of the brain injury mechanisms following ICH, there are currently no effective treatments to significantly improve patient functional outcomes. Curcumin, as a potential therapeutic agent for ICH, is limited by its poor water solubility and oral bioavailability. In this study, mPEG-PCL is used to encapsulate curcumin, forming curcumin nanoparticles, and utilized the intranasal administration route to directly deliver curcumin nanoparticles from the nasal cavity to the brain. By inhibiting pro-inflammatory neuroinflammation of microglia following ICH in mice, reprogramming pro-inflammatory microglia toward an anti-inflammatory function, and consequently reducing neuronal inflammatory death and hematoma volume, this approach improved blood-brain barrier damage in ICH mice and promoted the recovery of neurological function post-stroke. This study offers a promising therapeutic strategy for ICH to mediate neuroinflammatory microenvironments.
Collapse
Affiliation(s)
- Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wenjie Zhou
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xin Zan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Chao You
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
35
|
Jia B, Zhang B, Li J, Qin J, Huang Y, Huang M, Ming Y, Jiang J, Chen R, Xiao Y, Du J. Emerging polymeric materials for treatment of oral diseases: design strategy towards a unique oral environment. Chem Soc Rev 2024; 53:3273-3301. [PMID: 38507263 DOI: 10.1039/d3cs01039b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Oral diseases are prevalent but challenging diseases owing to the highly movable and wet, microbial and inflammatory environment. Polymeric materials are regarded as one of the most promising biomaterials due to their good compatibility, facile preparation, and flexible design to obtain multifunctionality. Therefore, a variety of strategies have been employed to develop materials with improved therapeutic efficacy by overcoming physicobiological barriers in oral diseases. In this review, we summarize the design strategies of polymeric biomaterials for the treatment of oral diseases. First, we present the unique oral environment including highly movable and wet, microbial and inflammatory environment, which hinders the effective treatment of oral diseases. Second, a series of strategies for designing polymeric materials towards such a unique oral environment are highlighted. For example, multifunctional polymeric materials are armed with wet-adhesive, antimicrobial, and anti-inflammatory functions through advanced chemistry and nanotechnology to effectively treat oral diseases. These are achieved by designing wet-adhesive polymers modified with hydroxy, amine, quinone, and aldehyde groups to provide strong wet-adhesion through hydrogen and covalent bonding, and electrostatic and hydrophobic interactions, by developing antimicrobial polymers including cationic polymers, antimicrobial peptides, and antibiotic-conjugated polymers, and by synthesizing anti-inflammatory polymers with phenolic hydroxy and cysteine groups that function as immunomodulators and electron donors to reactive oxygen species to reduce inflammation. Third, various delivery systems with strong wet-adhesion and enhanced mucosa and biofilm penetration capabilities, such as nanoparticles, hydrogels, patches, and microneedles, are constructed for delivery of antibiotics, immunomodulators, and antioxidants to achieve therapeutic efficacy. Finally, we provide insights into challenges and future development of polymeric materials for oral diseases with promise for clinical translation.
Collapse
Affiliation(s)
- Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Beibei Zhang
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianhua Li
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jinlong Qin
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yisheng Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Mingshu Huang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Yue Ming
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong, China
| | - Jingjing Jiang
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Ran Chen
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Yufen Xiao
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| |
Collapse
|
36
|
Li B, Liu Y, Chen D, Sun S. Comprehensive Analysis of Predictive Value and the potential therapeutic target of NLRP3 inflammasome in glioma based on tumor microenvironment. Clin Immunol 2024; 261:109918. [PMID: 38307475 DOI: 10.1016/j.clim.2024.109918] [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: 11/16/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Glioma exhibits high recurrence rates and poor prognosis. The nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome plays a crucial role in inflammation. There is a lack of research exploring the NLRP3 in glioma. METHODS We used several databases, networks, Western blotting, multiple immunofluorescence staining to analyze the role of NLRP3 in inflammatory tumor microenvironment (TME). RESULTS NLRP3 is higher-expression in glioma with a low mutation load. NLRP3 expression is linked to the infiltration of immune cells, chemokines, immunomodulators, and the TME. Signaling pathways, co-expression genes and interacting proteins contribute to the up-regulation of NLRP3. Patients responding to immunotherapy positively tend to have lower NLRP3 expression relating to the overall survival based on nomogram. Sensitivity to molecular medicines is observed in relation to NLRP3. CONCLUSION The NLRP3 inflammasome plays a pivotal role in TME which could serve as a higher predictive value biomarker and therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Bihan Li
- Nanjing municipal center for disease control and prevention, Nanjing, Jiangsu, China; Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Dawei Chen
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
37
|
Ai S, Li Y, Zheng H, Zhang M, Tao J, Liu W, Peng L, Wang Z, Wang Y. Collision of herbal medicine and nanotechnology: a bibliometric analysis of herbal nanoparticles from 2004 to 2023. J Nanobiotechnology 2024; 22:140. [PMID: 38556857 PMCID: PMC10983666 DOI: 10.1186/s12951-024-02426-3] [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: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Herbal nanoparticles are made from natural herbs/medicinal plants, their extracts, or a combination with other nanoparticle carriers. Compared to traditional herbs, herbal nanoparticles lead to improved bioavailability, enhanced stability, and reduced toxicity. Previous research indicates that herbal medicine nanomaterials are rapidly advancing and making significant progress; however, bibliometric analysis and knowledge mapping for herbal nanoparticles are currently lacking. We performed a bibliometric analysis by retrieving publications related to herbal nanoparticles from the Web of Science Core Collection (WoSCC) database spanning from 2004 to 2023. Data processing was performed using the R package Bibliometrix, VOSviewers, and CiteSpace. RESULTS In total, 1876 articles related to herbal nanoparticles were identified, originating from various countries, with China being the primary contributing country. The number of publications in this field increases annually. Beijing University of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, and Saveetha University in India are prominent research institutions in this domain. The Journal "International Journal of Nanomedicine" has the highest number of publications. The number of authors of these publications reached 8234, with Yan Zhao, Yue Zhang, and Huihua Qu being the most prolific authors and Yan Zhao being the most frequently cited author. "Traditional Chinese medicine," "drug delivery," and "green synthesis" are the main research focal points. Themes such as "green synthesis," "curcumin," "wound healing," "drug delivery," and "carbon dots" may represent emerging research areas. CONCLUSIONS Our study findings assist in identifying the latest research frontiers and hot topics, providing valuable references for scholars investigating the role of nanotechnology in herbal medicine.
Collapse
Affiliation(s)
- Sinan Ai
- China-Japan Friendship Hospital, Beijing, China
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, China
| | - Yake Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huijuan Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Meiling Zhang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiayin Tao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Beijing, China.
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Medical Science, China-Japan Friendship Hospital, Beijing, China.
| | - Zhen Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Yaoxian Wang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
38
|
Gao H, Yuan Z, Liang H, Liu Y. Integrating UPLC-Q-Orbitrap MS with serum pharmacochemistry network and experimental verification to explore the pharmacological mechanisms of Cynanchi stauntonii rhizoma et radix against sepsis-induced acute lung injury. Front Pharmacol 2024; 15:1261772. [PMID: 38584603 PMCID: PMC10995315 DOI: 10.3389/fphar.2024.1261772] [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: 07/19/2023] [Accepted: 01/03/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction: Patients with sepsis are at an incremental risk of acute lung injury (ALI). Baiqian, also known as Cynanchi stauntonii rhizoma et radix (Csrer), has anti-inflammatory properties and is traditionally used to treat cough and phlegm. This study aimed to demonstrate the multicomponent, multitarget, and multi-pathway regulatory molecular mechanisms of Csrer in treating lipopolysaccharide (LPS)-induced ALI. Methods: The bioactive components of Csrer were identified by ultrahigh-performance liquid chromatography Q-Orbitrap mass spectrometry (UPLC-Q-Orbitrap MS). Active targets predicted from PharmMapper. DrugBank, OMIM, TTD, and GeneCards were used to identify potential targets related to ALI. Intersection genes were identified for Csrer against ALI. The PPI network was analysed to identify prime targets. GO and KEGG analyses were performed. A drug-compound-target-pathway-disease network was constructed. Molecular docking and simulations evaluated the binding free energy between key proteins and active compounds. The protective effect and mechanism of Csrer in ALI were verified using an ALI model in mice. Western blot, Immunohistochemistry and TUNEL staining evaluated the mechanisms of the pulmonary protective effects of Csrer. Results: Forty-six bioactive components, one hundred and ninety-two potential cross-targets against ALI and ten core genes were identified. According to GO and KEGG analyses, the PI3K-Akt, apoptosis and p53 pathways are predominantly involved in the "Csrer-ALI" network. According to molecular docking and dynamics simulations, ten key genes were firmly bound by the principal active components of Csrer. The "Csrer-ALI" network was revealed to be mediated by the p53-mediated apoptosis and inflammatory pathways in animal experiments. Conclusion: Csrer is a reliable source for ALI treatment based on its practical components, potential targets and pathways.
Collapse
Affiliation(s)
- Hejun Gao
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ziyi Yuan
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haoxuan Liang
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Youtan Liu
- Department of Anesthesiology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Shen H, Pan L, Ning K, Fang Y, Muhitdinov B, Liu E, Huang Y. Asiatic acid cyclodextrin inclusion micro-cocrystal for insoluble drug delivery and acute lung injury therapy enhancement. J Nanobiotechnology 2024; 22:119. [PMID: 38494523 PMCID: PMC10946140 DOI: 10.1186/s12951-024-02387-7] [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: 01/15/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a fatal respiratory disease caused by overreactive immune reactions (e.g., SARS-CoV-2 infection), with a high mortality rate. Its treatment is often compromised by inefficient drug delivery barriers and insufficient potency of the currently used drugs. Therefore, developing a highly effective lung-targeted drug delivery strategy is a pressing clinical need. RESULTS In this study, the micro-sized inclusion cocrystal of asiatic acid/γ-cyclodextrin (AA/γCD, with a stoichiometry molar ratio of 2:3 and a mean size of 1.8 μm) was prepared for ALI treatment. The dissolution behavior of the AA/γCD inclusion cocrystals followed a "spring-and-hover" model, which meaned that AA/γCD could dissolve from the cocrystal in an inclusion complex form, thereby promoting a significantly improved water solubility (nine times higher than free AA). This made the cyclodextrin-based inclusion cocrystals an effective solid form for enhanced drug absorption and delivery efficiency. The biodistribution experiments demonstrated AA/γCD accumulated predominantly in the lung (Cmax = 50 µg/g) after systemic administration due to the micron size-mediated passive targeting effect. The AA/γCD group showed an enhanced anti-inflammatory therapeutic effect, as evidenced by reduced levels of pro-inflammatory cytokines in the lung and bronchoalveolar lavage fluids (BALF). Histological examination confirmed that AA/γCD effectively inhibited inflammation reactions. CONCLUSION The micro-sized inclusion cocrystals AA/γCD were successfully delivered into the lungs by pulmonary administration and had a significant therapeutic effect on ALI.
Collapse
Affiliation(s)
- Huan Shen
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Li Pan
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China
| | - Keke Ning
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China
| | - Yuefei Fang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Bahtiyor Muhitdinov
- Institute of Bioorganic Chemistry, Uzbekistan Academy of Sciences, 83 M. Ulughbek Street, Tashkent, 100125, Uzbekistan
| | - Ergang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
- State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, Zunyi Medical University, Zunyi, 563003, China.
- NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai, 201203, China.
| |
Collapse
|
40
|
Chen KY, Liu SY, Tang JJ, Liu MK, Chen XY, Liu ZP, Ferrandon D, Lai KF, Li Z. NLRP3 knockout in mice provided protection against Serratia marcescens-induced acute pneumonia by decreasing PD-L1 and PD-1 expression in macrophages. Int Immunopharmacol 2024; 129:111559. [PMID: 38330794 DOI: 10.1016/j.intimp.2024.111559] [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: 11/12/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024]
Abstract
Antibiotic-resistant Serratia marcescens (Sm) is known to cause bloodstream infections, pneumonia, etc. The nod-like receptor family, pyrin domain-containing 3 (NLRP3), has been implicated in various lung infections. Yet, its role in Sm-induced pneumonia was not well understood. In our study, we discovered that deletion of Nlrp3 in mice significantly improved Sm-induced survival rates, reduced bacterial loads in the lungs, bronchoalveolar lavage fluid (BALF), and bloodstream, and mitigated the severity of acute lung injury (ALI) compared to wild-type (WT) mice. Mechanistically, we observed that 24 h post-Sm infection, NLRP3 inflammasome activation occurred, leading to gasdermin D NH2-terminal (GSDMD-NT)-induced pyroptosis in macrophages and IL-1β secretion. The NLRP3 or NLRP3 inflammasome influenced the expression PD-L1 and PD-1, as well as the count of PD-L1 or PD-1-expressing macrophages, alveolar macrophages, interstitial macrophages, PD-L1-expressing neutrophils, and the count of macrophage receptors with collagenous structure (MARCO)-expressing macrophages, particularly MARCO+ alveolar macrophages. The frequency of MARCO+ alveolar macrophages, PD-1 expression, particularly PD-1+ interstitial macrophages were negatively or positively correlated with the Sm load, respectively. Additionally, IL-1β levels in BALF correlated with three features of acute lung injury: histologic score, protein concentration and neutrophil count in BALF. Consequently, our findings suggest that Nlrp3 deletion offers protection agaisnt acute Sm pneumonia in mice by inhibiting inflammasome activation and reducing Sm infection-induced PD-L1/PD-1 or MARCO expression, particularly in macrophages. This highlights potential therapeutic targets for Sm and other gram-negative bacteria-induced acute pneumonia.
Collapse
Affiliation(s)
- Kan-Yao Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory, Guangdong Provincial People's Hospital Zhuhai Hospital, Zhuhai, China
| | - Shu-Yan Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Juan-Juan Tang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Meng-Ke Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xu-Yang Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhi-Peng Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dominique Ferrandon
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China; Université de Strasbourg, RIDI UPR9022 du CNRS, F-67000 Strasbourg, France
| | - Ke-Fang Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China; The Second Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
41
|
Gao J, Weng Z, Zhang Z, Liu Y, Liu Z, Pu X, Yu S, Zhong Y, Bai D, Xin H, Wang X. Traditional Scraping (Gua Sha) Combined with Copper-Curcumin Nanoparticle Oleogel for Accurate and Multi-Effective Therapy of Androgenic Alopecia. Adv Healthc Mater 2024; 13:e2303095. [PMID: 38175177 DOI: 10.1002/adhm.202303095] [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: 12/13/2023] [Indexed: 01/05/2024]
Abstract
Androgenetic alopecia (AGA) is a prevalent systemic disease caused by diverse factors, for which effective treatments are currently limited. Herein, the oleogel (OG) containing copper-curcumin (CuR) nanoparticles is developed, designated as CuRG, which is also combined with traditional naturopathic scraping (Gua Sha, SCR) as a multifunctional therapy for AGA. With the assistance of lipophilic OG and SCR, CuR can efficaciously penetrate the epidermal and dermal regions where most hair follicles (HFs) reside, thereby releasing curcumin (CR) and copper ions (Cu2+) subcutaneously to facilitate hair regeneration. Concomitantly, the mechanical stimulation induced by SCR promotes the formation of new blood vessels, which is conducive to reshaping the microenvironment of HFs. This study validates that the combination of CuRG and SCR is capable of systematically interfering with different pathological processes, ranging from improvement of perifollicular microenvironment (oxidative stress and insufficient vascularization), regulation of inflammatory responses to degradation of androgen receptor, thus potentiating hair growth. Compared with minoxidil, a widely used clinical drug for AGA therapy, the designed synergistic system displays augmented hair regeneration in the AGA mouse model.
Collapse
Affiliation(s)
- Jie Gao
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Zhenzhen Weng
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330088, China
| | - Ze Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Yuanyuan Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Zikang Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Xinyue Pu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Simin Yu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330088, China
| | - Yanhua Zhong
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330088, China
| | - Danmeng Bai
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330088, China
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330088, China
| |
Collapse
|
42
|
Zhang L, Guo Q, An R, Shen S, Yin L. In vitro ischemic preconditioning mediates the Ca 2+/CaN/NFAT pathway to protect against oxygen-glucose deprivation-induced cellular damage and inflammatory responses. Brain Res 2024; 1826:148736. [PMID: 38141801 DOI: 10.1016/j.brainres.2023.148736] [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: 11/06/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
Oxygen-glucose deprivation (OGD) is a critical model for studying hypoxic-ischemic cerebrovascular disease in vitro. This paper is to investigate the protection of OGD-induced cellular damage and inflammatory responses by OGD preconditioning in vitro, to provide a theoretical basis for OGD preconditioning to improve the prevention and prognosis of ischemic stroke. OGD or OGD preconditioning model was established by culturing the PC12 cell line in vitro, followed by further adding A23187 (calcium ion carrier) or CsA (calcium ion antagonist). Cell viability was detected by MTT, apoptosis by Hoechst 33,258 staining, the levels of TNF-α and IL-1β mRNA by RT-qPCR and ELISA, and the levels of CaN, NFAT, COX-2 by RT-qPCR and Western blot. Cell viability was decreased, and apoptosis, inflammatory cytokines, and CaN, NFAT, and COX-2 levels were notably increased upon OGD, while OGD pretreatment significantly increased cell viability and decreased apoptosis, inflammation, and the Ca2+/CaN/NFAT pathway. Treatment with A23187 decreased cell viability, promoted apoptosis, and significantly increased TNF-α, IL-1β, CaN, NFAT, and COX-2 levels, while CsA treatment reduced the opposite results. In vitro OGD preconditioning mediates the Ca2+/CaN/NFAT pathway to protect against OGD-induced cellular damage and inflammatory responses.
Collapse
Affiliation(s)
- Lihong Zhang
- Department of Neurointervention and Neurocritical Care, Dalian Central Hospital, Affiliated to Dalian University of Technology, Dalian, China; Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian 116023, Liaoning, China
| | - Qingzi Guo
- Deprtment of Cardiothoracic Surgery, Royal Stoke University Hospital, Stoke-on-Trent ST4 6QG, UK
| | - Ran An
- Department of Neurology, Taihe Hospital, Shiyan 442099, Hubei, China
| | - Shuhan Shen
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121012, Liaoning, China
| | - Lin Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, No. 467 Zhongshan Road, Shahekou District, Dalian 116023, Liaoning, China.
| |
Collapse
|
43
|
Wang Y, Jia X, An S, Yin W, Huang J, Jiang X. Nanozyme-Based Regulation of Cellular Metabolism and Their Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301810. [PMID: 37017586 DOI: 10.1002/adma.202301810] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/29/2023] [Indexed: 06/19/2023]
Abstract
Metabolism is the sum of the enzyme-dependent chemical reactions, which produces energy in catabolic process and synthesizes biomass in anabolic process, exhibiting high similarity in mammalian cell, microbial cell, and plant cell. Consequently, the loss or gain of metabolic enzyme activity greatly affects cellular metabolism. Nanozymes, as emerging enzyme mimics with diverse functions and adjustable catalytic activities, have shown attractive potential for metabolic regulation. Although the basic metabolic tasks are highly similar for the cells from different species, the concrete metabolic pathway varies with the intracellular structure of different species. Here, the basic metabolism in living organisms is described and the similarities and differences in the metabolic pathways among mammalian, microbial, and plant cells and the regulation mechanism are discussed. The recent progress on regulation of cellular metabolism mainly including nutrient uptake and utilization, energy production, and the accompanied redox reactions by different kinds of oxidoreductases and their applications in the field of disease therapy, antimicrobial therapy, and sustainable agriculture is systematically reviewed. Furthermore, the prospects and challenges of nanozymes in regulating cell metabolism are also discussed, which broaden their application scenarios.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Xiaodan Jia
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Shangjie An
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Wenbo Yin
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Jiahao Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
| | - Xiue Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China (USTC), Hefei, Anhui, 230026, China
- Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
44
|
Bao X, Huo S, Wang Z, Yang S, Dou L, Liu Y, Huang J, Cai C, Fang B, Xu G. Multifunctional biomimetic hydrogel dressing provides anti-infection treatment and improves immunotherapy by reprogramming the infection-related wound microenvironment. J Nanobiotechnology 2024; 22:80. [PMID: 38418972 PMCID: PMC10902999 DOI: 10.1186/s12951-024-02337-3] [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: 01/04/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024] Open
Abstract
The advancement of biomaterials with antimicrobial and wound healing properties continues to present challenges. Macrophages are recognized for their significant role in the repair of infection-related wounds. However, the interaction between biomaterials and macrophages remains complex and requires further investigation. In this research, we propose a new sequential immunomodulation method to enhance and expedite wound healing by leveraging the immune properties of bacteria-related wounds, utilizing a novel mixed hydrogel dressing. The hydrogel matrix is derived from porcine acellular dermal matrix (PADM) and is loaded with a new type of bioactive glass nanoparticles (MBG) doped with magnesium (Mg-MBG) and loaded with Curcumin (Cur). This hybrid hydrogel demonstrates controlled release of Cur, effectively eradicating bacterial infection in the early stage of wound infection, and the subsequent release of Mg ions (Mg2+) synergistically inhibits the activation of inflammation-related pathways (such as MAPK pathway, NF-κB pathway, TNF-α pathway, etc.), suppressing the inflammatory response caused by infection. Therefore, this innovative hydrogel can safely and effectively expedite wound healing during infection. Our design strategy explores novel immunomodulatory biomaterials, offering a fresh approach to tackle current clinical challenges associated with wound infection treatment.
Collapse
Affiliation(s)
- Xiaogang Bao
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Shicheng Huo
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| | - Zhenhua Wang
- Department of Laboratory Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Shengyan Yang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Linyun Dou
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Yifei Liu
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Jian Huang
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Chang Cai
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China
| | - Bin Fang
- Department of Orthopedics, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, China.
| | - Guohua Xu
- Department of Orthopedic Surgery, The Spine Surgical Center, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, China.
| |
Collapse
|
45
|
Gu J, Zhang P, Li H, Wang Y, Huang Y, Fan L, Ma X, Qian X, Xi J. Cerium-Luteolin Nanocomplexes in Managing Inflammation-Related Diseases by Antioxidant and Immunoregulation. ACS NANO 2024; 18:6229-6242. [PMID: 38345570 DOI: 10.1021/acsnano.3c09528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Oxidative stress, characterized by an imbalance between reactive oxygen species (ROS) production and the antioxidant defense system, plays a pivotal role in inflammation-related diseases. Excessive ROS levels can induce cellular damage and impair normal physiological functions, triggering the release of inflammatory mediators and exacerbating the inflammatory response, ultimately leading to irreversible tissue damage. In this study, we synthesized cerium ion-luteolin nanocomplexes (CeLutNCs) by coordinating Ce ions with the natural product luteolin, aiming to develop a therapeutic agent with excellent antioxidant and immunoregulation properties for ROS-related inflammation treatment. In vitro experiments demonstrated that the prepared CeLutNCs effectively scavenged excess ROS, prevented cell apoptosis, down-regulated levels of important inflammatory cytokines, regulated the response of inflammatory macrophages, and suppressed the activation of the nuclear factor-κ-gene binding (NF-κB) pathway. In an acute kidney injury (AKI) animal model, CeLutNCs exhibited significant efficacy in improving kidney function, repairing damaged renal tissue, and reducing oxidative stress, inflammatory response, and cellular apoptosis. Moreover, the therapeutic potential of CeLutNCs in an acute lung injury (ALI) model was confirmed through the assessment of inflammatory responses and histopathological studies. This study emphasizes the effectiveness of these metal-natural product coordination nanocomplexes as a promising therapeutic approach for preventing AKI and other diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Jiake Gu
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225009, China
| | - Peiying Zhang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Huajun Li
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Yisen Wang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Ying Huang
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Lei Fan
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, China
| | - Xiao Ma
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaodong Qian
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Juqun Xi
- Medical College, Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
46
|
Gao F, Liang W, Chen Q, Chen B, Liu Y, Liu Z, Xu X, Zhu R, Cheng L. A Curcumin-Decorated Nanozyme with ROS Scavenging and Anti-Inflammatory Properties for Neuroprotection. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:389. [PMID: 38470720 PMCID: PMC10934375 DOI: 10.3390/nano14050389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/16/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024]
Abstract
Disordered reactive oxygen/nitrogen species are a common occurrence in various diseases, which usually cause cellular oxidative damage and inflammation. Despite the wide range of applications for biomimetic nanoparticles with antioxidant or anti-inflammatory properties, designs that seamlessly integrate these two abilities with a synergistic effect in a simple manner are seldom reported. In this study, we developed a novel PEI-Mn composite nanoparticle (PM NP) using a chelation method, and the curcumin was loaded onto PM NPs via metal-phenol coordination to form PEI-Mn@curcumin nanoparticles (PMC NPs). PMC NPs possessed excellent dispersibility and cytocompatibility, was engineered to serve as an effective nanozyme, and exhibited specific SOD-like and CAT-like activities. In addition, the incorporation of curcumin granted PMC NPs the ability to effectively suppress the expression of inflammatory cytokines in microglia induced by LPS. As curcumin also has antioxidant properties, it further amplified the synergistic efficiency of ROS scavenging. Significantly, PMC NPs effectively scavenged ROS triggered by H2O2 in SIM-A9 microglia cells and Neuro-2a cells. PMC NPs also considerably mitigated DNA and lipid oxidation in Neuro-2a cells and demonstrated an increase in cell viability under various H2O2 concentrations. These properties suggest that PMC NPs have significant potential in addressing excessive ROS and inflammation related to neural diseases.
Collapse
Affiliation(s)
- Feng Gao
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| | - Wenyu Liang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| | - Qixin Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| | - Bairu Chen
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Yuchen Liu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Zhibo Liu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
| | - Xu Xu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| | - Rongrong Zhu
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| | - Liming Cheng
- Department of Orthopedics, Tongji Hospital Affiliated to Tongji University, School of Medicine, Tongji University, Shanghai 200331, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, School of Life Science and Technology, Tongji University, Shanghai 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200065, China
| |
Collapse
|
47
|
Fan C, Zhang Z, Lai Z, Yang Y, Li J, Liu L, Chen S, Hu X, Zhao H, Cui S. Chemical Evolution and Biological Evaluation of Natural Products for Efficient Therapy of Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305432. [PMID: 38126681 PMCID: PMC10870070 DOI: 10.1002/advs.202305432] [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: 08/08/2023] [Revised: 11/01/2023] [Indexed: 12/23/2023]
Abstract
Acute lung injury (ALI) is one of the most common complications in COVID-19 and also a syndrome of acute respiratory failure with high mortality rates, but lacks effective therapeutic drugs. Natural products provide inspiration and have proven to be the most valuable source for bioactive molecule discovery. In this study, the chemical evolution of the natural product Tanshinone IIA (Tan-IIA) to achieve a piperidine-fused scaffold through a synthetic route of pre-activation, multi-component reaction, and post-modification is presented. Through biological evaluation, it is pinpointed that compound 8b is a standout candidate with remarkable anti-inflammation and anti-oxidative stress properties, coupled with low toxicity. The mechanistic study unveils a multifaceted biological profile of 8b and shows that 8b is highly efficient in vivo for the treatment of ALI. Therefore, this work not only provides an effective strategy for the treatment of ALI, but also offers a distinctive natural product-inspired drug discovery.
Collapse
Affiliation(s)
- Chengcheng Fan
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Zeyi Zhang
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Zhencheng Lai
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Yanzi Yang
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Jiaming Li
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Lei Liu
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Siyu Chen
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
| | - Xueping Hu
- Institute of Molecular Sciences and EngineeringInstitute of Frontier and Interdisciplinary ScienceShandong UniversityQingdao266237China
| | - Huajun Zhao
- College of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhou311402China
| | - Sunliang Cui
- Institute of Drug Discovery and DesignCollege of Pharmaceutical SciencesNational Key Laboratory of Advanced Drug Delivery and Release SystemsZhejiang University866 Yuhangtang RoadHangzhou310058China
- Jinhua Institute of Zhejiang UniversityJinhuaZhejiang321299China
| |
Collapse
|
48
|
Wang R, Li Q, Wu P, Ren K, Li Y, Wang Y, Zhu H, Lv C. Fe-Capsaicin Nanozymes Attenuate Sepsis-Induced Acute Lung Injury via NF-κB Signaling. Int J Nanomedicine 2024; 19:73-90. [PMID: 38187907 PMCID: PMC10771734 DOI: 10.2147/ijn.s436271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/17/2023] [Indexed: 01/09/2024] Open
Abstract
Background In sepsis, the lungs are one of the most severely affected organs, usually resulting in acute lung injury (ALI). Capsaicin (CAP) is a natural compound found in chili peppers that has pain-relieving and anti-inflammatory properties. Here, we report that nanoparticles containing capsaicin and iron (Fe-CAP NPs) exhibited anti-inflammatory effects in the treatment of ALI. Methods The morphological characteristics of nanozymes were detected. RAW 264.7 cells were divided into four groups: control, lipopolysaccharide (LPS), CAP+LPS and Fe-CAP+LPS groups. The expression of inducible nitric oxide synthase (iNOS), transforming growth factor-β (TGF-β), and tumor necrosis factor-α (TNF-α) was assessed by immunofluorescence, Western blot, and enzyme-linked immunosorbent assay (ELISA). Nuclear factor kappa-B (NF-κB) expression was determined by Western blot. C57 mice were divided into control, LPS, CAP+LPS and Fe-CAP+LPS groups. Interleukin-6 (IL-6) and iNOS expression in the lung was detected by Western Blot. IL-6 and TNF-α expression in serum was detected by ELISA. Extravasated Evans blue, histopathological evaluation and wet-to-dry (W/D) weight ratio were used to assess pulmonary capillary permeability. The blood and major organs (heart, liver, spleen, lung and kidney) of mice were tested for the toxicity of Fe-CAP NPs. Results In the LPS group, TNF-α, iNOS, p-NF-κB and p-IKBα expression increased. However, their expression was significantly decreased in the Fe-CAP+LPS group. TGF-β expression showed the opposite trend. In vivo, IL-6 and iNOS expression was notably increased in the lungs of LPS group of mice but decreased with Fe-CAP pretreatment. Fe-CAP significantly ameliorated lung EB leakage, improved the histopathology of lung tissue and reduced the W/D weight ratio. The nanoparticles showed non-cytotoxicity, when studying these biological activities. Conclusion Fe-CAP NPs could alleviated inflammation by inhibiting the expression of pro-inflammatory factors in macrophages, increasing the expression of anti-inflammatory factors, and alleviating lung tissue damage.
Collapse
Affiliation(s)
- Ruijie Wang
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, People’s Republic of China
| | - Quan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Pengxin Wu
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Ke Ren
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yang Wang
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Huadong Zhu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Chuanzhu Lv
- Research Unit of Island Emergency Medicine, Chinese Academy of Medical Sciences (No. 2019RU013), Hainan Medical University, Haikou, People’s Republic of China
- Emergency Medicine Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, People’s Republic of China
| |
Collapse
|
49
|
Yang P, Huang Q, Zhang J, Li Y, Gao H, Gu Z. Natural Polyphenolic Nanodots for Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308393. [PMID: 38010256 DOI: 10.1002/adma.202308393] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/07/2023] [Indexed: 11/29/2023]
Abstract
The abnormal amyloid-β accumulation is essential and obbligato in Alzheimer's disease pathogenesis and natural polyphenols exhibit great potential as amyloid aggregation inhibitors. However, the poor metabolic stability, low bioavailability, and weak blood-brain barrier crossing ability of natural polyphenol molecules fail to meet clinical needs. Here, a universal protocol to prepare natural polyphenolic nanodots is developed by heating in aqueous solution without unacceptable additives. The nanodots are able to not only inhibit amyloid-β fibrillization and trigger the fibril disaggregation, but mitigate the amyloid-β-plaque-induced cascade impairments including normalizing oxidative microenvironment, altering microglial polarization, and rescuing neuronal death and synaptic loss, which results in significant improvements in recognition and cognition deficits in transgenic mice. More importantly, natural polyphenolic nanodots possess stronger antiamyloidogenic performance compared with small molecule, as well as penetrate the blood-brain barrier. The excellent biocompatibility further guarantees the potential of natural polyphenolic nanodots for clinical applications. It is expected that natural polyphenolic nanodots provide an attractive paradigm to support the development of the therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Peng Yang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- Department of Health Products Technical Research and Development Center, Yunnanbaiyao Group Co. Ltd., Kunming, 650500, China
| | - Qianqian Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jianhua Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Yiwen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Zhipeng Gu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
50
|
Chen X, Tang Z. Novel application of nanomedicine for the treatment of acute lung injury: a literature review. Ther Adv Respir Dis 2024; 18:17534666241244974. [PMID: 38616385 PMCID: PMC11017818 DOI: 10.1177/17534666241244974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
Nanoparticles have attracted extensive attention due to their high degree of cell targeting, biocompatibility, controllable biological activity, and outstanding pharmacokinetics. Changing the size, morphology, and surface chemical groups of nanoparticles can increase the biological distribution of agents to achieve precise tissue targeting and optimize therapeutic effects. Examples of their use include nanoparticles designed for increasing antigen-specific immune responses, developing vaccines, and treating inflammatory diseases. Nanoparticles show the potential to become a new generation of therapeutic agents for regulating inflammation. Recently, many nanomaterials with targeted properties have been developed to treat acute lung injury/acute respiratory distress syndrome (ALI/ARDS). In this review, we provide a brief explanation of the pathological mechanism underlying ALI/ARDS and a systematic overview of the latest technology and research progress in nanomedicine treatments of ALI, including improved nanocarriers, nanozymes, and nanovaccines for the targeted treatment of lung injury. Ultimately, these nanomedicines will be used for the clinical treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Xianfeng Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Zhanhong Tang
- Department of Intensive Care Unit, the First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning 530021, China
| |
Collapse
|